WO2002057453A2 - Polypetides and nucleic acids encoding same - Google Patents

Polypetides and nucleic acids encoding same Download PDF

Info

Publication number
WO2002057453A2
WO2002057453A2 PCT/US2001/050331 US0150331W WO02057453A2 WO 2002057453 A2 WO2002057453 A2 WO 2002057453A2 US 0150331 W US0150331 W US 0150331W WO 02057453 A2 WO02057453 A2 WO 02057453A2
Authority
WO
WIPO (PCT)
Prior art keywords
nucleic acid
amino acid
polypeptide
protein
seq
Prior art date
Application number
PCT/US2001/050331
Other languages
French (fr)
Other versions
WO2002057453A3 (en
Inventor
Esha A. Gangolli
Meera Patturajan
Corine A. M. Vernet
Uriel M. Malyankar
Ramesh Kekuda
David J. Stone
David Anderson
Richard A. Shimkets
Catherine E. Burgess
Bryan D. Zerhusen
Xiaohong Liu
Kimberly A. Spytek
Stacie J. Casman
Ferenc L. Boldog
Glennda Smithson
Li Li
Weizhen Ji
Original Assignee
Curagen Corporation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curagen Corporation filed Critical Curagen Corporation
Priority to JP2002558505A priority Critical patent/JP2005508601A/en
Priority to EP01993342A priority patent/EP1352065A2/en
Priority to CA002433313A priority patent/CA2433313A1/en
Publication of WO2002057453A2 publication Critical patent/WO2002057453A2/en
Publication of WO2002057453A3 publication Critical patent/WO2002057453A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/12Ophthalmic agents for cataracts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Dermatology (AREA)
  • Hematology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Diabetes (AREA)
  • Obesity (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Pulmonology (AREA)
  • Biophysics (AREA)
  • Peptides Or Proteins (AREA)
  • Psychology (AREA)
  • Hospice & Palliative Care (AREA)
  • Vascular Medicine (AREA)

Abstract

Disclosed herein are nucleic acid sequences that encode novel polypeptides. Also disclosed are polypeptides encoded by these nucleic acid sequences, and antibodies, which immunospecifically-bind to the polypeptide, as well as derivatives, variants, mutants, or fragments of the aforementioned polypeptide, polynucleotide, or antibody. The invention further discloses therapeutic, diagnostic and research methods for diagnosis, treatment, and prevention of disorders involving any one of these novel human nucleic acids and proteins.

Description

POLYPEPTIDES AND NUCLEIC ACIDS ENCODING SAME
FIELD OF THE INVENTION
The invention generally relates to nucleic acids and polypeptides encoded thereby.
BACKGROUND OF THE INVENTION
The epidermal growth factor (EGF) superfamily comprises a diverse group of proteins that function as secreted signaling molecules, growth factors, and components of the extracellular matrix, many with a role in vertebrate development. EGF -related proteins with Cls-li e (CUB) domains have been reported. The CUB domain is found in 16 functionally diverse proteins such as the dorso-ventral patterning protein tolloid, bone niorphogenetic protein- 1, a family of spermadhesins, complement subcomponents Cls/Clr and the neuronal recognition molecule A5. Most of these proteins are known to be involved in developmental processes. The second domain is found mostly among developmentally-regulated proteins and spermadhesins.
The adipocyte complement related protein-3 (AC P3), is a 30 kDa serum protein made and secreted exclusively from adipocyte cells, which is implicated in energy homeostasis and obesity. ACRP3 is structurally similar to complement factor Clq and to a hibernation-specific protein isolated from the plasma of Siberian chipmunks; it forms large homo-oligomers that undergo a series of post-translational modifications (see, Scherer PE, et al, J Biol Chem 1995 Nov 10;270(45):26746-9). ACRP30 is a close homologue of the complement protein Clq,
which is involved in the recognition of microbial surfaces and antibody- antigen complexes in the classical pathway of complement. The crystal structure of a homotrimeric fragment of ACRP3 has been solved to 2.1 A resolution. The structure reveals homology to the tumor necrosis factor (TNF) family. Identical folding topologies, key residue conservations, and similarity of trimer interfaces and intron positions establish an evolutionary link between the TNF and Clq families.
Clq is the first subcomponent of the Cl complex of the classical pathway of complement activation. Several functions have been assigned to Clq, which include antibody- dependent and independent immune functions, and are considered to be mediated by Clq receptors present on the effector cell surface. There remains some uncertainty about the identities of the receptors that mediate Clq functions. Some of the previously described Clq receptor molecules, such as gClqR and cClqR, now appear to have less of a role in Clq functions than in functions unrelated to Clq. The problem of identifying receptor proteins with complementary binding sites for Clq has been compounded by the highly charged nature of the different domains in Clq. Although newer candidate receptors like ClqR(p) and CRl have emerged, full analysis of the Clq-Clq receptor interactions is still at an early stage. In view of the diverse functions that Clq is considered to perform, it has been speculated that several Clq-binding proteins may act in concert, as a Clq receptor complex, to bring about Clq mediated functions. Some major advances have been made in last few years. Experiments with gene targeted homozygous Clq-deficient mice have suggested a role for Clq in modulation of the humoral immune response, and also in protection against development of autoimmunity. The recently described crystal structure of ACRP-30, has revealed a new Clq/TNF superfamily of proteins. Although the members of this superfamily may have diverse functions, there may be a common theme in their phylogeny and modular organisation of their distinctive globular domains.
The first component of complement is a calcium-dependent complex of the 3 subcomponents Clq, Clr, and Cls. Subcomponent Clq binds to immunoglobulin complexes with resulting serial activation of Clr (enzyme), Cls (proenzyme) and the other 8 components of complement. Clq is composed of 3 different species of chains, called A, B, and C. Fragments of the A chain of Clq have been sequenced. The total A chain contains 190 amino acids. Clq shares with collagen the presence of hydroxyproline in its amino acid sequence.
Beta-adrenergic receptor kinase (beta-ARKl) phosphorylates the beta-2-adrenergic receptor and appears to mediate agonist-specific desensitization observed at high agonist concentrations. Beta-ARKl is an ubiquitous cytosolic enzyme that specifically phosphorylates the activated form of the beta-adrenergic and related G-protein-coupled receptors. The beta-ARKl gene spans approximately 23 kb and is composed of 21 exons. Beta-AR kinase (beta-ARKl) is known to be elevated in failing human heart tissue and its activity resulting in rapid desensitization via the abnormal coupling or uncoupling of beta- adrenergic receptor to G protein, receptor down-regulation, internalization and degradation, may account for some of the abnormalities of contractile function in the heart disease (see, Post, S. R., Hammond, H.K., Insel, P. A., 1999, Annu. Rev. Pharmacol. Vol. 39: 343-360) incoφorated by reference.
The TEN-M4 protein belongs to the ODZ/TENM family of proteins. This family was first identified in Drosophila as being a pair-rule gene affecting segmentation of the early embryo. It was the first pair-rule gene identified that was not a transcription factor, but a type II transmembrane protein. Vertebrate homologs of the TENM family have been identified in mouse and zebrafish. In the mouse, TEN-M4 expression was found to be on the cell surface, in the brain, trachea as well as developing limb and bone. Analysis of the TEN-MI protem reveals that it can bind to itself, making it likely that TEN-M4 may be a dimeric moiety as well. In cell culture experiments, fragments of the TEN-M proteins can bind the Drosophila PS2 integrins. In addition, members of the TEN-M family have been identified to be downstream of the endoplasmic reticulum stress response pathway, which alters the response of cells to their environment. This suggests that the ODZ/TENM family may be involved in cell adhesion, spreading and motility. Translocations leading to the fusion of this gene with the NRG1/HGL gene from chromosome 8 have been found to generate a paracrine growth factor for one mammary carcinoma cell line, termed gamma-heregulin.
Out At First is expressed in clusters of cells during germband extension, throughout the developing nervous system, and in the gonads of both sexes throughout the lifecycle. Mutation of the Drosophila gene is fatal and causes nervous system defects.
Butyrophilin plays several crucial roles in T-cell activation. The protein is known to be expressed in spleen and liver.
Sugar transport is a critical feature of many cell types in the body as energy storage and metabolism or defects thereof can cause a variety of human diseases. Glucose tranporter 4 (GLUT4) is critical to insulin-sensitive glucose uptake.
Mouse EphA6 (also known as m-ehk2) belongs to the superfamily of receptor tyrosine kinases, which constitute the largest family of oncogenes. This family includes prominent growth factor receptors such as those for epidermal growth factor, platelet-derived growth factor etc. Members of this superfamily influence cell shape, mobility, differentiation and proliferation. Within this superfamily, the Ephrin (Eph) receptors constitute the largest subfamily. Eph receptors and their ligands, ephrins, are known to be involved in several normal developmental processes, including formation of segmented structures, axon guidance, cell adhesion and development of vasculature. Ephrin receptors are classified into two main subtypes: EphA receptors bind to GPI-anchored ephrin-A ligands, while EphB receptors bind to ephrin-B proteins that have a transmembrane and cytoplasmic domain. The EphA6 receptor is highly expressed in the mouse brain and inner ear, including the cochlea. This receptor is also differentially expressed relative to the other ephrin receptors in certain regions of the primate neocortex during development. In addition, it is found in the developing retina and optic tectum in the chicken.
SUMMARY OF THE INVENTION
The present invention is based in part on nucleic acids encoding proteins that are members of the following protein families: EGF related SCUBEl-like proteins, Adipocyte Complement Related proteins, complement Clq tumor necrosis factor-like proteins, β- Adrenergic Receptor Kinase-like proteins, TENM4-like proteins, Out At First-like proteins, EphA6-ehk2-like proteins, Glucose Transporter-like proteins, Type la Membrane Sushi- Containing Domain-like proteins, Type la Membrane Sushi-Containing Domain proteins, Butyrophilin-like proteins, and Butyrophilin Precursor B7-DC-like proteins. The novel polynucleotides and polypeptides are referred to herein as NOV1, NOV2a, NOV2b, NOV2c, NOV2d, NOV3, NOV4, NOV5a, NOV5b, NOV6a, NOV6b, NOV7, NOV8, NOV9, NOVlOa, NOVlOb and NOV11. These nucleic acids and polypeptides, as well as derivatives, homologs, analogs and fragments thereof, will hereinafter be collectively designated as "NOVX" nucleic acid or polypeptide sequences.
In one aspect, the invention provides an isolated NOVX nucleic acid molecule encoding a NOVX polypeptide that includes a nucleic acid sequence that has identity to the nucleic acids disclosed in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 23, 25, 27, 29, 31 and 33. In some embodiments, the NOVX nucleic acid molecule will hybridize under stringent conditions to a nucleic acid sequence complementary to a nucleic acid molecule that includes a protein-coding sequence of a NOVX nucleic acid sequence. The invention also includes an isolated nucleic acid that encodes a NOVX polypeptide, or a fragment, homolog, analog or derivative thereof. For example, the nucleic acid can encode a polypeptide at least 80% identical to a polypeptide comprising the amino acid sequences of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34. The nucleic acid can be, for example, a genomic DNA fragment or a cDNA molecule that includes the nucleic acid sequence of any of SEQ D NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31 and 33.
Also included in the invention is an oligonucleotide, e.g., an oligonucleotide which includes at least 6 contiguous nucleotides of a NOVX nucleic acid (e.g., SEQ ID NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31 and 33) or a complement of said oligonucleotide. Also included in the invention are substantially purified NOVX polypeptides (SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32 and 34). In certain embodiments, the NOVX polypeptides include an amino acid sequence that is substantially identical to the amino acid sequence of a human NOVX polypeptide.
The invention also features antibodies that immunoselectively bind to NOVX polypeptides, or fragments, homologs, analogs or derivatives thereof.
In another aspect, the invention includes pharmaceutical compositions that include therapeutically- or prophylactically-effective amounts of a therapeutic and a pharmaceutically- acceptable carrier. The therapeutic can be, e.g. , a NOVX nucleic acid, a NOVX polypeptide, or an antibody specific for a NOVX polypeptide. In a further aspect, the invention includes, in one or more containers, a therapeutically- or prophylactically-effective amount of this pharmaceutical composition.
In a further aspect, the invention includes a method of producing a polypeptide by culturing a cell that includes a NOVX nucleic acid, under conditions allowing for expression of the NOVX polypeptide encoded by the DNA. If desired, the NOVX polypeptide can then be recovered.
In another aspect, the invention includes a method of detecting the presence of a NOVX polypeptide in a sample. In the method, a sample is contacted with a compound that selectively binds to the polypeptide under conditions allowing for formation of a complex between the polypeptide and the compound. The complex is detected, if present, thereby identifying the NOVX polypeptide within the sample.
The invention also includes methods to identify specific cell or tissue types based on their expression of a NOVX.
Also included in the invention is a method of detecting the presence of a NOVX nucleic acid molecule in a sample by contacting the sample with a NOVX nucleic acid probe or primer, and detecting whether the nucleic acid probe or primer bound to a NOVX nucleic acid molecule in the sample.
In a further aspect, the invention provides a method for modulating the activity of a NOVX polypeptide by contacting a cell sample that includes the NOVX polypeptide with a compound that binds to the NOVX polypeptide in an amount sufficient to modulate the activity of said polypeptide. The compound can be, e.g., a small molecule, such as a nucleic acid, peptide, polypeptide, peptidomimetic, carbohydrate, lipid or other organic (carbon containing) or inorganic molecule, as further described herein.
Also within the scope of the invention is the use of a therapeutic in the manufacture of a medicament for treating or preventing disorders or syndromes including, e.g., Von Hippel- Lindau (VHL) syndrome, cirrhosis, transplantation disorders, pancreatitis, obesity, diabetes, autoimmune disease, renal artery stenosis, interstitial nepliritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypercalcemia, Lesch-Nyhan syndrome, developmental defects, cataract, spinal cord injury, Alzheimer's disease, muscular dystrophy, acoustic trauma, cancer, learning and memory defects, infertility, cardiomyopathies, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect, atrioventricular canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect, valve diseases, tuberous sclerosis, scleroderma, endometriosis, hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, dementia, stroke, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, familial hypercholesterolemia, hyperlipoproteinemia II phenotype, tendinous xanthomas, corneal arcus, coronary artery disease, planar xanthomas, webbed digits, hypercholesterolemia, fertility, xanthomatosis, hepatitis C infection, regulation, synthesis, transport, recycling, or turnover of LDL receptors, cerebral arteriopathy with subcortical infarcts and leukoencephalopathy, epiphyseal dysplasia, multiple 1, ichthyosis, nonlamellar and nonerythrodermic, congenital, leukemia, T-cell acute lymphoblastoid, pseudoachondroplasia, SCID, autosomal recessive, T-negative/B-positive type, C3 deficiency, diabetes mellitus, insulin-resistant, with acanthosis nigricans, glutaricaciduria, type I, hypothyroidism, congenital, leprechaunism, liposarcoma, mucolipidosis IV, persistent mullerian duct syndrome, type I, Rabson-Mendenhall syndrome, thyroid carcinoma, with cell oxyphilia, erythrocytosis, malaria, to, bleeding disorder due to defective thromboxane A2 receptor, cerebellar ataxia, convulsions, familial febrile, cyclic hematopoiesis, fucosyltransferase-6 deficiency, GAMT deficiency, psoriasis, actinic keratosis, tuberous sclerosis, acne, hair growth, allopecia, pigmentation disorders, endocrine disorders, trauma, immunological disease, respiratory disease, gastro-intestinal diseases, reproductive health, neurological diseases, bone marrow transplantation, metabolic and endocrine diseases, allergy and inflammation, nephrological disorders, hematopoietic disorders, urinary system disorders, atopy; osteoporosis-pseudoglioma syndrome; Smith-Lemli-Opitz syndrome, type I; Smith- Lemli-Opitz syndrome, type II; xeroderma pigmentosum, Asthma, diabetes mellitus, susceptibility to DDDM; angioedema, paraganglioma, familial nonchromaffin, neuroprotection; Lambert-Eaton myasthenic syndrome, digestive system disorders, all or some of the protease/protease inhibitor deficiency disorders, acyl-CoA dehydrogenase, brachydactyly, carbamoylphosphate synthetase I deficiency, cardiomyopathy cataract Coppock-like, cataract crystalline aculeiform, cataract polymorphic congenital, cataract variable zonular pulverulent, cataracts punctate progressive juvenile-onse, choreoathetosis familial paroxysmal, craniofacial-deafhess-hand syndrome, ichthyosis lamellar, type 2, myopathy, desmin-related cardioskeletal, resistance/susceptibility to TB, rhabdomyosarcoma alveolar, Waardenburg syndrome type I and type III, Alport syndrome autosomal recessive, Bjoπistad syndrome, hematuria, hyperoxaluria primary, type 1, syndactyly type 1, hyperproglucagonemia, Bethlem myopathy, brachydactyly type E, brachydactyly-mental retardation syndrome, Finnish lethal neonatal metabolic syndrome, Simpson-Golabi-Behmel syndrome, Beckwith-Wiedemann syndrome, pathogen infections, heart disease, prostate cancer, angiogenesis and wound healing, modulation of apoptosis, neuropsychiatric disorders, age-related disorders, pathological disorders involving spleen, thymus, lung, and peritoneal macrophages and/or other pathologies and disorders of the like.
The therapeutic can be, e.g., a NOVX nucleic acid, a NOVX polypeptide, or a NOVX- specific antibody, or biologically-active derivatives or fragments thereof.
For example, the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed above and/or other pathologies and disorders of the like. The polypeptides can be used as immunogens to produce antibodies specific for the invention, and as vaccines. They can also be used to screen for potential agonist and antagonist compounds. For example, a cDNA encoding NOVX may be useful in gene therapy, and NOVX may be useful when administered to a subject in need thereof. By way of non-limiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
The invention further includes a method for screening for a modulator of disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like. The method includes contacting a test compound with a NOVX polypeptide and determining if the test compound binds to said NOVX polypeptide. Binding of the test compound to the NOVX polypeptide indicates the test compound is a modulator of activity, or of latency or predisposition to the aforementioned disorders or syndromes.
Also within the scope of the invention is a method for screening for a modulator of activity, or of latency or predisposition to disorders or syndromes including, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like by administering a test compound to a test animal at increased risk for the aforementioned disorders or syndromes. The test animal expresses a recombinant polypeptide encoded by a NOVX nucleic acid. Expression or activity of NOVX polypeptide is then measured in the test animal, as is expression or activity of the protein in a control animal which recombinantly- expresses NOVX polypeptide and is not at increased risk for the disorder or syndrome. Next, the expression of NOVX polypeptide in both the test animal and the control animal is compared. A change in the activity of NOVX polypeptide in the test animal relative to the control animal indicates the test compound is a modulator of latency of the disorder or syndrome.
In yet another aspect, the invention includes a method for determining the presence of or predisposition to a disease associated with altered levels of a NOVX polypeptide, a NOVX nucleic acid, or both, in a subject (e.g., a human subject). The method includes measuring the amount of the NOVX polypeptide in a test sample from the subject and comparing the amount of the polypeptide in the test sample to the amount of the NOVX polypeptide present in a control sample. An alteration in the level of the NOVX polypeptide in the test sample as compared to the control sample indicates the presence of or predisposition to a disease in the subject. Preferably, the predisposition includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like. Also, the expression levels of the new polypeptides of the invention can be used in a method to screen for various cancers as well as to determine the stage of cancers.
In a further aspect, the invention includes a method of treating or preventing a pathological condition associated with a disorder in a mammal by administering to the subject a NOVX polypeptide, a NOVX nucleic acid, or a NOVX-specific antibody to a subject (e.g., a human subject), in an amount sufficient to alleviate or prevent the pathological condition. In preferred embodiments, the disorder, includes, e.g., the diseases and disorders disclosed above and/or other pathologies and disorders of the like.
In yet another aspect, the invention can be used in a method to identity the cellular receptors and downstream effectors of the invention by any one of a number of techniques commonly employed in the art. These include but are not limited to the two-hybrid system, affinity purification, co-precipitation with antibodies or other specific-interacting molecules.
NOVX nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel NOVX substances for use in therapeutic or diagnostic methods. These NOVX antibodies may be generated according to methods lαiown in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. The disclosed NOVX proteins have multiple hydrophilic regions, each of which can be used as an immunogen. These NOVX proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
The NOVX nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In the case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Other features and advantages of the invention will be apparent from the following detailed description and claims.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides novel nucleotides and polypeptides encoded thereby.
Included in the invention are the novel nucleic acid sequences and their encoded polypeptides referred to herein as NOV1, NOV2a, NOV2b, NOV2c, NOV2d, NOV3, NOV4, NOV5a,
NOV5b, NOV6a, NOV6b, NOV7, NOV8, NOV9, NOVlOa, NOVlOb and NOV11. The sequences are collectively referred to herein as "NOVX nucleic acids" or "NOVX polynucleotides" and the corresponding encoded polypeptides are referred to as "NOVX polypeptides" or "NOVX proteins." Unless indicated otherwise, "NOVX" is meant to refer to any of the novel sequences disclosed herein. Table A provides a summary of the NOVX nucleic acids and their encoded polypeptides.
TABLE A. Sequences and Corresponding SEQ ID Numbers
Figure imgf000012_0001
NOVX nucleic acids and their encoded polypeptides are useful in a variety of applications and contexts. The various NOVX nucleic acids and polypeptides according to the invention are useful as novel members of the protein families according to the presence of domains and sequence relatedness to previously described proteins. Additionally, NOVX nucleic acids and polypeptides can also be used to identify proteins that are members of the family to which the NOVX polypeptides belong.
NOV1 is homologous to an EGF-Related SCUBEl-like family of proteins. Thus, the NOV1 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; cancer, obesity, endometriosis, trauma, viral, bacterial, or parasitic infections, allergy, asthma, endocrine disfunctions, diabetes, growth and reproductive disorders, and other diseases, disorders and conditions of the like.
NOV2 is homologous to the adipocyte complement Clq Tumor Necrosis Factor-like family of proteins. Thus NOV2 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral'bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV3 is homologous to a family of beta-adrenergic receptor kinase-like proteins. Thus, the NOV3 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example: cardiac disorders and disorders of myocontractility and the like.
NOV4 is homologous to the TEN-M4-like family of proteins. Thus, NOV4 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example: cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV5 is homologous to the Out At First (OAF)-like family of proteins. Thus NOV5 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in central nervous system diseases, disorders and conditions of the like.
NOV6 is homologous to the EphA6/ehk-2-like family of proteins. Thus NOV6 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example: cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV7 is homologous to members of the glucose transporter-like family of proteins. Thus, the NOV7 nucleic acids, polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; obesity, diabetes, cancer, inflammation, CNS diseases and other diseases, disorders and conditions of the like.
NOV8 is homologous to the Type la Membrane Sushi-Containing Domain-like family of proteins. Thus, NOV8 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV9 is homologous to the Type la Membrane Sushi-Containing Domain-like family of proteins. Thus, NOV9 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example: cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV10 is homologous to the butyrophilin-like family of proteins. Thus, NOV10 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like.
NOV11 is homologous to the cysteine sulfinic acid decarboxylase-like family of proteins. Thus, NOV11 nucleic acids and polypeptides, antibodies and related compounds according to the invention will be useful in therapeutic and diagnostic applications implicated in, for example; cancer, inflammation, neurological disorders, neuropsychiatric disorders, obesity, diabetes, viral/bacterial/parasitic infections, autoimmune diseases, renal artery stenosis, renal tubular acidosis, hypercalcemia, IgA nephropathy, Lesch-Nyhan syndrome, glomerulonephritis, interstitial nephritis, polycystic kidney disease, trauma, regeneration, Alzheimer's disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection, obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD) and other diseases, disorders and conditions of the like. The NOVX nucleic acids and polypeptides can also be used to screen for molecules, which inhibit or enhance NOVX activity or function. Specifically, the nucleic acids and polypeptides according to the invention may be used as targets for the identification of small molecules that modulate or inhibit, e.g., neurogenesis, cell differentiation, cell proliferation, hematopoiesis, wound healing and angiogenesis.
Additional utilities for the NOVX nucleic acids and polypeptides according to the invention are disclosed herein.
NOV1
A disclosed NOV1 nucleic acid of 3137 nucleotides (also referred to as CG55758-01) encoding a novel EGF-Related Protein (SCUBEl)-like protein is shown in Table 1 A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 78-80 and ending with a TGA codon at nucleotides 2973-2975. A putative untranslated region upstream from the initiation codon and downstream from the termination codon is underlined in Table 1 A. The start and stop codons are in bold letters.
Table 1A
NOV1 Polynucleotide
SEQ ID NO: 1
AGCGCCTGCGGGAGCGGCCGGTCGGTCGGGTCCCCGCGCCCCGCACGCCCGCACGCCCAGCGGGGCCCGC ATTGAGCATGGGCGCGGCGGCCGTGCGCTGGCACTTGTGCGTGCTGCTGGCCCTGGGCACACGCGGGCGG CTGGCCGGGGGCAGCGGGCTCCCAGGGTCAGTCGACGTGGATGAGTGCTCAGAGGGCACAGATGACTGCC ACATCGATGCCATCTGTCAGAACACGCCCAAGTCCTACAAATGCCTCTGCAAGCCAGGCTACAAGGGGGA AGGCAAGCAGTGTGAAGACATTGACGAGTGTGAGAATGACTACTACAATGGGGGCTGTGTCCACGAGTGC ATCAACATCCCGGGGAACTACAGGTGTACCTGCTTTGATGGCTTCATGCTGGCACACGATGGACACAACT GCCTGGATGTGGACGAGTGTCAGGACAATAATGGTGGCTGCCAGCAGATCTGCGTCAATGCCATGGGCAG CTACGAGTGTCAGTGCCACAGTGGCTTCCTCCTTAGTGACAACCAGCATACCTGCATCCACCGCTCCAAT GAGGGTATGAACTGCATGAACAAAGACCATGGCTGTGCCCACATCTGCCGGGAGACGCCCAAAGGTGGGG TGGCCTGCGACTGCAGGCCCGGCTTTGACCTTGCCCAAAACCAGAAGGACTGCACACTAACCTGTAATTA TGGAAACGGAGGCTGCCAGCACAGCTGTGAGGACACAGACACAGGCCCCACGTGTGGTTGCCACCAGAAG TACGCCCTCCACTCAGACGGTCGCACGTGCATCGAGACGTGCGCAGTCAATAACGGAGGCTGCGACCGGA CATGCAAGGACACAGCCACTGGCGTGCGATGCAGCTGCCCCGTTGGATTCACACTGCAGCCGGACGGGAA GACATGCAAAGACATCAACGAGTGCCTGGTCAACAACGGAGGCTGCGACCACTTCTGCCGCAACACCGTG GGCAGCTTCGAGTGCGGCTGCCGGAAGGGCTACAAGCTGCTCACCGACGAGCGCACCTGCCAGGACATCG ACGAGTGCTCCTTCGAGCGGACCTGTGACCACATCTGCATCAACTCCCCGGGCAGCTTCCAGTGCCTGTG TCACCGCGGCTACATCCTCTACGGGACAACCCACTGCGGAGATGTGGACGAGTGCAGCATGAGCAACGGG AGCTGTGACCAGGGCTGCGTCAACACCAAGGGCAGCTACGAGTGCGTCTGTCCCCCGGGGAGGCGGCTCC ACTGGAACGGGAAGGATTGCGTGGAGACAGGCAAGTGTCTTTCTCGCGCCAAGACCTCCCCCCGGGCCCA GCTGTCCTGCAGCAAGGCAGGCGGTGTGGAGAGCTGCTTCCTTTCCTGCCCGGCTCACACACTCTTCGTG CCACAAGACTCGGAAAATAGCTACGTCCTGAGCTGCGGAGTTCCAGGGCCGCAGGGCAAGGCGCTGCAGA AACGCAACGGCACCAGCTCTGGCCTCGGGCCCAGCTGCTCAGATGCCCCCACCACCCCCATCAAACAGAA GGCCCGCTTCAAGATCCGAGATGCCAAGTGCCACCTCCGGCCCCACAGCCAGGCACGAGCAAAGGAGACC GCCAGGCAGCCGCTGCTGGACCACTGCCATGTGACTTTCGTGACCCTCAAGTGTGACTCCTCCAAGAAGA GGCGCCGTGGCCGCAAGTCCCCATCCAAGGAGGTGTCCCACATCACAGCAGAGTTTGAGATCGAGACAAA GATGGAAGAGGCCTCAGGTACATGCGAAGCGGACTGCTTGCGGAAGCGAGCAGAACAGAGCCTGCAGGCC GCCATCAAGACCCTGCGCAAGTCCATCGGCCGGCAGCAGTTCTATGTCCAGGTCTCAGGCACTGAGTACG AGGTAGCCCAGAGGCCAGCCAAGGCGCTGGAGGGGCAGGGGGCATGTGGCGCAGGCCAGGTGCTACAGGA CAGCAAATGCGTTGCCTGTGGGCCTGGCACCCACTTCGGTGGTGAGCTCGGCCAGTGTGTGTCATGTATG CCAGGAACATACCAGGACATGGAAGGCCAGCTCAGTTGCACACCGTGCCCCAGCAGCGACGGGCTTGGTC TGCCTGGTGCCCGCAACGTGTCGGAATGTGGAGGCCAGTGTTCTCCAGGCTTCTTCTCGGCCGATGGCTT CAAGCCCTGCCAGGCCTGCCCCGTGGGCACGTACCAGCCTGAGCCCGGGCGCACCGGCTGCTTCCCCTGT GGAGGGGGTTTGCTCACCAAACACGAAGGCACCACCTCCTTCCAGGACTGCGAGGCTAAAGTGCACTGCT CCCCCGGCCACCACTACAACACCACCACCCACCGCTGCATCCGCTGCCCCGTCGGCACCTACCAGCCCGA GTTTGGCCAGAACCACTGCATCACCTGTCCGGGCAACACCAGCACAGACTTCGATGGCTCCACCAACGTC ACACACTGCAAAAGTCAGCACTGCGGCGGCGAGCTTGGTGACTACACCGGCTACATCGAGTCCCCCAACT ACCCTGGCGACTACCCAGCCAACGCTGAATGCGTCTGGCACATCGCGCCTCCCCCAAAGCGCAGGATCCT CATCGTGGTCCCTGAGATCTTCCTGCCCATCGAGGATGAGTGCGGCGATGTTCTGGTCATGAGGAAGAGT GCCTCTCCCACGTCCATCACCACCTATGAGACCTGCCAGACCTACGAGAGGCCCATCGCCTTCACCTCCC GCTCCCGCAAGCTCTGGATCCAGTTCAAATCCAATGAAGGCAACAGCGGCAAAGGCTTCCAAGTGCCCTA TGTCACCTACGATGGTAAGATCCACTGTCTTCACGGCCCACTGTGCACGGCTCAGGCGGGGCCCTGGAGA CACAGAGATGAGTCGCACGTCCCCGCCCTCAGGGAGCTGCGACCTGGCAGGTACAGACCTGGAAGCAGAA CGAACACTGTCAGGGGCCAGAGCCAGACAGGCTGAGGGTGGTACCGGGTGGTACAGGCAAGACAGCGGTT AGTGGCCTCTGCAGGCTTCAGCTGAGGTGCTGCCCAAGCAGGGTTTTGAGGGCTAAATAGGGGGTTCTTA GTGAAACCCCGAGGAGGACAATACAGGTGCAGGGAGCCCCAGGTTCAAAGGCACAGA
In a search of public sequence databases, the NOVl nucleic acid sequence, located on chromosome 22ql3, demonstrates 88% identity to Mus Musculus EGF-related protein SCUBEl (Genbank AF276425). Public nucleotide databases include all GenBank databases and the GeneSeq patent database.
In all BLAST alignments herein, the "E- value" or "Expect" value is a numeric indication of the probability that the aligned sequences could have achieved their similarity to the BLAST query sequence by chance alone, within the database that was searched. For example, the probability that the subject ("Sbjct") retrieved from the NOVl BLAST analysis, e.g., Mus Musculus EGF-related protein SCUBEl, matched the Query NOVl sequence purely by chance is Lie -17. The Expect value (E) is a parameter that describes the number of hits one can "expect" to see just by chance when searching a database of a particular size. It decreases exponentially with the Score (S) that is assigned to a match between two sequences. Essentially, the E value describes the random background noise that exists for matches between sequences.
The Expect value is used as a convenient way to create a significance threshold for reporting results. The default value used for blasting is typically set to 0.0001. , hi BLAST 2.0, the Expect value is also used instead of the P value (probability) to report the significance of matches. For example, an E value of one assigned to a hit can be interpreted as meaning that in a database of the current size one might expect to see one match with a similar score simply by chance. An E value of zero means that one would not expect to see any matches with a similar score simply by chance. See, e.g., http://www.ncbi.nlm.nih.gov/Education/BLASTinfo/. Occasionally, a string of X's or N's will result from a BLAST search. This is a result of automatic filtering of the query for low- complexity sequence that is performed to prevent artifactual hits. The filter substitutes any low-complexity sequence that it finds with the letter "N" in nucleotide sequence (e.g., "NNNNNNNNNNNNN") or the letter "X" in protein sequences (e.g., "XXXXXXXXX"). Low-complexity regions can result in high scores that reflect compositional bias rather than significant position-by-position alignment. (Wootton and Federhen, Methods Enzymol 266:554-571, 1996).
The disclosed NOVl polypeptide (SEQ ID NO:2) encoded by SEQ ID NO:l has 965 amino acid residues and is presented in Table IB using the one-letter amino acid codes. Signal P, Psort and/or Hydropathy results predict that NOVl has a signal peptide and is likely to be localized outside the cell with a certainty of 0.3700. In other embodiments, NOVl may also be localized to the lysosome (lumen) with a certainty of 0.1900, the nucleus with a certainty of 0.1800, or in the endoplasmic reticulum (membrane) with a certainty of 0.1000. The most likely cleavage site for a NOVl signal peptide is between amino acids 23 and 24, at: RLA-GG.
Table IB
NOVl Polypeptide
SEQ ID NO:2
MGAAAVRWHLCVLLALGTRGRLAGGSGLPGSVDVDECSEGTDDCHIDAICQNTPKSYKCLCKPGYKGEGK QCEDIDECE DYYNGGCVHECINIPGNYRCTCFDGFMLAHDGH CLDVDECQDN GGCQQICVNAMGSYE CQCHSGFLLSDNQHTCIHRSNEGMNCMNKDHGCAHICRETPKGGVACDCRPGFDLAQNQKDCTLTCNYGN GGCQHSCEDTDTGPTCGCHQKYALHSDGRTCIETCAVNNGGCDRTCKDTATGVRCSCPVGFTLQPDGKTC KDINECLV NGGCDHFCRNTVGSFECGCRKGYKLLTDERTCQDIDECSFERTCDHICINSPGSFQCLCHR GYILYGTTHCGDVDECSMSNGSCDQGCVWTKGSYECVCPPGRRLHWNGKDCVETGKCLSRAKTSPRAQLS CSKAGGVESCFLSCPAHTLFVPQDSENSYVLSCGVPGPQGKALQKRNGTSSGLGPSCSDAPTTPIKQKAR FKIRDAKCHLRPHSQARAKETARQPLLDHCHVTFVTLKCDSSKKRRRGRKSPSKEVSHITAEFEIETKME EASGTCEADCLRKRAEQSLQAAIKTLRKSIGRQQFYVQVSGTEYEVAQRPAKALEGQGACGAGQVLQDSK CVACGPGTHFGGELGQCVSCMPGTYQDMEGQLSCTPCPSSDGLGLPGARNVSECGGQCSPGFFSADGFKP CQACPVGTYQPEPGRTGCFPCGGGLLTKHEGTTSFQDCEAKVHCSPGHHYNTTTHRCIRCPVGTYQPEFG QNHCITCPGNTSTDFDGSTNVTHCKSQHCGGELGDYTGYIESPNYPGDYPANAECVWHIAPPPKRRILIV VPEIFLPIEDECGDVLVMRKSASPTSITTYETCQTYERPIAFTSRSRKLWIQFKSNEGNSGKGFQVPYVT YDGKIHCLHGPLCTAQAGP RHRDESHVPALRELRPGRYRPGSRTNTVRGQSQTG
A search of sequence databases reveals that the NOVl amino acid sequence has 145 of 489 amino acid residues (29%) identical to, and 216 of 489 amino acid residues (44%) similar to, the 2489 amino acid residue ptnr:SPTREMBL-ACC:Q16744 protein from Homo sapiens (Human) (COMPLEMENT RECEPTOR 1). Public amino acid databases include the GenBank databases, SwissProt, PDB and PIR.
NOVl is expressed in at least the pituitary gland, the ovaries, and the trachea. This information was derived by determining the tissue sources of the sequences that were included in the invention including but not limited to SeqCalling sources, public EST sources, literature sources, and/or RACE sources.
Homologies to the above NOVl polypeptide will be shared by the other NOVl protein insofar as they are homologous to each other as shown below. The disclosed NOVl polypeptide has homology to the amino acid sequences shown in the BLASTP data listed in Table lC.
Figure imgf000021_0001
Figure imgf000022_0004
The homology between these and other sequences is shown graphically in the ClustalW analysis shown in Table ID. In the ClustalW alignment of the NOVl protein, as well as all other ClustalW analyses herein, the black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that maybe required to preserve structural or functional properties), whereas non-highlighted amino acid residues are less conserved and can potentially be altered to a much broader extent without altering protein structure or function.
Table ID. ClustalW Analysis of NOVl
1) Novel NOVl (SEQ ID NO:l)
2) gi|l273884θ| (SEQ ID NO:35)
3) gi|l0190748| (SEQ ID NO:36)
4) gi|9910154| (SEQ ID NO:37)
5) gi|5050926| (SEQ ID NO:38)
6) gi|l3518037| (SEQ ID NO:39)
10 20 30 40 50
Figure imgf000022_0001
gi|l0190748| iJE^GRNRPGAAiA^ lilH ij PPLLgLA^VPgg GRAAGPQEJSSS gi| 9910154 I Ug gGCGRPREARAJJlrtlSjLJjLPP - -BLAA&Vpl5DRGLT GpgElBBB5Bli
Figure imgf000022_0002
gi|l3518037| E[EKMLAGCF LILGQIViϊJPAEA|2EβSRgRglSR3RHARTHPQ AIjL s|
60 70 80 90 100
NOVl EEMBSBEttJiEHiffl lL!«) JAtKtetaiakl»Ma.ι> ι.aBliSi- gi|l2738840| l l ii!Tetøκl^Rh!«tøtWt)5gEΪiil-
Figure imgf000022_0003
gi| 9910154 I gi 150509261 fe!aτltWt«liιltVΛW»VVI--p^κlSBaιffl38^κi3^_d»Ma.»lι^ «|ES] gi 113518037 | CENKRADΪ(VF0IDiSRjvN5i,aDYAKVKE|ijlvSlL@PLlϊJ3GPDVTRVGIιLQ
110 120 130 140 150
NOVl ^WAWiiiE[«IIiH^iM:MM^>Ie.^akAiUi|MiiiM«ιU gi|l2738840| WeMH iDWraii. gi|l0190748| --E: ^Mti*tøjJDmD^UW^I>t4!Olim9i^^l>rø$l!l!ϊϊϊlϋL«>YM]-' gi| 9910154 I --T: flIPGNYRCTCFDGFM AHDGHNCLD. gi|5050926| -D; Jfefef«kti!iEBIi^*5leHrt;W(.«talιMaθtWtι»|e)Jik[«t*ιl< gi|l3518037| YGSTVK EFSLKϊTKRKSE^RA^KRMEHLSTGTMT@ω CQ Alis|lAFSE
160 170 180 190 200
NOVl BϋSbS- - gx 112738840 I g8gθfc.--| SotBTOallle '^oBlHsleiJa.iMt. tiiliiltt^.Jφii i^ gi 110190748 I BTBIliB-- aHTlSB^iileh'i^cBKEltta.atfettlVMi tl^kfelEiSiaiigfa gi| 9910154 I SSIB -- SHt^i^lleH>s.«lRiaκEl.a-allh.ι_tι.».tlιιl«^^t;fctE^I^B gi| 5050926 j BsEopM fflolι<A<ilftB!8t!il o!aHsMa.3tfelt_ t.i i^fel gi 113518037 | A§GARP RENVPRVJ3ravTDgRPQDSVAEVAAI^ E!TGIL3FAIGV@QVD
210 220 230 240 250
NOVl
Figure imgf000023_0001
gi|12738840| gι|l0190748| MMMiMJIsBnarifal&fflqfBSI- -HEM;HriJEfl^κil^ !MιBra8WeBBg5BI
Figure imgf000023_0002
gi| 5050926 I
Figure imgf000023_0003
260 270 280 290 300
NOVl g^ElgJD Bg5τSG_gϊloKHAffiϊl3!»!gaΕlifιlSI gi|l273884θ) !ϊ^ε!ϊ^τ^^GBI5oSfflA{lgfe!aaϊlτHι!5l gi|l0190748| lϊSfo!5 D_^EHs_^PθHκ«^ lHeBlsHL!5lREDTVLEVTESNTT5VVDGDK gi I 9910154 I IS^ElJ^E^EBIsl^ RH HBklil^Siaa'laloEGTV EGTESNATSVADGDK gi|5050926|
Figure imgf000023_0004
310 320 330 340 350
NOVl ιl«tAtιtlιelef J;]ιι_«ti4tιlAiraRf<-H ^rt^ιtiBWPBlet^i-^itW gi|l2738840| bι!4A<ιiιi.^ t>^J ii Απ^Rpg8^4^ι.<«lpB!^ιl i4Λta gi|l0190748| RVKR I_ E «k IkVMet ιl;W ιWs1^ gi I 9910154 I VKRR LME «A<iiliMef ti:lιι. i«»Wa57^Hfa5T^τW^^«k» ^I_ i<ιVJl1 gi|5050926| gi|l3518037| L^ME'HNgEQlflVMVPGSFVgQEYS AiJAElHKBvAVn
360 370 380 390 400
NOVI & gg^]ji^ι^ j5j^E3G ^SES3l3R':!:'^3I_3l^FE_3il-B gi 112738840 I [^I ]MN!S^ 3]Ξ^^ Q^SβΞ^θΞ^-Ξ_ΞΞ_i^^--l^ΞI_^F ES3-Ξ
Figure imgf000024_0001
gi I 5050926 I gi 113518037 I Y^SE|JJH^ESΪ!Eggl D^YL3QgHE@EJΑ[ΪNPJ3KτSτKl[3γg^SSNHGS
410 420 430 440 450
NOVl i iBHSsigasraoSLBHrfHiBBiG - τ8Sπ8gBlv.15t8^sWsBDR3-{-gMκltl
Figure imgf000024_0002
gi|5050926| gi|l3518037| Q^Eg^TDD^S^gL^FΪ3ΪNPDKK gRRliϊ g li!J]KPGE'EHEBf(J]MEE
460 470 480 490 500
NOVl EBπBlvflplfflRϊ^itøMG.5HgfeGK^^^^
91 12738840| ^^vgPJ^R^JIHQiSEB&NGCLSI^K l AQBsEGiS ^VgNSJJ gi 10190748| ^SQBiE^κ3I_ElκS3_Si^KGLLpτN-^pRvsBHSGiSs^G GEi_i3 gi 9910154] ^^oBH!EF^SIHκ!2S&KGFppΦ-1^!pβvsBHi3GESs_SG?RS_S gi 5050926 I gi 135180371 ^YgRgHRS TJjDp[JJGiSTgS)A)HCAQQDHGCE^ CijN T--EpSFVC
510 520 530 540 550
NOVl siaPAHT FVPODSEtaRYVtlSGGKpGpHnKAT,o!5RlSflTaS LG -_H5lDAPT gi] 12738840 I sSLGHSLFMP-DSEySYIJECGEPG QGK gRJΪlGTSSS GPGS^lAPT
Figure imgf000024_0003
gi|5050926| gi|l3518037| Qffls EGFlS5jEDUKTCSRVDYCLLgDHG C
560 570 580 590 600
NOVl TPjKQKARFKIRpAKCHLRgHSQARASEΪA^QPLLDHCHVT^VTJΪKgD^S gi|l2738840| TP fQKA FKIRW KCHLQgRSQER lOU' lRHP D CHV fVTH gD^S gi|l0190748| gi| 9910154 I
Figure imgf000024_0004
gi|5050926| gi|l3518037| gYSCVNMDRSFACQC@EGHVLRSDGK CAKJjDSφli
610 620 630 640 650
NOVl S -- S2κ^PS[^SHEBAS31lil KME]3ASJG|rSEADg'RiS2AiQs|ϊq
Figure imgf000024_0005
660 670 680 690 700
Figure imgf000025_0001
gi|l273884θ| A l^lS^Kø.TGJ^SlYVlV-^TEYEViQΪcf ^LEG-TGT^I^I QJG gi|l0190748| Kgϊ^ li^VHSE^lgώslMN b SKEilP^TSERQAEsEv®3^11 gi I 9910154 I κ L^ JJKRi^SEg|H @IjsSMD DMg gs^VSGQHEE ^ ^GHE
Figure imgf000025_0002
710 720 730 740 750
NOVl !^ABGP.SfeFGBELGP^S^PΞ^^M!g__CJIl.gHτiϊ5rasSDGLBI- - -ijp
Figure imgf000025_0003
gi|l0190748| d^s!8R !8HYYDl^RE^JLBPN^Fl^E!55!^τBEl3SHRPG sElA KTP g I 9910154 I Q^sgRA^Ϊr2D@SQEΪ^iLEPNJ33F jEHvτSE|S3RPENLgS KlS gi|5050926| gi|l3518037| I^NNGN SYICκgSE@FVLAES5SήR-BκκSTEGp- "ΪD
760 770 780 790 800
NOVl G^RiJ]gE^SQSs|EF^SDSSκ[^*Spiβ2iΞE'I,EΞ3GSFPBS^!L gi|l273884θ| G^iSylE^SQBsγ'&lSiκi^aSpiβSEpSSSGSFP-^-SL gi 110190748 ] gi I 9910154 I Egw SD^^LgQ|^E El!βA[^LSAkS3FS3DvS33s@LS^^!P gx|5050926| gi|l3518037| LVFVΪDGSKSLGEENFEWKQIVTGIIDSS-SISSKAASVG LQYSTQVH
810 820 830 840 850
NOVl j^E^- -ττ^EϊEa-κΞHE^HΞS_EiSSS_E^^-__iSΞ3i-EδiSi| gi|l2738840| WSSTSI- - τ MA.ι!«l^κ^! ^J«lMitiιιlιιhιli.;M^ JvS-iιtf^j^jel Ste gi|l0190748| - -AτMA.J 3τ«ffiot itJeillF(ii ιlιιi..J J 5h^-lιιW8.!i.!ill l^ gi I 9910154 I - -AτMA.J aτ^o! ^J.ilF^t ιl)il.i;T ;l JιS-lιιW5.5.jlκfc gi|5050926| gx|l3518037| 0BFTLR FNgAi_J3M^ A2 HMKYMGKG^MjG AI,^HMFE SlTQGESA P
860 870 880 890 900
Figure imgf000025_0004
gi|5050926| gi|l3518037| FS^RVPRAAIVJ3TDGRAQDDVSE ASKAI^ANGIEMSAVGVG K^IE
910 920 930 940 950
NOVl i^τlft^ΑHJJi4J;tA* ωd!A^Λ^ι<MFaaB {B-^ gi|l2738840| .^7i^πΑ 5tøi_lriAΛ WJaHatøfl^ElSt^ gi|l0190748| IMτlffiτ^iH _l.llA_tMd;>H; Ji>&i^^ gi I 9910154 I gi|5050926| gi|l3518037| |ELQEE3ASEgτNKH[jFYAEnFSTMDi lSEKLKKGICϊ!A EDSDGRQDSPA 960 970 980 990 1000
Figure imgf000026_0001
gi I 9910154 j fA^^r^W^iM^KliCTiSπira^TOGMA^AW^W-llk' ilEDYOE IEDIV gi | 5050926 I
Figure imgf000026_0002
1010 1020 1030 1040 1050
NOVl LHGPJJCT& GP RriξωislάrpS RB RPGRYRPGSRTNTVRG^SQTG - - gx|l273884θ| CHSP|JCA§QG AWGLfeNfiLHWpSsDRAQTQR@KLGLGNAETQGV gx|l0190748| RDGRJJYAfENHQEI φϊCKL !K§LFθV AHPgNYFKYTAQESEtSi FPRSF gi|9910154| RDGRS A§E HQEI ^ 3 KLjκSLFigvLAHPgM FK TAQES lMF RSF gi|5050926| gi|l3518037| STQKJJSHgTKPSGSPLS^KHDQCKCBNLIMFSN ANEEVRK TQRLEEMT
1060
NOVl gi|l273884θ| gi|l0190748| IRLLRSKVSRFLRPYK gi I 9910154 I IR RSKVSRFLRPYK gi|5050926| gi|l3518037| QRMEALENRLRYR
The presence of identifiable domains in NOVl, as well as all other NOVX proteins, was determined by searches using software algorithms such as PROSITE, DOMAIN, Blocks, Pfam, ProDomain, and Prints, and then determining the Interpro number by crossing the domain match (or numbers) using the Interpro website (http:www.ebi.ac.uk/ interpro). DOMAIN results for NOVl as disclosed in Table IE, were collected from the Conserved Domain Database (CDD) with Reverse Position Specific BLAST analyses. This BLAST analysis software samples domains found in the Smart and Pfam collections. For Table IE and all successive DOMAIN sequence alignments, fully conserved single residues are indicated by black shading or by the sign (|) and "strong" semi-conserved residues are indicated by grey shading or by the sign (+). The "strong" group of conserved amino acid residues may be any one of the following groups of amino acids: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW.
Table IE lists the domain description from DOMAIN analysis results against NOVl. This indicates that the NOVl sequence has properties similar to those of other proteins known to contain this domain. Table IE. Domain Analysis of NOVl
gnl|Smart|smart00042, CUB, Domain first found in Clr, Cls, uEGF, and bone moφhogenetic protein; This domain is found mostly among developmentally-regulated proteins. Spermadhesins contain only this domain.
CD-Length = 114 residues, 99.1% aligned Score = 85.5 bits (210), Expect = le-17
Query : 799 CGGΞ GDYTGYIESPNYPGDYPA AECWHIAPPPKRRILIWPEIFLPIEDΞCG-DVLV 857
CGG L +G I SPNYP YP N CVW 1+ PP Rl 4- + D C D + Sbj ct : 1 CGGTLTASSGTITSPNYPNSYPKF LNCVWTISAPPGYRIE KFTDFDLESSDNCTYDYVE 60
Query : 858 MRKSASPTSITTYETCQTYERPIAFTSRSRKLWIQFKSNEGNSGKGFQVPYVT 910 + S +S C + P +S S + + F S+ +GF Y
Sbj ct : 61 IYDGPSTSSPL GRFCGSE PPPIISSSSNSMTVTFVSDSSVQKRGFSARYSA 113
The epidermal growth factor (EGF) superfamily comprises a diverse group of proteins that function as secreted signaling molecules, growth factors, and components of the extracellular matrix, many with a role in vertebrate development. A novel mammalian gene encoding an EGF-related protein with a CUB (Cls-like) domain that defines a new mammalian gene family. The SCUBEl (signal peptide-CUB domain-EGF-related 1) gene was isolated from a developing mouse urogenital ridge cDNA library and is expressed prominently in the developing gonad, nervous system, somites, surface ectoderm, and limb buds. Mouse SCUBEl was mapped to chromosome 15 and shown that it is orthologous to a human gene in the syntenic region of chromosome 22ql 3. EGF-related proteins with Cls-like (CUB) domains have been reported. The CUB domain is found in 16 functionally diverse proteins such as the dorso-ventral patterning protein tolloid, bone morphogenetic protein-1, a family of spermadhesins, complement subcomponents Cls/Clr and the neuronal recognition molecule A5. Most of these proteins are known to be involved in developmental processes. The second domain is found mostly among developmentally-regulated proteins and spermadhesins.
The disclosed NOVl nucleic acid of the invention encoding an EGF-Related Protein
(SCUBEl)-like protein includes the nucleic acid or a fragment thereof whose sequence is provided in Table 1 A. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 1 A while still encoding a protein that maintains its EGF-Related Protein (SCUBEl)-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 30% percent of the bases may be so changed.
The disclosed NOVl protein of the invention includes an EGF-Related Protein (SCUBEl)-like protein whose sequence is provided in Table IB. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table IB while still encoding a protein that maintains its EGF-Related Protein (SCUBEl)-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 12% percent of the residues may be so changed.
The invention further encompasses antibodies and antibody fragments, such as Fa or (Fab)2,that bind immunospecifically to any of the proteins of the invention.
The above defined information for this invention suggests that this EGF-Related Protem (SCUBEl)-like protein (NOVl) may function as a member of a EGF-Related Protein (SCUBEl)-like protein family. Therefore, the NOVl nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The NOVl nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in cancer including but not limited to various pathologies and disorders as indicated below. For example, a cDNA encoding EGF-Related Protein (SCUBEl)-like protein (NOVl) may be useful in gene therapy, and the EGF-Related Protein (SCUBEl)-like protein (NOVl) may be useful when administered to a subject in need thereof. By way of nonlimiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from cancer, trauma, viral/bacterial/parasitic infections, endometriosis, fertility, astlima, allergy, endocrine dysfunctions, diabetes, obesity, growth and reproductive disorders and other diseases, disorders and conditions of the like. The NOVl nucleic acid encoding the EGF-Related Protein (SCUBEl)-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOVl nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel NOVl substances for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. The disclosed NOVl proteins have multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOVl epitope is from about amino acids 400 to 450. hi other embodiments, a NOVl epitope is from about amino acids 500 to 600, from about 1000-1100, from about 1500-1600 and 2500-2800. These novel proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
NOV2
NOV2 includes four adipocyte complement-related Clq Tumor Necrosis Factor-like proteins and nucleic acids encoding the same. The disclosed sequences are identified herein as NOV2a, NOV2b, NOV2c, and NOV2d.
NOV2a
A disclosed NO V2a nucleic acid of 874 nucleotides identified as SEQ IDNO:3 (also referred to as CG55724-01) encoding an adipocyte complement-related Clq Tumor Necrosis Factor-like protein is shown in Table 2A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 11-13 and ending with a TGA codon at nucleotides 674-676. Putative upstream and downstream untranslated regions are underlined.
Figure imgf000029_0001
CTCGGCGGCACGCACCAGCCCCCTGGAGGGCACGTCGGAGATGGCGGTGACCTTCGACAA 300
GGTGTACGTGAACATCGGGGGCGACTTCGACGCGGCGGCCGGCGTGTTCCGCTGCCGTCT 360
GCCCGGCGCCTACTTCTTCTCCTTCACGCTGGGCAAGCTGCCGCGTAAGACGCTGTCGGT 420
TAAGCTGATGAAGAACCGCGACGAGGTGCAGGCCATGATTTACGACGACGGCGCGTCGCG 480
GCGCCGCGAGATGCAGAGCCAGAGCGTGATGCTGGCCCTGCGGCGCGGCGACGCCGTCTG 540
GCTGCTCAGCCACGACCACGACGGCTACGGCGCCTACAGCAACCACGGCAAGTACATCAC 600
CTTCTCCGGCTTCCTGGTGTACCCCGACCTCGCCCCCGCCGCCCCGCCGGGCCTCGGGGC 660
CTCGGAGCTACTGTGAGCCCCGGGCCAGAGAAGAGCCCGGGAGGGCCAGGGGCGTGCATG 720
CCAGGCCGGGCCCGAGGCTCGAAAGTCCCGCGCGAGCGCCACGGCCTCCGGGCGCGCCTG 780
GACTCTGCCAATAAAGCGGAAAGCGGGCACGCGCAGCGCCCGGCAGCCCAGGACTAAGCC 840 GAATCTGCAAAATCCATCAACTGCCGGCGCTGAA
The disclosed NOV2a nucleic acid sequence, localized to chromosome 11, has 294 of 485 bases (60%) identical to a gb:GENBANK-ID:AF192499|acc:AF192499.1 mRNA from Mus musculus (Mus musculus putative secreted protein ZSIG37 (Zsig37) mRNA, complete eds).
A NOV2a polypeptide (SEQ ID NO:4) encoded by SEQ ID NO:3 has 221 amino acid residues and is presented using the one-letter code in Table 2B. Signal P, Psort and/or Hydropathy results predict that NOV2b does not have a signal peptide and the NOV2a polypeptide is likely to be localized to the cytoplasm with a certainty of 0.4500. I other embodiments, NOV2a may also be localized to peroxisomal microbodies with a certainty of 0.2688, lysosomes with a certainty of 0.1937, or the mitochondrial matrix space with a certainty of 0.1000.
Figure imgf000030_0001
The disclosed NO V2a amino acid sequence has 55 of 158 amino acid residues (34%) identical to, and 84 of 158 amino acid residues (53%) identity to the 244 amino acid residue pntr:SWISSPROT ACC:Q15848 protein from Homo sapiens (Human) (30 kDa adipocyte complement related protein precursor, ACRP30). The NOV2a adipocyte complement-related protein precursor disclosed in this invention is expressed in at least the following tissues: testis, kidney, whole embryo. This information was derived by determining the tissue sources of the sequences that were included in the invention including but not limited to SeqCalling sources, public EST sources, literature sources, and/or RACE sources. In addition, the sequence is predicted to be expressed in the following tissues because of the expression pattern of (GENBANK-ID: gb:GENBANK-ID:AF192499|acc:AF192499.1) a closely related Mus musculus putative secreted protein ZSIG37 (Zsig37) mRNA, complete eds homolog in species Mus musculus: adipocytes.
NOV2b
A disclosed NOV2b nucleic acid of 1277 nucleotides (also referred to as CG55724-03) encoding a complement related Clq Tumor Necrosis Factor-like protein is shown in Table 2C as SEQ ID NO:5. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 225-227 and ending with a TGA codon at nucleotides 1077-1079. Putative upstream and downstream untranslated regions are underlined.
Table 2C
NOV2b Polynucleotide
SEQ ID NO:5
GAATTCGGCACGAGGCGCCCGGCCCCTGGCCCCAGCACCCTGTCCGCTGCCGCCTCAGAG 60
CCGGGAAAAGCAGCCGGAGCCCCCGCCGCCCCTGCCGCAGCGCGGGCGGTCAGCGCGCAG 120
CCCGGCACCCGCAGCCTGCAGCCTGCAGCCCGCAGCCCGCAGCCCGGAGCCAGATCGCGG 180
GCTCAGACCGAACCCGACTCGACCGCCGCCCCCAGCCAGGCGCCATGCTGCCGCTTCTGC 240
TGGGCCTGCTGGGCCCAGCGGCCTGCTGGGCCCTGGGCCCGACCCCCGGCCCGGGATCCT 300
CTGAGCTGCGCTCGGCCTTCTCGGCGGCACGCACCACCCCCCTGGAGGGCACGTCGGAGA 360
TGGCGGTGACCTTCGACAAGGTGTACGTGAACATCGGGGGCGACTTCGATGTGGCCACCG 420
GCCAGTTTCGCTGCCGCGTGCCCGGCGCCTACTTCTTCTCCTTCACGGCTGGCAAGGCCC 480
CGCACAAGAGCCTGTCGGTGATGCTGGTGCGAAACCGCGACGAGGTGCAGGCGCTGGCCT 540
TCGACGAGCAGCGGCGGCCAGGCGCGCGGCGCGCAGCCAGCCAGAGCGCCATGCTGCAGC 600
TCGACTACGGCGACACAGTGTGGCTGCGGCTGCATGGCGCCCCGCAGTACGCGCTAGGCG 660
CGCCCGGCGCCACCTTCAGCGGCTACCTAGTCTACGCCGACGCCGAGTTCGTCAACATTG 720
GCGGCGACTTCGACGCGGCGGCCGGCGTGTTCCGCTGCCGTCTGCCCGGCGCCTACTTCT 780
TCTCCTTCACGCTGGGCAAGCTGCCGCGTAAGACGCTGTCGGTTAAGCTGATGAAGAACC 840
GCGACGAGGTGCAGGCCATGATTTACGACGACGGCGCGTCGCGGCGCCGCGAGATGCAGA 900
GCCAGAGCGTGATGCTGGCCCTGCGGCGCGGCGACGCCGTCTGGCTGCTCAGCCACGACC 960
ACGACGGCTACGGCGCCTACAGCAACCACGGCAAGTACATCACCTTCTCCGGCTTCCTGG 1020
TGTACCCCGACCTCGCCCCCGCCGCCCCGCCGGGCCTCGGGGCCTCGGAGCTACTGTGAG 1080
CCCCGGGCCAGAGAAGAGCCCGGGAGGGCCAGGGGCGTGCATGCCAGGCCGGGCCCGAGG 1140
CTCGAAAGTCCCGCGCGAGCGCCACGGCCTCCGGGCGCGCCTGGACTCTGCCAATAAAGC 1200
GGAAAGCGGGCACGCGCAGCGCCCGGCAGCCCAGGACTAAGCCGAATCTGCAAAATCCAT 1260 CAACTGCCGGCGCTGAA 1277
The disclosed NOV2b nucleic acid sequence, localized to chromosome 11, has 767 of 814 bases (94%) identical to a gb:GENBANK-ID:AF329838|acc:AF329838.1 mRNA from Homo sapiens (Homo sapiens complement Clq Tumor Necrosis Factor-related protein CTRP4 mRNA, complete eds).
A NOV2b polypeptide (SEQ ID NO:6) encoded by SEQ ID NO:5 has 284 amino acid residues and is presented using the one-letter code in Table 2D. Signal P, Psort and/or Hydropathy results predict that NOV2b has a signal peptide and is likely to be localized outside the cell with a certainty of 0.4801. In other embodiments, NOV2b may also be localized to microsomal bodies with a certainty of 0.2178, the endoplasmic reticulum (membrane or lumen) with a certainty of 0.1000. The most likely cleavage site for a NOV2b signal peptide is between amino acids 16 and 17, at: CWA-LG.
Figure imgf000032_0001
The disclosed NOV2b amino acid sequence has 55 of 158 amino acid residues (34%) identical to, and 84 of 158 amino acid residues (53%) identity to the 244 amino acid residue pntr:SPTREMBL ACC:Q9BXJ3 protein from Homo sapiens (Human) (complement Clq Tumor Necrosis Factor-related protein). The NOV2b complement-Clq tumor necrosis factorlike gene disclosed in this invention is expressed in at least the following tissues: brain, germ cell, kidney, pooled, testis, whole embryo. Expression information was derived from the tissue sources of the sequences that were included in the derivation of the sequence of CuraGen Ace. No. CG55724-03, CG55724-04, or CG55724-06.
NOV2c
A disclosed NOV2c nucleic acid of 1322 nucleotides (also referred to as CG55724-04) encoding a complement related Clq Tumor Necrosis Factor-like protein is shown in Table 2E as SEQ ID NO:7. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 225-227 and ending with a TGA codon at nucleotides 1122-1124. Putative upstream and downstream untranslated regions are underlined.
Table 2E
NO 2c Polynucleotide
SEQ ID NO:7
GAATTCGGCACGAGGCGCCCGGCCCCTGGCCCCAGCACCCTGTCCGCTGCCGCCTCAGAG 60
CCGGGAAAAGCAGCCGGAGCCCCCGCCGCCCCTGCCGCAGCGCGGGCGGTCAGCGCGCAG 120
CCCGGCACCCGCAGGCTGCAGCCTGCAGCCCGCAGCCCGCAGCCCGGAGCCAGATCGCGG 180
GCTCAGACCGAACCCGACTCGACCGCCGCCCCCAGCCAGGCGCCATGCTGCCGCTTCTGC 240
TGGGCCTGCTGGGCCCAGCGGCCTGCTGGGCCCTGGGCCCGACCCCCGGCCCGGGATCCT 300
CTGAGCTGCGCTCGGCCTTCTCGGCGGCACGCACCACCCCCCTGGAGGGCACGTCGGAGA 360
TGGCGGTGACCTTCGACAAGGTGTACGTGAACATCGGGGGCGACTTCGATGTGGCCACCG 420
GCCAGTTTCGCTGCCGCGTGCCCGGCGCCTACTTCTTCTCCTTCACGGCTGGCAAGGCCC 480
CGCACAAGAGCCTGTCGGTGATGCTGGTGCGAAACCGCGACGAGGTGCAGGCGCTGGCCT 540
TCGACGAGCAGCGGCGGCCAGGCGCGCGGCGCGCAGCCAGCCAGAGCGCCATGCTGCAGC 600
TCGACTACGGCGACACAGTGTGGCTGCGGCTGCATGGCGCCCCGCACTACGCGCTAGGCG 660
CGCCCGGCGCCACCTTCAGCGGCTACCTAGTCTACGCCGACGCCGACGCTGGCCCCGGGC 720
CGCGGCACCAACCACTCGCCTTCGACACCGAGTTCGTCAACATTGGCGGCGACTTCGACG 780
CGGCGGCCGACGTGTTCCGCTGCCGTCTGCCCGGCGCCTACTTCTTCTCCTTCACGCTGG 840 GCAAGCTGCCGCGTAAGACGCTGTCGGTTAAGCTGATGAAGAACCGCGACGAGGTGCAGG 900
CCATGATTTACGACGACGGCGCGTCGCGGCGCCGCGAGATGCAGAGCCAGAGCGTGATGC 960
TGGCCCTGCGGCGCGGCGACGCCGTCTGGCTGCTCAGCCACGACCACGACGGCTACGGCG 1020
CCTACAGCAACCACGGCAAGTACATCACCTTCTCCGGCTTCCTGGTGTACCCCGACCTCG 1080
CCCCCGCCGCCCCGCCGGGCCTCGGGGCCTCGGAGCTACTGTGAGCCCCGGGCCAGAGAA 1140
GAGCCCGGGAGGGCCAGGGGCGTGCATGCCAGGCCGGGCCCGAGGCTCGAAAGTCCCGCG 1200
CGAGCGCCACGGCCTCCGGGCGCGCCTGGACTCTGCCAATAAAGCGGAAAGCGGGCACGG 1260
GCAGCGCCCGGCAGCCCAGGACTAAGCCGAATCTGCAAAATCCATCAACTGCCGGCGCTG 1320 AA
The disclosed NO V2c nucleic acid sequence, localized to chromosome 11, has 949 of 1136 bases (83%) identical to a gb:GENBANK-ID:AF329838|acc:AF329838.1 mRNA from Homo sapiens (Homo sapiens complement Clq Tumor Necrosis Factor-related protein CTRP4 mRNA, complete eds).
A NOV2c polypeptide (SEQ ID NO:8) encoded by SEQ ID NO:7 has 299 amino acid residues and is presented using the one-letter code in Table 2F. Signal P, Psort and/or Hydropathy results predict that NOV2c has a signal peptide and is likely to be localized outside the cell with a certainty of 0.4801. In other embodiments, NOV2c may also be localized to microsomal bodies with a certainty of 0.2178, the endoplasmic reticulum (membrane or lumen) with a certainty of 0.1000. The most likely cleavage site for aNOV2c signal peptide is between amino acids 16 and 17, at: CWA-LG.
Table 2F
NOV2c Polypeptide
SEQ ID NO:8
MLP LLGLLGPAAC ALGPTPGPGSSELRSAFSAARTTPLEGTSEMAVTFD VYVNIGGD 60
FDVATGQFRCRVPGAYFFSFTAGKAPHKS SVMLVRNRDEVQA AFDEQRRPGARRAASQ 120
SAM QLDYGDTV LRLHGAPHYALGAPGATFSGYLVYADADAGPGPRHQP AFDTEFVNI 180
GGDFDAAADVFRCR PGAYFFSFT GKLPRKTLSVKLM NRDEVQAMIYDDGASRRREMQ 240 SQSVMLALRRGDAVWL SHDHDGYGAYSNHGKYITFSGFLVYPDLAPAAPPG GASEL
The disclosed NOV2c amino acid sequence has 164 of 170 amino acid residues (96%) identical to, and 164 of 170 amino acid residues (96%) identity to the 329 amino acid residue pntr:SPTREMBL ACC:Q9BXJ3 protein from Homo sapiens (Human) (complement Clq Tumor Necrosis Factor-related protein). The NOV2c complement-Clq tumor necrosis factorlike gene disclosed in this invention is expressed in at least the following tissues: brain, germ cell, kidney, pooled, testis, whole embryo. Expression information was derived from the tissue sources of the sequences that were included in the derivation of the sequence of CuraGen Ace. No. CG55724-03, CG55724-04, or CG55724-06.
NOV2d A disclosed NO V2d nucleic acid of 409 nucleotides (also referred to as CG55724-06) encoding a complement related Clq Tumor Necrosis Factor-like protein is shown in Table 2G as SEQ ID NO:X. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 4-6 and ending with a TGA codon at nucleotides 403-405. Putative upstream and downstream untranslated regions are underlined.
Table 2G
NOV2d Polynucleotide
SEQ ID NO:9
ATTATGCTGCCGCTTCTGCTGGGCCTGCTGGGCCCAGCGGCCTGCTGGGCCCTGGGCCCG 60
ACCCCCGGCCCGGGATCCTCTGAGCTGCGCTCGGCCTTCTCGGCGGCACGCACCACCCCC 120
CTGGAGGGCACGTCGGAGATGGCGGTGACCTTCGACAAGGTGTACGTGAACATCGGGGGC 180
GACTTCGATGTGGCCACCGGCCAGTTTCGCTGCCGCGAGATGCAGAGCCAGAGCGTGATG 0
CTGGCCCTGCGGCGCGGCGACGCCGTCTGGCTGCTCAGCCACGACCACGACGGCTACGGC 300
GCCTACAGCAACCACGGCAAGTACATCACCTTCTCCGGCTTCCTGGTGTACCCCGACCTC 360
GCCCCCGCCGCCCCGCCGGGCCTCGGGGCCTCGGAGCTACTGTGAGCCC 409
The disclosed NOV2d nucleic acid sequence, localized to chromosome 11, has 239 of 260 bases (91%) identical to a gb:GENBANK-ID:AF329838|acc:AF329838.1 mRNA from Homo sapiens (Homo sapiens complement Clq Tumor Necrosis Factor-related protein CTRP4 mRNA, complete eds).
A NOV2d polypeptide (SEQ ID NO: 10) encoded by SEQ ID NO:9 has 133 amino acid residues and is presented using the one-letter code in Table 2H. Signal P, Psort and/or Hydropathy results predict that NOV2d has a signal peptide and is likely to be localized outside the cell with a certainty of 0.4801. In other embodiments, NOV2d may also be localized to microsomal bodies with a certainty of 0.1972, the endoplasmic reticulum (membrane or lumen) with a certainty of 0.1000. The most likely cleavage site for a NOV2d signal peptide is between amino acids 16 and 17, at: CWA-LG.
Figure imgf000034_0001
The disclosed NOV2d amino acid sequence has 164 of 170 amino acid residues (96%) positives to, and 164 of 170 amino acid residues (96%) positives to the 329 amino acid residue pntπSPTREMBL ACC:Q9BXJ3 protein from Homo sapiens (Human) (complement Clq Tumor Necrosis Factor-related protein). The NOV2d complement-Clq tumor necrosis factorlike gene disclosed in this invention is expressed in at least the following tissues: brain, germ cell, kidney, pooled, testis, whole embryo. Expression information was derived from the tissue sources of the sequences that were included in the derivation of the sequence of CuraGen Ace. No. CG55724-03, CG55724-04, or CG55724-06.
The disclosed NOV2 nucleic acids of the present invention are expressed in at least bone marrow, brain, thalamus, testis, lung, kidney, and germ cells. This information was derived by determining the tissue sources of the sequences that were included in the invention. SeqCalling sources: Adrenal gland/Suprarenal gland, Amygdala, Bone, Bone Marrow, Brain, Colon, Coronary Artery, Dennis, Epidermis, Foreskin, Hair Follicles, Heart, Hippocampus, Hypothalamus, Kidney, Liver, Lung, Lymph node, Lymphoid tissue, Mammary gland/Breast, Esophagus, Ovary, Pancreas, Parathyroid Gland, Peripheral Blood, Pineal Gland, Pituitary Gland, Placenta, Prostate, Retina, Salivary Glands, Small Intestine, Spleen, Stomach, Testis, Thalamus, Thymus, Tonsils, Trachea, Umbilical Vein, and Uterus.
NOV2 also has homology to the amino acid sequences shown in the BLASTP data listed in Table 21.
Figure imgf000035_0001
gi 116550291 [dbj | BAB unnamed 248 36 49 2e-16 70947. ll (AK055541) protein product [Homo sapiens]
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 23.
Table 2J. ClustalW Analysis of NO 2 i) NOV2a (SEQ ID NO: 3)
2) NOV2b (SEQ ID NO: 5) 3) NOV2C (SEQ ID NO: 7} 4) NOV2d (SEQ ID NO: 9) 5) gi|l3994273 (SEQ ID NO:40) 6) gijl2835488 (SEQ ID NO: 41) 7) gi|l3385666 (SEQ ID NO:42) 8) gi|l3994278 (SEQ ID NO:43) 9) gi 116550291 (SEQ ID NO:44)
10 20 30 40 50
NOV2
KOV2b
NOV2C
NOV2d gi|l3994273| gi|l2835488] gi|l338566S| gi|l3994278| MQ LRVRESPGEATGHRVTMGTAALGPVAALLLFLLMCEIPMVELTFDR gi|l655029l|
SO 70 80 90 100
NOV2a -HP GLV
NOV2b -- -l8HpiliιL[e|
NOV2C -Mpffriifl
NOV2d -ICTIPJILL|S|
Figure imgf000036_0001
gi 112835488] -USojjDYg gi 113994278 I AVASGCQRCCDSEDPLDPAHrøSSASSSGRPHALPEIRPYINITpjJKGDK gi 1165502911 MYPA TAV--PQINITΪ|JκGEI!
110 120 130 140 150
NOV2a SG — AJJPCSVEQS{^PEDEALRRSQMRKGSLSVVDAVSGP[JΓ{3PGLPGR
NOV2b LBGP|ACWJJSE
Figure imgf000036_0002
NOV2d LUGPIACMEHEI gi|l3994273| LBGPE3AO«EEEI @τg-
Figure imgf000036_0003
gi 113994278 | DPGPMG3PGYMGREGPQG--E-- --PGPQGS GDKGEMGδS- gi 1165502911 DRGDRGJJQGKYGKTGSAG--A-- • -RGHTGPKGQKGSMGA0-
160 170 180 190 200
NOV2a GRAGLSGKNGFPlelDRSaAMl;fet!>3fe M;i)l-il.dlEfesiB A- -EτS3κffl
NOV2b -EBG sEl l3^iH3life i>; ιLnl3lElgτsfe'MA Eτ||![κj5
NOV2C -l33 ^.ql5lτMJiafeaA5lιιlιi-JlEl8-3S!BMA ETB
Figure imgf000037_0001
gi| 13994273 I -B3Gssl5lL!3^Jal5fe J; »Lnl3lElel'iJsBMA ETSJKB gi|l2835488| -ffiAapjgpl;WJJ^ J;llfesilvte!SDAAPGPRHRPl]Al3a E gi|l3385666| -I^A^pigpl.WJJ^ J;J>tesfflvlBSDAAPGPRHRPtøl3aTE gi|l3994278| -HΑPC0KRFFjragVGl3κπAfflHSGE JF0T k_L||E!Jj gi|l655029l| -BlERCκsHΥΑBBEIvGigκκl3M'Hsιmγγoτ EI[33 E
210 220 230 240 250
Figure imgf000037_0002
N0V2C -Ξ ilι<c_t.t tlvgiBlθT3raSigVPGAYFFSFTAGKA HKS SVMLVRMRDEVOA
N0V2d KefeiJaltl CTBlnBaa-
Figure imgf000037_0003
gi|l3994278| ;FH 3cS3κi_ϊ_EQiϊAAp -
Figure imgf000037_0004
260 270 280 290 300
N0V2a
N0V2b LAFDEQRRPGARRAASQSAMLQLDYGDTV LRLHGAPQYALGAPGATFSG
N0V2C LAFDEQRRPGARRAASQSAMLQLDYGDTVWLRLHGAPHYALGAPGATFSG
N0V2d gi 113994273 | LAFDEQRRPGARRAASQSAMLQLDYGDTV LRLHGAPHYALGAPGATFSG
gi|l3385666| gi|l3994278| gi|l655029l|
310 320 330 340 350
N0V2a
N0V2b YLVYADA
N0V2C YLVYADADAGP- -GPRHQPLAF
N0V2d gι| 13994273 I YLVYADADADAPARGPPAPPEPRSAFSAARTRSLVGSDAGPGPRHQPLAF gi|l2835488| gi|l3385666| gi|l3994278| gi|l655029l|
360 370 380 390 400 NOV2a
NOV2b --EFVNIGGDFDAAAGVFRCR
NOV2C DTEFVNIGGDFDAAADVFRCB LPGAYFFSFTLGKLPRKTLSVKL K RDE
NOV2d gi 113994273 I . DTEFVNIGGDFDAAAGVFRCK iTFFSFTLGKLPRKTLSVKLMKNRD gi|l2835488| gi|l3385666| LPGAYFFSFTLGKLPRKTLSVKLMKNRDE gi 113994278 I |JR@lEE^LN&SWNYJ ά|E!H5qκ§ gi 1165502911 vt^LMaMLMΛfflT MoBETΩ^H-lBMEEig
410 420 430 440 450
NOV2a _»w»Bttjaι>Ai«)a?wιfe ii.ιw«B>ii»wiiτewefeVft NOV2b !ιfM»κtfawΑii)3aafeVi'.><wιaiiiBnreHtEw»
NOV2C jTOWt tjawAiiiarTeroawiwiMiiB^iTwefev^
NOV2d ^iM. B3aa^lW*feti.li.l ' A^--■ gi 113994273 I QAMIYDDGASRRRE OSOSVML s*M«.\\^,M:\i,.i..WMMz gi|l2835488| H»ym*tιmRtf mm τs*TAmt*viΛaati,ΛmtΛi^utiimtvmsκ» gi|l3385666| gi 113994278 | - - flLYAQPgEΞs iBJIΪSEfflMjilAYl^RBBViR FKROREN^I YSMJFD
Figure imgf000038_0001
460 470
NOV2a IWiιJn.aMeia-AWiJιi*il^Al33GilGASi5BlL
NOV2b Ji4' Jn.ab.ia-A Jd*JAJAl3gGllGASlSniL
Nov2c ΞH3ESSHS3ΪSΞSAΞSGΪZGASE32
NOV2d J^J».afet.a-fc iJιlιfcl^_ l3gGilGASlSπiL gi 1 3994273 I iii' in.afe..a-fc 'iJdΛtWJ tSgGllGAsEBlL gi 112835488 I ..J'i iJAWAWJrifelAEJlGigg fflKPPiiiffl- gi 113385666 I k4'i.Wd!iMslfc dtVftJftgπ.53ΑllκPPl5π-
Figure imgf000038_0002
Tables 2K list the domain description from DOMAIN analysis results against NOV2. This indicates that the NOV2 sequence has properties similar to those of other proteins known to contain this domain.
Table 2K. Domain Analysis of NOV2 gnllSmartlsmartOOllO, C1Q, Complement component Clq domain.; Globular domain found in many collagens and eponymously in complement Clq. When part of full length proteins these domains form a 'bouquet' due to the multimerization of heterotrimers. The Clq fold is similar to that of tumour necrosis factor.
CD-Length = 132 residues, 84.1% aligned Score = 86.7 bits (213), Expect = le-18
Query : 91 MAVTFD?^ΪVNIGGDFDAAAGVFRCR PGAYFFSFT GKLPRKTLSV LMKNRDEVQAMI 150
V FDKV N G +D + G F C +PG Y+FS+ + + + + V LMKN +V Sbjct: 20 QPVRFDKVLYNQQGHYDPSTGKFTCPVPGVYYFSYHI-ESKGRHVKVSLMNGIQVMRE- 77
Query: 151 YDDGASRRREMQSQSVMLALRRGDAV L SHDHDGYGAYSNHGKYITFSGFLVY 204
D+ ++ S + R+GD V L D G Y+ TFSGFL++ Sbjct: 78 CDEYQKGLYQVASGGALLQLRQGDQV - ELDDKKNG YAGEEVDSTFSGFL F 130
Clq is the first subcomponent of the Cl complex of the classical pathway of complement activation. Several functions have been assigned to Clq, which include antibody- dependent and independent immune functions, and are considered to be mediated by Clq receptors present on the effector cell surface. There remains some uncertainty about the identities of the receptors that mediate Clq functions. Some of the previously described Clq receptor molecules, such as gClqR and cClqR, now appear to have less of a role in Clq functions than in functions unrelated to Clq. The problem of identifying receptor proteins with complementary binding sites for Clq has been compounded by the highly charged nature of the different domains in Clq. Although newer candidate receptors like ClqR(p) and CRl have emerged, full analysis of the Clq-Clq receptor interactions is still at an early stage. In view of the diverse functions that Clq is considered to perform, it has been speculated that several Clq-binding proteins may act in concert, as a Clq receptor complex, to bring about Clq mediated functions. Some major advances have been made in last few years. Experiments with gene targeted homozygous Clq-deficient mice have suggested a role for Clq in modulation of the humoral immune response, and also in protection against development of autoimmunity. The recently described crystal structure of ACRP-30, has revealed a new Clq/TNF superfamily of proteins. Although the members of this superfamily may have diverse functions, there may be a common theme in their phylogeny and modular organisation of their distinctive globular domains.
The novel polypeptide described in this application is homologous to adipocyte complement related protein 3 (ACRP3). The ACRP3 protein is made exclusively in adipocytes and its mRNA is induced over 100-fold during adipocyte differentiation. ACRP3 is structurally similar to complement factor Clq and to a hibernation-specific protein isolated from the plasma of Siberian chipmunks; it forms large homo-oligomers that undergo a series of post-translational modifications. A similar protein has a cluster of aromatic residues near the C terminus having high local similarity with collagens X and VIII and complement factor Clq. Clq is a subunit of the Cl enzyme complex that activates the serum complement system. Clq comprises 6 A, 6 B and 6 C chains. These share the same topology, each possessing a small, globular N-terminal domain, a collagen-like Gly/Pro-rich central region, and a conserved C-terminal region, the Clq domain. The Clq protein is produced by collagen- producing cells and shows sequence and structural similarity to collagens VIII and X, (see, Scherer PE, et al., J Biol Chem 1995 Nov 10;270(45):26746-9 and Maeda K, et al., Biochem Biophys Res Commun 1996 Apr 16;221(2):286-9), incorporated herein by reference.
The present invention includes chimeric or fusion proteins of the complement-Clq tumor necrosis factor-like protein, in which the complement-Clq tumor necrosis factor-like protein of the present invention is joined to a second polypeptide or protein that is not substantially homologous to the present novel protein. The second polypeptide can be fused to either the amino-terminus or carboxyl-terminus of the present CG55724-01, CG55724-03, CG55724-04, or CG55724-06 polypeptide. In certain embodiments a third nonhomologous polypeptide or protein may also be fused to the novel complement-Clq tumor necrosis factorlike protein such that the second nonhomologous polypeptide or protein is joined at the amino terminus, and the third nonhomologous polypeptide or protein is joined at the carboxyl terminus, of the CG55724-01, CG55724-03, CG55724-04, or CG55724-06 polypeptide. Examples of nonhomologous sequences that may be incorporated as either a second or third polypeptide or protein include glutathione S-transferase, a heterologous signal sequence fused at the amino terminus of the complement-Clq tumor necrosis factor-like protein, an immunoglobulin sequence or domain, a serum protein or domain thereof (such as a serum albumin), an antigenic epitope, and a specificity motif such as (His)6. The invention further includes nucleic acids encoding any of the chimeric or fusion proteins described above.
The disclosed NOV2 nucleic acids of the invention encoding a complement-related Clq Tumor Necrosis Related Protein-like protein includes the nucleic acidswhose sequence is provided in Table 2A, 2C, 2E and 2G or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 2A, 2C, 2E and 2G while still encoding a protein that maintains its complement-related Clq Tumor Necrosis Related Protein-like protein activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 40% (NOV2a), 6% (NOV2b), 6% (NOV2c) and 9% (NOV2d) of the bases may be so changed.
The disclosed NON2 protein of the invention includes the complement-related Clq Tumor Necrosis Related Protein-like protein whose sequence is provided in Table 2B, 2D, 2F and 2G. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 2B, 2D, 2F and 2G while still encoding a protein that maintains its the complement-related Clq Tumor Necrosis Related Protein-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 66% (NOV2a), 2% (NON2b, ΝON2c), and 9% (ΝOV2d) of the residues may be so changed.
The NOV2 nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in cancers, adrenoleukodystrophy , Alzheimer's disease, autoimmune disease, allergies, addiction, anxiety, ataxia-telangiectasia, asthma, ARDS, atherosclerosis, behavioral disorders, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, allergy, cerebral palsy, congenital adrenal hyperplasia, cirrhosis, cardiomyopathy, congenital heart defects, diabetes, diverticular disease, epilepsy, emphysema, endometriosis, endocrine dysfunctions, graft versus host disease, glomerulonephritis, graft versus host disease (GVHD), growth and reproductive disorders, hemophilia, hypercoagulation, hypercalceimia, Huntington's disease, hypertension, hypogonadism, fertility, idiopathic thrombocytopenic purpura, immunodeficiencies, interstitial nephritis, IgA nephropathy, lymphaedema, inflammatory bowel disease, Lesch-Nyhan syndrome, leukodystrophies, multiple sclerosis, muscular dystrophy, myasthenia gravis, neurodegeneration, neuroprotection,obesity, Parkinson's disease, pain, polycystic kidney disease, pulmonary stenosis, pancreatitis, renal artery stenosis, renal tubular acidosis, stroke, systemic lupus erythematosus, scleroderma, subaortic stenosis, transplantation, tuberous sclerosis, Von Hippel-Lindau (VHL) syndrome, ventricular septal defect (VSD), valve diseases, Von Hippel-Lindau (VHL) syndrome, ulcers, and other diseases, pathologies and disorders. The NOV2 nucleic acid encoding the complement-related Clq Tumor Necrosis Related Protein-like protein, and the protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOV2 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. The disclosed NOVa, NOV2b, NOV2c, and NOV2d proteins have multiple hydrophilic regions, each of which can be used as an immunogen.
In one embodiment, a contemplated NOV2a epitope is from about amino acids 25 to 100. h another embodiment, a contemplated NOV2a epitope is from about amino acids 110 to 275. h other specific embodiments, contemplated NOVl epitopes are from about amino acids 280 to 325, 350 to 425, 450 to 625, 650 to 690, 700 to 825, and 850 to 965. hi one embodiment, a contemplated NOV2b epitope is from about amino acids 20 to 50. In another embodiment, a contemplated NOV2b epitope is from about amino acids 55 to 65. h other specific embodiments, contemplated NO V2b epitopes are from about amino acids 90 to 145, 195 to 235, and 240 to 260.
In one embodiment, a contemplated NOV2c epitope is from about amino acids 20 to 50. hi another embodiment, a contemplated NOV2c epitope is from about amino acids 55 to 65. h other specific embodiments, contemplated NOV2c epitopes are from about amino acids 90 to 145, 195 to 235, and 240 to 260.
In one embodiment, a contemplated NOV2d epitope is from about amino acids 18 to 40. hi another embodiment, a contemplated NOV2d epitope is from about amino acids 42 to 47. hi other specific embodiments, contemplated NOV2d epitopes are from about amino acids 60 to 80, 85 to 105, and 106 to 110.
NOV3
A disclosed NO V3 nucleic acid of 3073 nucleotides is set forth as SEQ ID NO: 11 (also referred to as CG50345-01) encoding a beta adrenergic receptor kinase-like protein is shown in Table 3A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 108-110 and ending with a TGA codon at nucleotides 2112-2114.
Table 3A.
NOV3 Polynucleotide
SEQ ID NO:ll
GGGTACCGAGCTCGAATTCCGGCTCGGCCTCGGGCGCGGCCGAGCGCCGCGCGAGCAGGA 60
GCGGCGGCGGCGGCGGCGGCGGCGGGAGGAGGCAGCGCCGGCCCAAGATGGCGGACCTGG 120
AGGCGGTGCTGGCCGACGTGAGCTACCTGATGGCCATGGAGAAGAGCAAGGCCACGCCGG 180
CCGCGCGCGCCAGCAAGAAGATACTGCTGCCCGAGCCCAGCATCCGCAGTGTCATGCAGA 240
AGTACCTGGAGGACCGGGGCGAGGTGACCTTTGAGAAGATCTTTTCCCAGAAGCTGGGGT 300
ACCTGCTCTTCCGAGACTTCTGCCTGAACCACCTGGAGGAGGCCAGGCCCTTGGTGGAAT 360
TCTATGAGGAGATCAAGAAGTACGAGAAGCTGGAGACGGAGGAGGAGCGTGTGGCCCGCA 420
GCCGGGAGATCTTCGACTCATACATCATGAAGGAGCTGCTGGCCTGCTCGCATCCCTTCT 480
CGAAGAGTGCCACTGAGCATGTCCAAGGCCACCTGGGGAAGAAGCAGGTGCCTCCGGATC 5 0
TCTTCCAGCCATACATCGAAGAGATTTGTCAAAACCTCCGAGGGGACGTGTTCCAGAAAT 600
TCATTGAGAGCGATAAGTTCACACGGTTTTGCCAGTGGAAGAATGTGGAGCTCAACATCC 660
ACCTGACCATGAATGACTTCAGCGTGCATCGCATCATTGGGCGCGGGGGCTTTGGCGAGG 720
TCTATGGGTGCCGGAAGCGTGACACAGGCAAGATGTACGCCATGAAGTGCCTGGACAAAA 780
AGCGCATCAAGATGAAGCAGGGGGAGACCCTGGCCCTGAACGAGCGCATCATGCTCTCGC 840
TCGTCAGCACTGGGGACTGCCCATTCATTGTCTGCATGTCATACGCGTTCCACACGCCAG 900
ACAAGCTCAGCTTCATCCTGGACCTCATGAACGGTGGGGACCTGCACTACCACCTCTCCC 960
AGCACGGGGTCTTCTCAGAGGCTGACATGCGCTTCTATGCGGCCGAGATCATCCTGGGCC 1020
TGGAGCACATGCACAACCGCTTCGTGGTCTACCGGGACCTGAAGCCAGCCAACATCCTTC 1080
TGGACGAGCATGGCCACGTGCGGATCTCGGACCTGGGCCTGGCCTGTGACTTCTCCAAGA 1140
AGAAGCCCCATGCCAGCGTGGGCACCCACGGGTACATGGCTCCGGAGGTCCTGCAGAAGG 1200
GCGTGGCCTACGACAGCAGTGCCGACTGGTTCTCTCTGGGGTGCATGCTCTTCAAGTTGC 1260
TGCGGGGGCACAGCCCCTTCCGGCAGCACAAGACCAAAGACAAGCATGAGATCGACCGCA 1320
TGACGCTGACGATGGCCGTGGAGCTGCCCGACTCCTTCTCCCCTGAACTACACTCCCTGC 1380
TGGAGGGGTTGCTGCAGAGGGATGTCAACCGGAGATTGGGCTGCCTGGGCCGAGGGGCTC 1440
AGGAGGTGAAAGAGAGCCCCTTTTTCCGCTCCCTGGACTGGCAGATGGTCTTCTTGCAGA 1500
GGTACCCTCCCCCGCTGATCCCCCCACGAGGGGAGGTGAACGCGGCCGACGCCTTCGACA 1560
TTGGCTCCTTCGATGAGGAGGACACAAAAGGAATCAAGCAGGAGGTGGCAGAGACTGTCT 1620
TCGACACCATCAACGCTGAGACAGACCGGCTGGAGGCTCGCAAGAAAGCCAAGAACAAGC 1680
AGCTGGGCCATGAGGAAGACTACGCCCTGGGCAAGGACTGCATCATGCATGGCTACATGT 1740
CCAAGATGGGCAACCCCTTTCTGACCCAGTGGCAGCGGCGGTACTTCTACCTGTTCCCCA 1800
ACCGCCTCGAGTGGCGGGGCGAGGGCGAGGCCCCGCAGAGCCTGCTGACCATGGAGGAGA 1860
TCCAGTCGGTGGAGGAGACGCAGATCAAGGAGCGCAAGTGCCTGCTCCTCAAGATCCGCG 1920
GTGGGAAACAGTTCATTTTGCAGTGCGATAGCGACCCTGAGCTGGTGCAGTGGAAGAAGG 1980
AGCTGCGCGACGCCTACCGCGAGGCCCAGCAGCTGGTGCAGCGGGTGCCCAAGATGAAGA 2040
ACAAGCCGCGCTCGCCCGTGGTGGAGCTGAGCAAGGTGCCGCTGGTCCAGCGCGGCAGTG 2100
CCAACGGCCTCTGACCCGCCCACCCGCCTTTTATAAACCTCTAATTTATTTTGTCGAATT 2160
TTTATTATTTGTTTTCCCGCCAAGCGAAAAGGTTTTATTTTGTAATTATTGTGATTTCCC 2220
GTGGCCCCAGCCTGGCCCAGCTCCCCCGGGAGGCCCCGCTTGCCTCGGCTCCTGCTGCAC 2280
CAACCCAGCCGCTGCCCGGCGCCCTCTGTCCTGACTTCAGGGGCTGCCCGCTCCCAGTGT 2340
CTTCCTGTGGGGGAAGAGCACAGCCCTCCCGCCCCTTCCCCGAGGGATGATGCCACACCA 2400
AGCTGTGCCACCCTGGGCTCTGTGGGCTGCACTTGTGCCATGGGACTGTGGGTGGCCCAT 2460
CCCCCCTCACCAGGGGCAGGCACAGCACAGGGATCCGACTTGAATTTTCCCACTGCACCC 2520
CCTCCTGCTGCAGAGGGGCAGGCCCTGCACTGTCCTGCTCCACAGTGTTGGCGAGAGGAG 2580
GGGCCCGTTGTCTCCCTGGCCCTCAAGGCTCCCACAGTGACTCGGGCTCCTGTGCCCTTA 2640
TTCAGGAAAAGCCTCTGTGTCACTGGCTGCCTCCACTCCCACTTCCCTGACACTGCGGGG 2700
CTTGGCTGAGAGAGTGGCATTGGCAGCAGGTGCTGCTACCCTCCCTGCTGTCCCCTCTTG 2760
CCCCAACCCCCAGCACCCGGGCTCAGGGACCACAGCAAGGCACCTGCAGGTTGGGCCATA 2820
CTGGCCTCGCCTGGCCTGAGGTCTCGCTGATGCTGGGCTGGGTGCGACCCCATCTGCCCA 2880
GGACGGGGCCGGCCAGGTGGGCGGGCAGCACAGCAAGGAGGCTGGCTGGGGCCTATCAGT 2940
GTGCCCCCCATCCTGGCCCATCAGTGTACCCCCGCCCAGACTGGCCAGCCCCACAGCCCA 3000
CGTCCTGTCAGTGCCGCCGCCTCGCCCACCGCATGCCCCCTGTGCCAGTGCTCTGCCTGT 3060 GTGTGTGCACTCT The disclosed NOV3 nucleic acid sequence maps to chromosome 1 lql3 and has 1638 of 1666 bases (98%) identical to a gb:GENBANK-ID:HSBARK|acc:X61157.1 mRNA from Homo sapiens (H. sapiens mRNA for beta-adrenergic receptor kinase).
A disclosed NOV3 protein (SEQ ID NO: 12) encoded by SEQ ID NO:ll has 668 amino acid residues, and is presented using the one-letter code in Table 3B. Signal P, Psort and/or Hydropathy results predict that NOV3 does have a signal peptide, and is likely to be localized to the nucleus with a certainty of 0.8800. In other embodiments NOV3 is also likely to be localized to perioxisomal microbodies with a certainty of 0.1582, mitochondrial matrix space with a certainty of 0.1000, to the lysosomal lumen with a certainty of 0.1000.
Table 3B
NOV3 Polypeptide
SEQ ID NO:12
MADLEAVLADVSY MA EKSKATPAARASKKIL PEPSIRSVMQKY EDRGEVTFEKIFS 60
QKLGYLLFRDFCLNHLEEARPLVEFYEEIKKYEKLETEEERVARSREIFDSYIMKELLAC 120
SHPFSKSATEHVQGHLGKKQVPPD FQPYIEEICQNLRGDVFQKFIESDKFTRFCQWKNV 180
ELNIHLTMNDFSVHRIIGRGGFGEVYGCRKRDTGK YAMKCLDKKRIKMKQGETLALNER 240
IMLSLVSTGDCPFIVCMSYAFHTPDKDSFILDLMNGGDLHYHLSQHGVFSEADMRFYAAE 300
IILG EHMHNRFWYRD KPAMILLDEHGHVRISDLG ACDFSKKKPHASVGTHGYMAPE 360
VLQKGVAYDSSAD FS GCMLFKLLRGHSPFRQHKT DKHEIDRMTLTMAVELPDSFSPE 420
LHSLLEGL QRDVNRRLGCLGRGAQEVKESPFFRSLD QMVFLQRYPPPLIPPRGEVNAA 480
DAFDIGSFDEEDTKGIKQEVAETVFDTINAETDRLEARKKA NKQLGHEEDYALGKDCI 5 0
HGYMSKMGNPFLTQ QRRYFYLFPNRLE RGEGEAPQSLLTMEEIQSVEETQIKERKC L 600
LKIRGGKQFILQCDSDPE VQ KKELRDAYREAQQLVQRVPKMKNKPRSPWELSKVP V 660 QRGSANGL
The disclosed NOV3 amino acid has 359 of 642 amino acid residues (55%) identical to, and 497 of 497 amino acid residues (100%) similar to 497 of the 689 amino acid residue ptnr:SWISSNEW ACC:P25098 protein from Homo sapiens (Human) beta-adrenergic receptor kinase 1 (beta-ARKl, G-Protein Coupled Receptor Kinase 2).
The NOV3 sequence is expressed in at least the following tissues: brain-the Adrenal Gland/Suprarenal gland, Amygdala, Aorta, Bone, Bone Marrow, Brain, Cerebellum, Cervix, Chorionic Villus,Cochlea, Colon, Dermis, Epidermis, Foreskin, Hair Follicles, Heart, Hippocampus, Hypothalamus, Kidney, Liver, Lung, Lymph node, Lymphoid tissue, Mammary gland/Breast, Muscle, Myometrium, Ovary, Pancreas, Parotid Salivary glands, Pituitary Gland, Placenta, Prostate, Proximal Convoluted Tubule, Small Intestine, Spinal Chord, Retina, Spleen, Stomach, Substantia Nigra, Testis, Thymus, Thyroid, Tonsils, Umbilical Vein, Urinary Bladder, Uterus.
NOV3 also has homology to the amino acid sequences shown in the BLASTP data listed in Table 3C.
Figure imgf000045_0001
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 3D.
Table 3D. ClustalW Analysis of NO 3
1) NOV3 (SEQ ID NO.ll)
2) gi| 6978467 I (SEQ ID N0.45)
3) gi|4206092| (SEQ ID NO:46)
4) gi 11141511 (SEQ ID NO:47) , 5) gi 1114154 I (SEQ ID NO:48)
6) gi| 5139484 I (SEQ ID N0.49)
10 20 30 40 50 NOV3 gi I 6978467 I MADLEAVLADVSY AMEKSKATPAARASKK WΛΛMMUMimuΛ gi I 4206092 I ΪADLEAVLADVSYLMAMEKSKATPAARASKKI fLPEPSIRSVMi iBWMIaPra
Figure imgf000046_0001
gi1114154 I lAD EAV ADVSY MAMEKSI ATPAARASKKIfflLPEPSIRSVMOKYliE i gi|5139484| ii^.^^ M.tf ^ι^^u^^j^;ι^aRι^Λ^^4 ^ι.>i^iΑsig
60 70 80 90 100
NOV3 G[|vi33EiSi sSS:[S2SBRi3E!_SH:i;,E_SRi3LEKl lκtffatøfeτl53lB gi 169784671 ιfeϊ&iaήBt3TOιBgl1TOκig3ϊWEiGi5 >vrao[5] gi I 4206092 i GJ3 iBEi2BS ?S^^KEi_ESSHKIE!EKSLi
Figure imgf000046_0002
110 120 130 140 150
NOV3 ISIV'idg.raBi-^syii-l&Wartl.^^
Figure imgf000046_0003
gi I 5139484 I IMc!3^oπ^^^J .i Jι*M^..J^.fe^^i tf»fe.ii1lsiaa^THτpi^
160 170 180 190 200
NOV3 ^^^^m^^TJΛiΛsUiAώUM^άiAktikkMMJs^^i^SS
210 220 230 240 250
NOV3 MawataiiftwaaRwroaaBiKWB M»wa»ιifeHWiaaHBiiBt»wiaιι.»)» gi|6978467| MϋMstfgBΪM gi|4206092| GFGEVYGCRKADTGKMYAMKCLDKKRIKMKQGETLALNERIMLSLVSTGD
Figure imgf000046_0005
gi1114154 I ΪFGEVYGCRKADTGKMYAMKCLD KRIK KOGETLALNERIMLSLVSTGI gi] 5139484 I .VYGCRKADTGKMYAMKCLDKKRIKMKOGETLALNERIM SLVSTG
260 270 280 290 300
Figure imgf000046_0006
Figure imgf000046_0007
Figure imgf000046_0008
310 320 330 340 350
■unco ι***;eJΛstJIΛIJι CL»Vrt;Ji)ΛMΛ Λ*ι)SY[eli»*i;*«:)i_ι*fe.*Λ«iι)^^I«M!.^Λ aFiaimawaitaiEWϊTii.
11 G EHMHNRFWYRDLKPANI LDEHGHVRI SDLG ACDFSKKKPHAS
ILGLEHMHNRFVVYRDLKPANILLDEHGHaRISDLGLACDFSKKKPHA
360 370 380 390 400
Figure imgf000047_0001
gi|4206092| WtMUmΛMiOmsAIMΛMΛUΛΛMiMiUMiMύΛ^UUm gi 11141511 MDΞSADWFSLGCMLFKLLRGHSPFROHKTKDKI
gi]5139484]
410 420 430 440 450
Figure imgf000047_0002
gi I 69784671 yafc _Adaιt-lhιigιaBBθW3g-B-l3BI3aHlι^ππ[5M
Figure imgf000047_0003
gi 11141511 l>HΦl&lιdlιι. tiatJ-dιfel3gl33^^
Figure imgf000047_0004
460 470 480 490 500
NOV3 pl^Ksl]fiMyιBigl-BRfaaaaιι.ιι Ma«^^^ —
Figure imgf000047_0005
510 520 530 540 550
No 3 iiWAHa .immmMMmmMMMU
Figure imgf000047_0006
gi I H41541 c&ddiif(l.ElvEfet-i.iW
Figure imgf000047_0007
560 570 580 590 600
NOV3 gi|6978467]
Figure imgf000047_0008
gi] 114154]
Figure imgf000048_0001
610 620 630 640 650
NOV3 gi|6978467| IAJι!ιtόH_^*^^ιl«>J^tkl 44Λl vJtte>!t_^ft!aEM»lJdF l^L!55πτcτ gi|4206092|
Figure imgf000048_0002
gi|ll4154| gi| 5139484 I
660 670 680
NOV3 tosSgoffl ^c iM3BcMκiSt^J;ld ^5HI3JM31[vo^Gfe!^iHI
Figure imgf000048_0003
gi|4206092| !ϊBJSξ^{2Q§{^κB^p^3ΞSIτQΞ ^B
Figure imgf000048_0004
gi|5139484] SF 5_ s.;JGτB5flpigpsBcHig-ΗsB?π
Table 3E lists the domain description from DOMAIN analysis results against NOV3. This indicates that the NOV3 sequence has properties similar to those of other proteins known to contain this domain.
Table 3E Domain Analysis of NOV3
gnl|Smart|smart00220, S_TKc, Serine/Threonine protein kinases, catalytic domain; Phosphotransferases. Serine or threonine-specific kinase subfamily.
CD-Length = 256 residues, 100.0% aligned Score = 237 bits (605), Expect = le-63
Query : 191 FSVHRIIGRGGFGEVYGCRKRDTGKMYAMKC DKKRIKMKQGETLA NERIMLSLVSTGD 250
+ + ++G+G FG+VY R + TGK+ A+K + K+++K K+ E L E +L + D
Sbjct: 1 YELLEV GKGAFGKVY ARDKKTGKLVAIKVIKKEKLKKK RER-ILREIKI KK D
56
Query : 251 CPFIVCMSYAFHTPDKLSFILDLMNGGDLHYHLSQHGVFSEADMRFYAAEIILGLEHMHN 310
P IV + F DKL +++ GGDL L + G SE + RFYA +1+ LE++H+ Sbjct: 57 HPNIVKLYDVFEDDDKLY VMΞYCEGGDLFD KKRGR SEDEARFYARQILSALEYLHS 116
Quer : 311 RFWYRD KPANI LDEHGHVRISDLGLACDFSKKKPHAS- -VGTHGYMAPEVLQKGVAY 368
+ +++RDLKP NIL D GHV+++D GLA + VGT YMAPEVL G Y
Sbjct : 117 QGIIHRDLKPENI LDSDGHVKLADFG AKQLDSGGTLLTTFVGTPEYMAPEVL-LGKGY 175
Query: 369 DSSADWFSLGCMLFKLLRGHSPFRQHKTKDK-HEIDRMT TMAVELPDSFSPELHS LΞG 427
+ D +SLG +L++LL G PF + SPE L++
Sbj Ct : 176 GKAVDI SLGVILYELLTGKPPFPGDDQ ALF KIGKPPPPFPPPE KISPEAKD IKK 235
Query: 428 L QRDVNRR GC GRGAQEVKESPFF 453
L +D +R A+E E PFF Sbjct: 236 L VKDPEKRL TAEEALEHPFF 256
Beta-adrenergic receptor kinase (beta-ARKl) phosphorylates the beta-2-adrenergic receptor and appears to mediate agonist-specific desensitization observed at high agonist concentrations. Beta-ARKl is an ubiquitous cytosolic enzyme that specifically phosphorylates the activated form of the beta-adrenergic and related G-protein-coupled receptors. The beta-ARKl gene spans approximately 23 kb and is composed of 21 exons. Beta-AR kinase (beta-ARKl) is known to be elevated in failing human heart tissue and its activity resulting in rapid desensitization via the abnormal coupling or uncoupling of beta- adrenergic receptor to G protein, receptor down-regulation, internalization and degradation, may account for some of the abnormalities of contractile function in the heart disease (see, Post, S. R., Hammond, H.K., Insel, P.A.,1999, Annu. Rev. Pharmacol. Vol. 39: 343-360) incorporated by reference.
Beta-adrenergic receptor kinase (beta-ARKl) phosphorylates the beta-2-adrenergic receptor and appears to mediate agonist-specific desensitization observed at high agonist concentrations. Beta-ARKl is an ubiquitous cytosolic enzyme that specifically phosphorylates the activated form of the beta-adrenergic and related G-protein-coupled receptors. The beta- ARKl gene spans approximately 23 kb and is composed of 21 exons. Beta-AR kinase (beta- ARKl) is known to be elevated in failing human heart tissue and its activity resulting in rapid desensitization via the abnormal coupling or uncoupling of beta-adrenergic receptor to G protein, receptor down-regulation, internalization and degradation, may account for some of the abnormalities of contractile function in the heart disease (see, Post, S. R., Hammond, H.K., hisel, P.A.,1999, Annu. Rev. Pharmacol. Vol. 39: 343-360, incoφorated. herein by reference)
The protein similarity information, expression pattern, and map location for the Beta- adrenergic receptor kinase-like protein and nucleic acid disclosed herein suggest that this Beta-adrenergic receptor kinase may have important structural and/or physiological functions characteristic of the Serine-threonine protein kinase family. Therefore, the nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo (vi) biological defense weapon.
The nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications implicated in various diseases and disorders described below and/or other pathologies. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from: cardiac diseases, myocardial contractility in failing heart and other diseases, disorders and conditions of the like. The disclosed NOV3 nucleic acid of the invention encoding a beta adrenergic receptor kinase -like protein includes the nucleic acid whose sequence is provided in Table 3 A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 3A while still encoding a protein that maintains beta adrenergic receptor kinase-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 2 percent of the bases may be so changed.
The disclosed NOV3 protein of the invention includes the beta adrenergic receptor kinase-like protein whose sequence is provided in Table 3B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 3B while still encoding a protein that maintains beta adrenergic receptor kinase-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 1 percent of the residues may be so changed.
The protein similarity information, expression pattern, and map location for the beta adrenergic receptor kinase-like protein and nucleic acid (NOV3) disclosed herein suggest that NOV3 may have important structural and/or physiological functions characteristic of the beta adrenergic receptor kinase-like family. Therefore, the NOV3 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo.
The NOV3 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications implicated in various diseases and disorders described below. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from cancer, inflammation, retinal disorders, neurological disorders, neuropsychiatric disorders, obesity, diabetes, bleeding disorders and/or other pathologies. The NOV3 nucleic acid, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOV3 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. The disclosed NOV3 polypeptide has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV3 epitope is from about amino acids 20 to 70. In another embodiment, a contemplated NOV3 epitope is from about amino acids 95 to 115. hi other specific embodiments, contemplated NOV3 epitopes are from about amino acids 120 to 190, 280 to 300, 305 to 375, 395 to 420, and 415 to 660.
NOV4
A disclosed NOV4 nucleic acid of 8354 nucleotides is set forth as SEQ ID NO:13 (designated CuraGen Ace. No. CG50301-01) encoding a TEN-M4-like protein is shown in Table 4A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 35-37 and ending with a TAG codon at nucleotides 8342-8344. Putative untranslated regions are indicated by underline.
Table 4A.
NOV4 Polynucleotide
SEQ ID NO:13
GTTTGTGGATGTGGAGGAGCGCGGGCCGGAGGCCATGGACGTGAAGGAGAGGAAGCCTTA 60
CCGCTCGCTGACCCGGCGCCGCGACGCCGAGCGCCGCTACACCAGCTCGTCCGCGGACAG 120
CGAGGAGGGCAAAGCCCCGCAGAAATCGTACAGCTCCAGCGAGACCCTGAAGGCCTACGA 180
CCAGGACGCCCGCCTAGCCTATGGCAGCCGCGTCAAGGACATTGTGCCGCAGGAGGCCGA 240
GGAATTCTGCCGCACAGGTGCCAACTTCACCCTGCGGGAGCTGGGGCTGGAAGAAGTAAC 300
GCCCCCTCACGGGACCCTGTACCGGACAGACATTGGCCTGCCCCAATGCGGCTACTCCAT 360
GGGGGCTGGCTCTGATGCCGACATGGAGGCTGACACGGTGCTGTCCCCTGAGCACCCCGT 20
GCGTCTGTGGGGCCGGAGCACACGGTCAGGGCGCAGCTCCTGCCTGTCCAGCCGGGCCAA 480
TTCCAATCTCACACTCACCGACACCGAGCATGAAAACACTGAGACTGATCATCCGGGCGG 540
CCTGCAGAACCACGCGCGGCTCCGGACGCCGCCGCCGCCGCTCTCGCACGCCCACACCCC 600
CAACCAGCACCACGCGGCCTCCATTAACTCCCTGAACCGGGGCAACTTCACGCCGAGGAG 660
CAACCCCAGCCCGGCCCCCACGGACCACTCGCTCTCCGGAGAGCCCCCTGCCGGCGGCGC 720
CCAGGAGCCTGCCCACGCCCAGGAGAACTGGCTGCTCAACAGCAACATCCCCCTGGAGAC 780
CAGGAACCTAGGCAAGCAGCCATTCCTAGGGACATTGCAGGACAACCTCATTGAGATGGA 840
CATTCTCGGCGCCTCCCGCCATGATGGGGCTTACAGTGACGGGCACTTCCTCTTCAAGCC 900
TGGAGGCACCTCCCCGCTCTTCTGCACCACATCACCAGGGTACCCACTGACGTCCAGCAC 960
AGTGTACTCTCCTCCGCCCCGACCCCTGCCCCGCAGCACCTTCGCCCGGCCGGCCTTTAA 1020
CCTCAAGAAGCCCTCCAAGTACTGTAACTGGAAGTGCGCAGCCCTGAGCGCCATCGTCAT 1080
CTCAGCCACTCTGGTCATCCTGCTGGCATACTTTGTGGCCATGCACCTGTTTGGCCTAAA 1140
CTGGCACCTGCAGCCGATGGAGGGGCAGATGTATGAGATCACGGAGGACACAGCCAGCAG 1200
TTGGCCTGTGCCAACCGACGTCTCCCTATACCCCTCAGGGGGCACTGGCTTAGAGACCCC 1260
TGACAGGAAAGGCAAAGGAACCACAGAAGGAAAGCCCAGTAGTTTCTTTCCAGAGGACAG 1320
TTTCATAGATTCTGGAGAAATTGATGTGGGAAGGCGAGCCTCCCAGAAGATTCCTCCTGG 1380
CACTTTCTGGAGATCTCAAGTGTTCATAGACCATCCTGTGCATCTGAAATTCAATGTGTC 1440
TCTGGGAAAGGCAGCCCTGGTTGGCATTTATGGCAGAAAAGGCCTCCCTCCTTCACATAC 1500
ACAGTTTGACTTTGTGGAGCTGCTGGATGGCAGGAGGCTCCTAACCCAGGAGGCGCGGAG 1560
CCTAGAGGGGACCCCGCGCCAGTCTCGGGGAACTGTGCCCCCCTCCAGCCATGAGACAGG 1620
CTTCATCCAGTATTTGGATTCAGGAATCTGGCACTTGGCTTTTTACAATGACGGAAAGGA 1680
GTCAGAAGTGGTTTCCTTTCTCACCACTGCCATTGAGTCGGTGGATAACTGCCCCAGCAA 1740
CTGCTATGGCAATGGTGACTGCATCTCTGGGACCTGCCACTGCTTCCTGGGTTTCCTGGG 1800
CCCCGACTGTGGCAGAGCCTCCTGCCCCGTGCTCTGTAGCGGAAATGGCCAATACATGAA 1860
AGGCAGATGCTTGTGCCACAGTGGCTGGAAAGGCGCTGAGTGCGATGTGCCCACCAACCA 1920
GTGTATCGATGTGGCCTGCAGCAACCATGGCACCTGCATCACGGGCACCTGCATCTGCAA 1980
CCCTGGCTACAAGGGCGAGAGCTGTGAGGAAGTGGACTGCATGGACCCCACATGTTCAGG 2040
CCGGGGTGTCTGCGTGAGAGGCGAATGCCATTGCTTTGTGGGATGGGGAGGCACCAACTG 2100
CGAGACCCCCAGGGCCACATGCTTAGACCAGTGTTCAGGCCACGGAACCTTCCTCCCGGA 2160
CACCGGGCTTTGCAGCTGTGACCCAAGCTGGACTGGACACGACTGTTCTATCGAGATCTG 2220 TGCTGCCGACTGTGGTGGCCATGGCGTGTGCGTAGGGGGCACCTGCCGCTGCGAGGATGG 280
CTGGATGGGGGCAGCCTGCGACCAGCGGGCCTGCCACCCGCGCTGTGCCGAGCATGGGAC 2340
CTGCCGCGACGGCAAGTGCGAGTGCAGCCCTGGCTGGAATGGCGAACACTGCACCATCGC 2 00
TCACTATCTGGATAGGGTAGTTAAAGAGGGTTGCCCTGGGTTGTGCAATGGGAACGGCAG 2 60
ATGTACCTTAGACCTGAATGGTTGGCACTGCGTCTGCCAGCTGGGCTGGAGAGGAGCTGG 5 0
CTGTGACACTTCCATGGAGACTGCCTGCGGTGACAGCAAAGACAATGATGGAGATGGCCT 580
GGTGGACTGCATGGACCCTGACTGCTGCCTCCAGCCCCTGTGCCATATCAACCCGCTGTG 26 0
CCTTGGCTCCCCTAACCCTCTGGACATCATCCAGGAGACACAGGTCCCTGTGTCACAGCA 2700
GAACCTACACTCCTTCTATGACCGCATCAAGTTCCTCGTGGGCAGGGACAGCACGCACAT 2760
AATCCCCGGGGAGAACCCCTTTGATGGAGGGCATGCTTGTGTTATTCGTGGCCAAGTGAT 2820
GACATCAGATGGAACCCCCCTGGTTGGTGTGAACATCAGTTTTGTCAATAACCCTCTCTT 880
TGGATATACAATCAGCAGGCAAGATGGCAGCTTTGACTTGGTGACAAATGGCGGCATCTC 2940
CATCATCCTGCGGTTCGAGCGGGCACCTTTCATCACACAGGAGCACACCCTGTGGCTGCC 3000
ATGGGATCGCTTCTTTGTCATGGAAACCATCATCATGAGACATGAGGAGAATGAGATTCC 3060
CAGCTGTGACCTGAGCAATTTTGCCCGCCCCAACCCAGTCGTCTCTCCATCCCCACTGAC 3120
GTCCTTCGCCAGCTCCTGTGCAGAGAAAGGCCCCATTGTGCCGGAAATTCAGGCTTTGCA 3180
GGAGGAAATCTCTATCTCTGGCTGCAAGATGAGGCTGAGCTACCTGAGCAGCCGGACCCC 3240
TGGCTACAAATCTGTCCTGAGGATCAGCCTCACCCACCCGACCATCCCCTTCAACCTCAT 3300
GAAGGTGCACCTCATGGTAGCGGTGGAGGGCCGCCTCTTCAGGAAGTGGTTCGCTGCAGC 3360
CCCAGACCTGTCCTATTATTTCATTTGGGACAAGACAGACGTCTACAACCAGAAGGTGTT 3420
TGGGCTTTCAGAAGCCTTTGTTTCCGTGGGTTATGAATATGAATCCTGCCCAGATCTAAT 3 80
CCTGTGGGAAAAAAGAACAACAGTGCTGCAGGGCTATGAAATTGACGCGTCCAAGCTTGG 35 0
AGGATGGAGCCTAGACAAACATCATGCCCTCAACATTCAAAGTGGTATCCTGCACAAAGG 3600
GAATGGGGAGAACCAGTTTGTGTCTCAGCAGCCTCCTGTCATTGGGAGCATCATGGGCAA 3660
TGGGCGCCGGAGAAGCATCTCCTGCCCCAGCTGCAACGGCCTTGCTGACGGCAACAAGCT 3720
CCTGGCCCCAGTGGCCCTCACCTGTGGCTCTGACGGGAGCCTCTATGTGGGTGATTTCAA 3780
CTACATTAGAAGGATCTTCCCCTCTGGAAATGTCACCAACATCCTAGAGCTGAGGAATAA 3840
AGATTTCAGACATAGTCACAGTCCAGCACACAAATACTACCTGGCCACAGACCCCATGAG 3900
TGGGGCCGTCTTCCTTTCTGACAGCAACAGCCGGCGGGTCTTTAAAATCAAGTCCACTGT 3960
GGTGGTGAAGGACCTTGTCAAGAACTCTGAGGTGGTTGCGGGGACAGGTGACCAGTGCCT 4020
CCCCTTTGATGACACTCGCTGCGGGGATGGTGGGAAGGCCACAGAAGCCACACTCACCAA 4080
TCCCAGGGGTATTACAGTGGACAAGTTTGGGCTGATCTACTTCGTGGATGGCACCATGAT 4140
CAGACGCATCGATCAGAATGGGATCATCTCCACCCTGCTCGGCTCTAATGATCTCACATC 4 00
AGCCCGGCCACTCAGCTGTGATTCTGTCATGGATATTTCCCAGGTAAGACTGGAGTGGCC 4260
CACAGACTTAGCCATCAACCCAATGGACAACTCACTTTATGTCCTCGACAACAATGTGGT 4320
CCTGCAAATCTCTGAAAACCACCAGGTGCGCATTGTCGCCGGGAGGCCCATGCACTGCCA ,4380
GGTCCCTGGCATTGACCACTTCCTGCTAAGCAAGGTGGCCATCCACGCAACCCTGGAGTC 4440
AGCCACCGCTTTGGCTGTTTCACACAATGGGGTCCTGTATATTGCTGAGACTGATGAGAA 4500
AAAGATCAACCGCATCAGGCAGGTCACCACTAGTGGAGAGATCTCACTCGTTGCTGGGGC 4560
CCCCAGTGGCTGTGACTGTAAAAATGATGCCAACTGTGATTGTTTTTCTGGAGACGATGG 46 0
TTATGCCAAGGATGCAAAGTTAAATACCCCATCTTCCTTGGCTGTGTGTGCTGATGGGGA 4680
GCTCTACGTGGCCGACCTTGGGAACATCCGAATTCGGTTTATCCGGAAGAACAAGCCTTT 4740
CCTCAACACCCAGAA.CATGTATGAGCTGTCTTCACCAATTGACCAGGAGCTCTATCTGTT 4800
TGATACCACCGGCAAGCACCTGTACACCCAAAGCCTGCCCACAGGAGACTACCTGTACAA 4860
CTTCACCTACACTGGGGACGGCGACATCACACTCATCACAGACAACAATGGCAACATGGT 4920
AAATGTCCGCCGAGACTCTACTGGGATGCCCCTCTGGCTGGTGGTCCCAGATGGCCAGGT 4980
GTACTGGGTGACCATGGGCACCAACAGTGCACTCAAGAGTGTGACCACACAAGGACACGA 5040
GTTGGCCATGATGACATACCATGGCAATTCCGGCCTTCTGGCAACCAAAAGCAATGAAAA 5100
CGGATGGACAACATTTTATGAGTACGACAGCTTTGGCCGCCTGACAAATGTGACCTTCCC 5160
TACTGGCCAGGTGAGCAGTTTCCGAAGTGATACAGACAGTTCAGTGCATGTCCAGGTAGA 5220
GACCTCCAGCAAGGATGATGTCACCATAACCACCAACCTGTCTGCCTCAGGCGCCTTCTA 5280
CACACTGCTGCAAGACCAAGTCCGGAACAGCTACTACATCGGGGCCGATGGCTCCTTGCG 5340
GCTGCTGCTGGCCAACGGCATGGAGGTGGCGCTGCAGACTGAGCCCCACTTGCTGGCTGG 5400
CACCGTCAACCCCACCGTGGGCAAGAGGAATGTCACGCTGCCCATCGACAACGGCCTCAA 5460
CCTGGTGGAGTGGCGCCAGCGCAAAGAGCAGGCTCGGGGCCAGGTCACTGTCTTTGGGCG 5520
CCGGCTGCGGGTGCACAACCGAAATCTCCTATCTCTGGACTTTGATCGCGTAACACGCAC 5580
AGAGAAGATCTATGATGACCACCGCAAGTTCACCCTTCGGATTCTGTACGACCAGGCGGG 5640
GCGGCCCAGCCTCTGGTCACCCAGCAGCAGGCTGAATGGTGTCAACGTGACATACTCCCC 5700
TGGGGGTTACATTGCTGGCATCCAGAGGGGCATCATGTCTGAAAGAATGGAATACGACCA 5760
GGCGGGCCGCATCACATCCAGGATCTTCGCTGATGGGAAGACATGGAGCTACACATACTT 5820
AGAGAAGTCCATGGTGCTGCTACTACACAGCCAGAGGCAGTATATCTTTGAGTTCGACAA 5880
GAATGACCGCCTCTCTTCTGTGACGATGCCCAACGTGGCGCGGCAGACACTAGAGACCAT 5940
CCGCTCAGTGGGCTACTACAGAAACATCTATCAGCCCCCTGAGGGCAATGCCTCAGTCAT 6000
ACAGGACTTCACTGAGGATGGGCACCTCCTTCACACCTTCTACCTGGGCACTGGCCGCAG 6060
GGTGATATACAAGTATGGCAAACTGTCAAAGCTGGCAGAGACGCTCTATGACACCACCAA 6120
GGTCAGTTTCACCTATGACGAGACGGCAGGCATGCTGAAGACCATCAACCTACAGAATGA 6180
GGGCTTCACCTGCACCATCCGCTACCGTCAGATTGGGCCCCTGATTGACCGACAGATCTT 6240
CCGCTTCACTGAGGAAGGCATGGTCAACGCCCGTTTTGACTACAACTATGACAACAGCTT 6300 CCGGGTGACCAGCATGCAGGCTGTGATCAACGAGACCCCACTGCCCATTGATCTCTATCG 6360
CTATGATGATGTGTCAGGCAAGACAGAGCAGTTTGGGAAGTTTGGTGTCATTTACTATGA 6420
CATTAACCAGATCATCACCACAGCTGTCATGACCCACACCAAGCATTTTGATGCATATGG 6480
CAGGATGAAGGAAGTGCAGTATGAGATCTTCCGCTCGCTCATGTACTGGATGACCGTCCA 6540
GTATGATAACATGGGGCGAGTAGTGAAGAAGGAGCTGAAGGTAGGACCCTACGCCAATAC 6600
CACTCGCTACTCCTATGAGTATGATGCTGACGGCCAGCTGCAGACAGTCTCCATCAATGA 6660
CAAGCCACTCTGGCGCTACAGCTACGACCTCAATGGGAACCTGCACTTACTGAGCCCTGG 6720
GAACAGTGCACGGCTCACACCACTACGGTATGACATCCGCGACCGCATCACTCGGCTGGG 6780
TGACGTGCAATACAAGATGGATGAGGATGGCTTCCTGAGGCAGCGGGGCGGTGATATCTT 6840
TGAGTACAACTCAGCTGGCCTGCTCATCAAGGCCTACAACCGGGCTGGCAGCTGGAGTGT 6900
CAGGTACCGCTACGATGGCCTGGGGCGGCGCGTGTCCAGCAAGAGCAGCCACAGCCACCA 6960
CCTGCAGTTCTTCTATGCAGACCTGACCAACCCCACCAAGGTCACCCACCTGTACAACCA 70 0
CTCCAGCTCTGAGATCACCTCCCTCTACTACGACTTGCAAGGACACCTCTTTGCCATGGA 7080
GCTGAGCAGTGGTGATGAGTTTTACATAGCTTGTGACAACATCGGGACCCCTCTTGCTGT 7140
CTTTAGTGGAACAGGTTTGATGATCAAGCAAATCCTGTACACAGCCTATGGGGAGATCTA 7200
CATGGATACCAACCCCAACTTTCAGATCATCATAGGCTACCATGGTGGCCTCTATGATCC 7260
ACTCACCAAGCTTGTCCACATGGGCCGGCGAGATTATGATGTGCTGGCCGGACGCTGGAC 7320
TAGCCCAGACCACGAGCTGTGGAAGCACCTTAGTAGCAGCAACGTCATGCCTTTTAATCT 7380
CTATATGTTCAAAAACAACAACCCCATCAGCAACTCCCAGGACATCAΛGTGCTTCATGAC 7440
AGATGTTAACAGCTGGCTGCTCACCTTTGGATTCCAGCTACACAACGTGATCCCTGGTTA 7500
TCCCAAACCAGACATGGATGCCATGGAACCCTCCTACGAGCTCATCCACACACAGATGAA 7560
AACGCAGGAGTGGGACAACAGCAAGTCTATCCTCGGGGTACAGTGTGAAGTACAGAAGCA 7620
GCTCAAGGCCTTTGTCACCTTAGAACGGTTTGACCAGCTCTATGGCTCCACAATCACCAG 7680
CTGCCAGCAGGCTCCAAAGACCAAGAAGTTTGCATCCAGCGGCTCAGTCTTTGGCAAGGG 7740
GGTCAAGTTTGCCTTGAAGGATGGCCGAGTGACCACAGACATCATCAGTGTGGCCAATGA 7800
GGATGGGCGAAGGGTTGCTGCCATCTTGAACCATGCCCACTACCTAGAGAACCTGCACTT 7860
CACCATTGATGGGGTGGATACCCATTACTTTGTGAAACCAGGACCTTCAGAAGGTGACCT 7920
GGCCATCCTGGGCCTCAGTGGGGGGCGGCGAACCCTGGAGAATGGGGTCAACGTCACTGT 7980
GTCCCAGATCAACACAGTACTTAATGGCAGGACTAGACGCTACACAGACATCCAGCTCCA 8040
GTACGGGGCACTGTGCTTGAACACACGCTACGGGACAACGTTGGATGAGGAGAAGGCACG 8100
GGTCCTGGAGCTGGCCCGGCAGAGAGCCGTGCGCCAAGCGTGGGCCCGCGAGCAGCAGAG 8160
ACTGCGGGAAGGGGAGGAAGGCCTGCGGGCCTGGACAGAGGGGGAGAAGCAGCAGGTGCT 8 20
GAGCACAGGGCGGGTGCAAGGCTACGACGGCTTTTTCGTGATCTCTGTCGAGCAGTACCC 8280
AGAACTGTCAGACAGCGCCAACAACATCCACTTCATGAGACAGAGCGAGATGGGCCGGAG 8340 GTGACAGAGAGGAC
A disclosed NOV4 nucleic acid maps to chromosome 11, and is found in at least brain, spinal chord, testis, heart, lung, parathyroid, stomach, breast, colon, epidermis, ovary and kidney. A NOV4 nucleic acid has 7504 of 8359 bases (89%) identical to a gb:GENBANK- ID:AB025413lacc: AB025413.1 mRNA from Mus musculus TEN-M4.
A NOV4 polypeptide (SEQ ID NO: 14) encoded by SEQ ID NO: 13 is 2769 amino acid residues and is presented using the one letter code in Table 4B. Signal P, Psort and/or Hydropathy results predict that NOV4 does not have a signal peptide and is likely to be localized mitochondrial inner membrane with a certainty of 0.8363. hi other embodiments, NOV4 may also be localized to the plasma membrane with a certainty of 0.65 or to the nucleus with a certainty of 0.6000, or microbody with a certainty of 0.3936. Table 4B.
NOV4 Polypeptide
SEQ ID NO:14 DVKERKPYRSLTRRRDAERRYTSSSADSEEGKAPQKSYSSSETLKAYDQDARLAYGSRV 60
KDIVPQEAEEFCRTGANFTLRELGLEEVTPPHGTLYRTDIGLPQCGYSMGAGSDADMEAD 120
TVLSPEHPVRL GRSTRSGRSSCLSSRANSNLT TDTEHENTETDHPGGLQNHARLRTPP 180
PPLSHAHTPNQHHAASINSLNRGNFTPRSNPSPAPTDHSLSGEPPAGGAQEPAHAQENW 240
LNSNIP ETRN GKQPFLGTLQDNLIEMDILGASRHDGAYSDGHF FKPGGTSP FCTTS 300
PGYPLTSSTVYSPPPRPLPRSTFARPAFNLKKPSKYCNWKCAALSAIVISAT VILLAYF 360
VAMHLFGLNWHLQPMEGQMYEITEDTASSWPVPTDVS YPSGGTG ETPDRKGKGTTEGK 420
PSSFFPEDSFIDSGEIDVGRRASQKIPPGTF RSQVFIDHPVH KFNVS GKAALVGIYG 480
RKGLPPSHTQFDFVEL DGRRLLTQEARSLEGTPRQSRGTVPPSSHETGFIQYLDSGI H 540
LAFYNDGKESEWSFLTTAIESVDNCPSNCYGNGDCISGTCHCFLGFLGPDCGRASCPVL 600
CSGNGQYMKGRC CHSGWKGAECDVPTNQCIDVACSNHGTCITGTCICNPGYKGESCEEV 660
DCMDPTCSGRGVCVRGECHCFVG GGTNCETPRATCLDQCSGHGTFLPDTG CSCDPS T 720
GHDCSIElCAADCGGHGVCVGGTCRCEDGWMGAACDQRACHPRCAEHGTCRDGKCECSPG 780
WNGEHCTIAHYLDRWKEGCPGLCNGNGRCTLDLNG HCVCQ G RGAGCDTSMETACGD 840
SKD DGDGLVDCMDPDCCLQPLCHINP CLGSPNP DIIQETQVPVSQQNLHSFYDRIKF 900
LVGRDSTHIIPGENPFDGGHACVIRGQVMTSDGTPLVGVNISFVIMPLFGYTISRQDGSF 960
D VTNGGISIILRFERAPFITQEHTLW PWDRFFVMETIIMRHEENEIPSCDLSNFARPN 1020
PWSPSPLTSFASSCAEKGPIVPEIQALQEEISISGCKMRLSY SSRTPGYKSVLRISLT 1080
HPTIPFNL KVH MVAVEGRLFRKWFAAAPDLSYYFIWDKTDVYNQ VFGLSEAFVSVGY 1140
EYESCPDLILWEKRTTVLQGYEIDASK GGWSLDKHHALNIQSGILHKGNGENQFVSQQP 1200
PVIGSIMGNGRRRSISCPSCNGLADGN LLAPVA TCGSDGSLYVGDFNYIRRIFPSGNV 1260
TNILELR KDFRHSHSPAHKYYLATDPMSGAVFLSDSNSRRVFKI ST WKDLVKNSEV 1320
VAGTGDQCLPFDDTRCGDGGKATEAT TNPRGITVDKFGLIYFVDGTMIRRIDQNGIIST 1380
LLGS D TSARPLSCDSVMDISQVRLEWPTD AINPMDNSLYVLD NWLQISENHQVRI 1440
VAGRPMHCQVPGIDHF LSKVAIHAT ESATA AVSH GV YIAETDEKKINRIRQVTTS 1500
GEISLVAGAPSGCDCKMDANCDCFSGDDGYAKDAK NTPSSLAVCADGELYVADLGNIRI 1560
RFIRKNKPF NTQ MYELSSPIDQELYLFDTTGKHLYTQS PTGDY YNFTYTGDGDIT 1620
ITD NGNMVNVRRDSTGMPLWLVVPDGQVY VTMGTNSA KSVTTQGHELAMMTYHGNSG 1680 bATKSNENG TTFYEYDSFGRLTNVTFPTGQVSSFRSDTDSSVHVQVETSSKDDVTITT 1740
NLSASGAFYTLLQDQVR SYYIGADGSLRLL A GMEVA QTEPHLLAGTVNPTVG RNV 1800
TLPIDNGLNLVE RQRKEQARGQVTVFGRRLRVH RNLLS DFDRVTRTEKIYDDHRKFT 1860
LRI YDQAGRPS WSPSSR NGV VTYSPGGYIAGIQRGIMSERMEYDQAGRITSRIFAD 1920
GKT SYTYLEKSMV L HSQRQYIFEFDKNDR SSVTMPNVARQTLETIRSVGYYR IYQ 1980
PPEGNASVIQDFTEDGHLLHTFYLGTGRRVIYKYGKLSKLAETLYDTTKVSFTYDETAGM 2040 KTIN QNEGFTCTIRYRQIGPLIDRQIFRFTEEGMVNARFDYNYDNSFRVTSMQAVINE 2100
TPLPID YRYDDVSGKTEQFGKFGVIYYDINQIITTAVMTHTKHFDAYGRMKEVQYEIFR 2160
SLMY MTVQYDNMGRWKKELKVGPYA TTRYSYEYDADGQ QTVSI DKPL RYSYD N 2220
GNLHLLSPGNSARLTP RYDIRDRITRLGDVQYKMDEDGFLRQRGGDIFEYNSAGLLIKA 2280
YNRAGS SVRYRYDGLGRRVSS SSHSHHLQFFYAD TNPTKVTH YNHSSSEITSLYYD 2340
LQGHLFAMELSSGDEFYIACDNIGTP AVFSGTG MIKQILYTAYGE1YMDTNPNFQIII 2400
GYHGGLYDPLTKVHMGRRDYDVLAGR TSPDHEL KH SSSNVMPFNLYMFKNMNPISN 460
SQDIKCFMTDVNSWLLTFGFQLHNVIPGYPKPDMDAMEPSYELIHTQ KTQE DNSKSIIi 2520
GVQCEVQKQLKAFVTLERFDQLYGSTITSCQQAPKTKKFASSGSVFGKGVKFAL DGRVT 580
TDIISVANEDGRRVAAILNRAHYLEN HFTIDGVDTHYFVKPGPSEGDLAILGLSGGRRT 640
LENGVNVTVSQINTVLNGRTRRYTDIQLQYGA CLNTRYGTTLDEEKARVLELARQRAVR 700
QAWAREQQR REGEEGLRAWTEGEKQQVLSTGRVQGYDGFFVISVEQYPE SDSA NIHF 760 MRQSEMGRR
The full amino acid sequence of the protein of the invention was found to have 2688 of 2771 amino acid residues (97%) identical to, and 2728 of 2771 amino acid residues (98%) similar to, the 2771 amino acid residue ptnr:SPTREMBL-ACC:Q9WTS7 protein from Mus musculus TEN-M4.
NOV4 also has homology to the amino acid sequences shown in the BLASTP data listed in Table 4C.
Figure imgf000056_0001
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 4D.
Table 4D ClustalW Analysis of NOV4
Tables 4E lists the domain description from DOMAIN analysis results against NOV4. This indicates that the NOV4 sequence has properties similar to those of other proteins known to contain this domain.
1) N0V4 (SEQ ID NO -.13)
2) gi| 16551957 (SEQ ID NO.50)
3) gi| 7657417 (SEQ ID NO:51)
4) gi|l3649010 (SEQ ID N0.52)
5) gi| 1079143 (SEQ ID NO:53)
6) gi| 8922444 (SEQ ID N0.54) 10 20 30 40 50
N0V4 røVKERKPYRS T-RRRDAERRYTSSSADSEEGKAP-QKSYSSSET KAY gi 1 16551957 I gi | 7657417 | MDVKERRPYCSLTKSRREKERRYTNSSADNEECRVPTQKSYSSSETLKAF gi 1 13649010 | MEQTDCKPYQPLPKVKHEMDLAYTSSSDESEDGRKP-RQSYNSRETLHEY gi|l079143| gi I 8922444 I
60 70 80 90 100
NOV4 DQD-ARLAYGSRVKDIVPQEAEEFCRTGANFTLREtJG EgVTPPHGT YR
gi I 7657417 | DHDYSRLLYGNRVKDLVHREADEYTRQGQNFTLRQΪj v SATRRGVAFC gi 113649010 I NQELR MN-YNSQSRKRKEVEKSTQE^EFCETSHT CSGYQ gi 11079143 I jMNFKJ VARCSSPW gi I 8922444 I
110 120 130 140 150
NOV4 TDjG-i|PQCGYSMGAGSDADMEADTVLSPEHPVK£ GRSTR GR& CLf^
gi I 7657417 | AEJKJG-fc|PHRGYS|3jSAGSDADTENEAV] ^PEHAMRjJWGRGVK@GRlS^CLg gi 1 13649010 | Tϋ fHS SRHGY^EMGSDVDTETEGAASPDHALR J IRGMK E_ ^CLf^ gi 1 1079143 I FGjj{GS|lSVLFAFWMLII, TTTGVIK S(QSPPCS^ VGNEA§E^erfAKgf gi I 8922444 I
160 170 180 190 200
N0V4 RA^SNLTLTD^EHENTETDH PGGLQN gi 1 16551957 I -- gi I 7657417 I
Figure imgf000057_0001
ΞEQPSN gi 1 13649010 | RAijSALSLTD^DHERKSDGENGFKFSPVCCDMEAQAGSTQDVQSSPHNQF
Figure imgf000057_0002
gi I 8922444 I
210 220 230 240 250
N0V4 HARLRTPPPPLSHAHTPNQHHAASINgliNRGNFTPRSNP P^PTDHSLgG
gi I 7657417 | NPGQPTLQP PPSHKQHPAQHHPSIT|LNRNSLTNRRNQi3PgpPAALPjSJE gi 113649010 | TFRPLPPPPPPPHACTCARKPPPAADgLQRRSMTTRSQPSjPlAPAPPTgT
Figure imgf000057_0003
gi I 8922444 I
260 270 280 290 300
N0V4 EPPAGGAQEPAHAQEN LLNSNIPI.ETRNLGKQPFLGT QDNLIEMDILG
gi| 7657417| LQTTP ESVQLQDSWVLGSNVPLESR gi] 13649010| QDS VH HNSWVLNSNIPLETR
Figure imgf000057_0004
gi I 8922444 I
310 320 330 340 350
N0V4 ASRHDGAYSDGHF FKPGGTSPLFCTT^PGYPLTSSTVYSjPppgpLøRS^ gi I 7657417 | HF FKTGTGTTPLFSTA^PGYTMASGSVY^PPTJ2P 0Rl i gi|l3649010| HFLFKHGSGSSAIFSAA^QNYP TSN V ^ PPSP SRSI
Figure imgf000058_0001
gi I 8922444 I
360 370 380 390 400
NOV4 FARPAFNLKKPSKYCN KCAALSAIVISAT VILLAYFVAMHLFG N HL
gi I 7657417 | LSRSAFKFKKSSKYCS RCTALCAVGVSV LAILLSYFIAMHLFGLN HL gι|l364901θ| FSRPAFTFNKPYRCCNWKCTALSATAITVT ALLIAYVIAVHLFGLT QL gi 11079143] FP P gι| 8922444 I
410 420 430 440 450
NOV4 QPMEGQMYEITEDTASSWPVPTDVSLYPSGGTGLETPDRKGKGTTEGKPS gi|l6551957| gi I 7657417 I QQTENDTFENGKVNSD- -T VPTNTVSLPSGDN -GKLG-- gi 113649010 I QPVEGE YANGVSKGNRGTESMDTTYSPIGGKVS - - -DKSEK- gi 11079143 I DGTTFG- gi I 8922444 I
460 470 480 490 500
N0V4 SFFPEDSFIDSGEΣDiϊS ϊi! SQ I [3GTS, S VFIDHSvHlSs feiGfe gi I 7657417 I GFTHENNTIDSG jjDl ] R^IjqEψPJJ3GIFJ2RS@ FIDQEQF^^l|§JjQK gi 113649010 I KVFQKGRAIDTGKTO_ESA1QVM^ip5pgGLF3l F@ITIHH lYp[βlI^ώΑ!K gi 11079143 | g5ΩSζ^ ^KEΪQ@YSg|NM@FYQSE@A ^gϊ^X Pj|; gi] 8922444 I
510 520 530 540 550
N0V4 A ,l_,^I^^IζG gsSSEFD!l^^DSRRLLTΩfe'SI,EG^PRQSRG'rvp gi I 7657417 | DA1LϊSh^^KG Pl5'slϊ5iBfeDl3vB--i3-iDl-ls r.ιIAJEOHNLVEaERAGROARS gi j 13649010 ] I^ g:^ EaiIPJ^S^DJ3^KlιMD@KQLVKQDSKGSDDjt;QHSPRN IL gi 11079143 I G&Sjgϊ^lKNALgTgϊ^HgKB lJsgFSAST^TASAAHIjglTR gi I 8922444 I
560 570 580 590 600
N0V4 SSHETGFIQg_XJS@I0lά_AFSS3-@ ES[ VSFL TAlΪESVDNSpSNSγ gi 116551957 I gi I 7657417 | VSLHEAGFIoRLDSiailfa- EW-HCTPigoVSFNTIVlteSVVElgPRNBlH gi 113649010 I SLQE GFIEgtffi,Qgp^IAFSSi3-@KKMi QVFVL AlfelMDDSs NgN gi|l079143| EV fflpgH^FVS'LΞJJSDgDVQlLTF AAVAE MTQNgPNGgs gi j 8922444 I
610 620 630 640 650 NOV4 EJy_fo ssβτ|!^F] .iS:ι |pi^^
gi I 7657417 I gl^Eg^S@τSHSFPSL@PJ!gSRA SSBSGJ!?_|8SSR;SFSτ
Figure imgf000059_0001
gi 110791431 HQEi(LSHSQSNpS3GlDESΞSESVEijΞEsQϊSEΞτNSES:ϊ:BNpSS gi I 8922444 I
660 670 680 690 700
NOV4 |3D^P NQglDVAgsNH@ 3-ςT@τSlSNP^^ESS3|JJEMDSτ^GR@
gi I 7657417 I [^^PTTθJlDPQSGGR§lgrMgs3ASNS^^ENS3|δSSl'DBGENHΞ
Figure imgf000059_0002
gi I 8922444 I
710 720 730 740 750
■■..|....ι....|....i....|....|....,....|....|....ι
Figure imgf000059_0003
gi I 7657417 I
Figure imgf000059_0004
gx 113649010 I lS K@EgHgsτ3GSv»3E P Iι VSθEθ!8lite.i.^jgT,T,lΑnvias|S|ripτ
Figure imgf000059_0005
gi I 8922444]
760 770 780 790 800
NOV4 E^Hi^IE∑S5EGl& 'GGTSRS GiI i^QSASHpκSAil!IS gι|76574i7| E^p_^^ϊBs^EsESv^GGSBR^EGEτΞpASNQSASHpRSaililΞ
Figure imgf000059_0006
gι| 8922444]
810 820 830 840 850
,...|....|....|....|....|....|.. .|.. .|....|....|
N0V4 τgRggκrgEgsP^l^E}i.«lιAJAHYLDRVVKEGffiGLHHG^R^TΪΩLM^l-IS
gi I 7657417 | gEDlκSEgscβ^E!IS__32fflγLD IVKEGBSGLSNs^^τ^DQNS-El
Figure imgf000059_0007
gi I 8922444 I
860 870 880 890 900
N0V4 HS^QLHR§ _^TS^TA3GS3SKJ3J^G[^Jvi^Mj33D^LQP gHINP
Figure imgf000059_0008
gi I 8922444 I 910 920 930 940 950
NOV4 L0 GSJ^lSϊiQETQVPVSQQN HSJ γi3J lKJ3J^GRDg HIIPGENP[|i gi 116551957 I gi I 7657417 I YSRG gDEQSSlϊSQSLQ PSQQAAKSgYDi sSϊfeSDl HVLPGESPJ
Figure imgf000060_0001
gx I 8922444 I --
960 970 980 990 1000
OV GG cE53S^TSDΞτpB^ ¥lFVipi'FGiSI_sΞQ_EsS3ϊvJτlβ gi 116551957 I gx 17657417 I KSI^sH^QfljTADgTPl l^I^FLHYSEYG^ΪTSQ^MJ Si^ANgS gi 113649010 | SRR&C_ff^O^ ID^TPfl^Sffi%)gF HHSnY l'πiSigo!a^sf^»^ lBgl gi 11079143 | ESRfeASSR^TSISMGR^TTTLLEG-|FSiϊτi^B'S3_^lβ gi I 8922444 I
1010 1020 1030 1040 1050
N0V4 iSjlljRJlES^ITg'EHTETOl^RFFpMBTPliJlRH EENEIPgg gi 116551957 I
Figure imgf000060_0002
gx| 13649010| I^ lBl@D!gsS3LPiKR J. ϊS3_HQF:ι:PHI^'IΕiQR WSDPPlg gx 11079143 | G VTJjQSGE^ R ^SRI Q S EV jri it vSSMSEE GI-AVTTT- rg gi I 8922444 I
1060 1070 1080 1090 1100
N0V4 DLSNfA PN|V^Sp p TSi3AS^CA|κGPEϊ^P| lgAϊjQ EIS0SGCIΦ( i3
gi I 7657417 | DLSG_jJVRPSJ3jJVSgP STi|FRi5SPSDS Si §T@WJH[ E T0PGTDljKiϊ gi 113649010 | DISreξISPNgl P PL S[3GG^CPE Gτ[J^P§ gv?Q EIPSPSSF^RJJ gi 11079143 I FAHDJDLMK(3v^ § KHGi|QG^CP SA|3ll S s^ξQ SLQ3PGTGϊιN3 g I 8922444 I
1110 1120 1130 1140 1150
N0V4 S0LH β^^RΪSΪ ^PT^FNBMKSffl^A^E R3FRKwf3 SAPD gi] 16551957 | gi I 7657417 | S0I^3AA^K^ KIT^QAV^FN^MκS?i|3M^A'V!V@ |3]FQgwSpSsPN gi 113649010 | S0LHτ ^K^I_Λ3iLl^Sτ3^G^IKSS A*EgRiJτQζWSp|AIN gi 11079143 I BH!33gAA^]-1^T JKθlflPD B3τΞI_Hl-.t!HlR!ϊτiEiaifflFEgil3EgDPG gi I 8922444 I
1160 1170 1180 1190 1200
N0V4 βSXYyi|DK qvK^ϊ!j5Flϊ]SESF3sB^ESESgpJ3LI^E!I*jJτ fΪQ@
gx 176574171 φ^α^i JKT A^j φ^SES Esj^ ^sgi^Lαt^ra^vjjQSY
Figure imgf000060_0003
gi 110791431 SK^T^AlS iiiiHRiaRiPyigaTTgvffiK-g-HόBTDiaTia- ιViBαιnjύjτκflslelH gx I 8922444 I
1210 1220 1230 1240 1250
Figure imgf000061_0001
gi I 8922444 I
1260 1270 1280 1290 1300
NOV4 ^S^sSPS@^_^DGj^L!SE!TCGSi^LTβΞ_EfXX!33_lFPSvT
Figure imgf000061_0002
gi I 8922444 I
1310 1320 1330 1340 1350
NOV4 Kttijg]jRNKDFRHSHSPaH^llTD||«SGA^I^3SN1g feF^IKS VVV
Figure imgf000061_0003
gi I 8922444 I
1360 1370 1380 1390 1400
....| ....| ....| ....] ....| ....| ....| ....| ....| ....| NOV4 KDLVκ!aΞi5vffiΑB^dS^FfoτRB!^aκg Egτ!lτι 3ι^r.τvnκFGi.1£
Figure imgf000061_0004
gχ|l3649010| KDLSK]S]F§ S5A@ϊ§3Qg33Fi3QSH^^GR|stef sBNSJjJRglJ D HGFp1
Figure imgf000061_0005
gi I 8922444 I
1410 1420 1430 1440 1450
N0V4 3vj^^Ij^Q iIS3ia^Si^LT@-A!^!5_jSg^S^D^'sβTO^E2
Figure imgf000061_0006
gi I 8922444 I
1460 1470 1480 1490 1500
NOV4 H_ΞlHJ3 ^SIιYW^p ^li_JEmQi^IV^ ^OVPGID^FI.I,SK gx] 16551957] gi ] 7657417 ] D|^1^3β^WΪ S^^lf1E^R32X^SrβVP E^PVG-K
Figure imgf000062_0001
gi I 1079143 | EJ^røgiS ^^ ϊi ^ Gicftl^^I.j^TASTA'iξDTD--
1510 1520 1530 1540 1550
....|....|....]....|....|....|....]....|....|....|
NOV4 vglPIAHESA A AVSHN@v[iYlAJ|TJ3EJKIjJj3lRQ 0S@E0SL gg P
Figure imgf000062_0002
gi I 7657417 I Hg Q JΪΪESA ^igVSYS@vJ3Y^ |fJ3E^CKlHl^Q^τSDgEBsL ^IP gi ] 13649010 I glHSHjESAR&Ϊ^VSHSgLJΪ jlTSE KViϊSl JSNSEJS IlSg P
Figure imgf000062_0003
gi I 8922444 I
1560 1570 1580 1590 1600
N0V4 Jc^l8κwnAMBITOFsπDDGγSκDro NTPSgH^CA!feEi.γ^^τ,G!Sιigπ gi] 16551957 I --
Figure imgf000062_0004
gi ] 13649010 ] tolfoWKIDPNBTOgSGDGGYgKP^ K igSBSvSPffiGTLYVgBlLGBivign)
Figure imgf000062_0005
gi I 8922444 I
1610 1620 1630 1640 1650
,...|....|....|....|....|....|....|.. |....|....|
N0V4 SFΪRK^KPFI^TQNrβώS^SlD^lSLiD Sκ2J YΪ3QSIJPSGΪ. Lg i3
gi I 7657417 I j^,Siq|KPLI^SMNFj^Aδ@ D^l^l[ DINgτ||JQY^δl4VgGJ LglSl[3
Figure imgf000062_0006
gi] 8922444 I
1660 1670 1680 1690 1700
Figure imgf000062_0007
gi|7657417| Js@ SiroNDgT gj^SNgJJTIilil' SSPNRMPV V SPDNg I JjT^G
Figure imgf000062_0008
1710 1720 1730 1740 1750
... ... I .... I .... I .... I .... I .... I .... I .... I .... I
N0V4 SAS S^TQGHELAM Tg^^^SJ5jAJEsNEN@w¥TF^Eg3sF|R|5^ T
gx I 7657417 ] GCJJκs«|AQGLELV FτSH@ -^fl5jAJEs3E @ T F 52SEgR©V
Figure imgf000062_0009
gx ] 1079143 I KT^E&S PDNYlTV YEKH@pfE^3 J3j EST@ ^ VYNS3EFg 3J-lAV gi I 8922444 I
1760 1770 1780 1790 1800
.... I .... I .... I .... I ... M ... I ..M.... I • • • • I • • • • I
NOV4 Flϊ^oBssFRsffiτgssvHgoVEΦHsι^-lD8τϊτππs^sG FBτEιJ8!Dov gi I 7657417 I iTNLHGJ^KAIT3DSlBSSSRiESBisS^S SSIDSFETj3fUQ gi|l364901θ| |EEsSFHS|3LgKLTκ2ELDTS Ei-^ STiJ3τATSTI0liljKgENT gi|l079143| IE SF^S EKGAQVK SE^QKEMSS IQG,&,TViy NGAA gi I 8922444 I SJKAI S5DΪES|SR^ED3sp,sSBs^:ι:DS J FΞτRffiQL
1810 1820 1830 1840 1850
NOV4 tøsJSγiGAl»!«^^r.j.j. ijfeB ΑL τ!5B?ι_B^τviπS^G(g-
gi I 7657417 I βsfQIGi^ΞrFYgsSaaSHYQTlSvl^TAHSVAJS gg gi|l364901θ| sτBRtøpMM<;faτF^s5!κiEτπLstei5ππtιH^8lAgtSiGB eg gi] 1079143 I ESSR T DMHTTSITPWSHNLQMEVABYT(I1IAEQS@LIJGBSYPVP^QR gi|8922444| EgSSQΪGJ3^BIIYΞ5lϊΛSHY^TΪH[^TAH3VAiJS
1860 1870 1880 1890 1900
Figure imgf000063_0001
gi 116551957 I gi I 7657417 I
Figure imgf000063_0002
EKBw taSlgldHΪSfel Bπtl
Figure imgf000063_0003
gi ] 1079143 | πEIAGdlAJSRFlS^FVRSiobPLOAGKOSK PPRPffiTE iaϊlKi-tϊ^GDliSlV1 gi| 8922444 I
Figure imgf000063_0004
1910 1920 1930 1940 1950
NOV4 B15ιJiHB5vfl)^BlBEB!Bfl3raτfl^ gi]l6551957|
Figure imgf000063_0005
1960 1970 1980 1990 2000
NOV4 gPGgγiAG[ESiMsiRME53QAS23τΞ2lϊ'i3ϊΞΞ_ΞE_5ϊ ΞI,p-'!2Ξ gi|l6551957] gi|7657417| lsι§oΪAsB^ττs15lιcrø^soB3!lv5^ga^^ιl tti^!i.gϊfedim
Figure imgf000063_0006
gi 11079143 I DRFER≤VSWS^LQ^YS^SRNg^IiNEIKiGEsSM gAjJKDMFGSg iϊ
Figure imgf000063_0007
2010 2020 2030 2040 2050
NOV4 WB!5 — QKj33|F]3κi3i3ss"vi3!i^ASQTOEi3iRS^^_SE^Qi& gi|l6551957| gi|7657417| g^0sa2Ji3s' igi3,Mξ3ilι^S3ϊ__BIJΞSE; gi|l364901θ| ϊ3QP[3cijLSlβΞSMVSIiSIjΞMLSv|0HEEτ[33 gi| 1079143 I κ\raτpRRSDSaLθ5^AGfl30^ll3τBRGHI!ιiRFSLOTlϊ^FFgγoΞBI gi|8922444| 3 j3R0s ιi3!SsvAi2JjτMQi5rRSι^22SSSNS
2060 2070 2080 2090 2100 NOV4 G ASV[3Q]3FTJEJ^H[HH|3FYJ^GHI ^G^ S^ ^T|3ES^J^F0 gi|l6551957| gi|7657417| |S^A|lEτJS0NEEf [3jQEAFiE3si22BLFj^RlξQTi3Jsi liSSSs'0 Esϊ'0 gi|l364901θ| ^τgFj3Q]3S R^βQELHΞGΘSβτ^Qfti2!s!3,β2ϊE^LΞ gi| 1079143 I ^RHPFEILgKfD^QIjjAKIHPHQSGK2M| 3D AG!3JETlJ!AGLgs HYi3 gi| 8922444 I _3K [^iπτ|^EEiaLlWθ^Fff^sl3^ιFBaRfe τ(MsTgi!-TOΞBliϊ5sgπ
2110 2120 2130 2140 2150
NOV4 Hnlϊiflafil HIB^^ gx| 165519571 J£lfiBfaiflBftffiEBr_Aιfc4^d«Md^^
Figure imgf000064_0001
gi 11079143 I BQBJ3TSΪ, S E^EPEELRRE^YHAGJ ,KD^KL[3|GSCNSPA62 1K
Figure imgf000064_0002
2160 2170 2180 2190 2200
NOV4 i^'.^^j^JdiωA&^_i*j^^'.feB^aτ|ψ)^{^^77ffl
Figure imgf000064_0003
2210 2220 2230 2240 2250
N0V4 gi|l6551957| gi|7657417]
Figure imgf000064_0004
gi)l079143| T^IQDSAKQFFAIVD^Qι^^sSLMtTV;KNIDt?E;Rk,ElJDg3LRig κs gi|8922444|
2260 2270 2280 2290 2300
Figure imgf000064_0005
gi|76574i7| Elι_%ι3B3ιH8BHHκHa88^^
Figure imgf000064_0006
2310 2320 2330 2340 2350
N0V4
Figure imgf000064_0007
gi|7657417| sE3S332^tS3aS!_!E!^R-yE!ΞlϊSSTEEE_3Ss ιasτ gi]l3649010| κ^3S_ΞZS2i-S33S_i3EEI κ^ϊ,SϊΞ3Z SI"!D5SSIlϊ;lΞΞi2
Figure imgf000064_0008
gx| 8922444 I 2360 2370 2380 2390 2400
NOV4 faBι»AG.q| .qjSRtø_Vitleilty ^
Figure imgf000065_0001
gi|8922444| ^^ SGf^lgg5^{^Ξ__9fS^QΞΞ3__ΞΞEEΞγi3^I
2410 2420 2430 2440 2450
NOV4
gi|7657417| li^^fc'A'i._J..3SB^
Figure imgf000065_0002
gi|8922444|
2460 2470 2480 2490 2500
Figure imgf000065_0003
gi 176574171 ffeoBatSlSroFlB^laaat. li-MJi^lJBI31^
Figure imgf000065_0004
gi 11079143 I feHKR0pSRΪJlκEκJ5EJ|FfVPSD[^^ϊlJEHJ32E TE 2QS3PH §d gi|8922444| imoraa^aaroFiasi^ictM.^^jiWdA'Jdiii^iiBiFBEia^ga^^
2510 2520 2530 2540 2550
NOV4
Figure imgf000065_0005
gi|l364901θ| E5ErøHp$gQyNL-LEκ[33EEsΞE
Figure imgf000065_0006
gx|8922444| 3ι|ιfE_^Gκ- SAS|jϊSSRHJi^κiH5|j2DBιϊ32|jgj^
2560 2570 2580 2590 2600
NOV4 ^QΪEi 3E^^iϊ iMi33_3SϊHTQMκιSEHisκsøL^^egδ__! gi|l6551957| ^QH[l/ϊ3^_j3κj^j^gAMJ3^^XHTQMK.ϊ E^ sKS0L^^E2QS gx|7657417]
Figure imgf000065_0007
seBEitovppH lS85hrS6Ak! gi|l364901θ|
Figure imgf000065_0008
gi I 8922444 I 83S BBIa{K8feB t5feiaL'ri5HSB5Htolκ sHθMD.iPpHFBffl8lngϋ8AH
2610 2620 2630 2640 2650
NOV4 @L^3V^ F"l(YGf i SCQQAPKTlάffliSG_IE_S^KF KDE-3
Figure imgf000065_0009
gi I 7657417 | g JHLSJjGKMAE^Q gRRK-AGAEQS WJE^^lS^JMLgvjSOSS
Figure imgf000065_0010
Figure imgf000066_0001
gx I 8922444 I g H^E^SCWQ S RR-AGGAQSWLWJfeVK llSSBMlStSQEΞ
2660 2670 2680 2690 2700
■■■■i....|....ι....ι....ι....|....ι....|....i-...ι
NOV4' BJTfalSvESTOGRRt^lTI SHk'itø
Figure imgf000066_0002
gi| 1079143 I VSlV¥DGallS1sV ftD-^SRgFll^sBFI,Eπiπgs^Dd^FH^gigD
Figure imgf000066_0003
2710 2720 2730 2740 2750
.... I .... I .... I ....i.... i ....|....] ... i .... i .... I
Figure imgf000066_0004
gi I 7657417 I fSϊBRJiTsS^SS^XSH^^S ΪB^SΞSζAfe^fS i^
Figure imgf000066_0005
2760 2770 2780 2790 2800
N0V4 BBtriiiia-ft-A-iljBIShfflalijBigBSBBBRolAt^drf
Figure imgf000066_0006
2810 2820 2830 2840 2850
NOV BBτBκB85Bffigf 1gA^^isB j. MHBHiaJE
Figure imgf000066_0007
gi|76574i7| ^ΑHκWaggϊlgγ^ιH^AWJ^lAiϊlSaSBMol5i3JSgιgκig
Figure imgf000066_0008
2860
NOV4 gi|l6551957| gi]7657417| gi|l364901θ| gi 11079143 I SNRRQLKFGELSA gi] 8922444 I Table 4E. Domain Analysis of NON4 gnl|Pfam|pfam01500, Keratin_B2, Keratin, high sulfur B2 protein. High sulfur proteins are cysteine-rich proteins synthesized during the differentiation of hair matrix cells, and form hair fibers in association with hair keratin intermediate filaments. This family has been divided up into four regions, with the second region containing 8 copies of a short repeat. This family is also known as B2 or KAP1.
CD-Length = 144 residues, 87.5% aligned Score = 38.9 bits (89), Expect = 0.004
Query : 630 CIDVACSΝHGTCITGTCICΝPGYKGESCEEVDCMDPTCSGRGVC GECHCFVGWGGTΝC 689
C CS GTC + C + SC + C P CS C R C + C
Sb ct : 5 CGFPTCST GTCGSSCC QPPSCCQPSCCQPVCSQTTCC-RPTCFQSSCCRPSCC
57
Query : 690 ETP- -RATC DQCSGHGTFLPDTGLCSCDPS TGHDCSIEICAaDCGGHGVCVGGTCRCE 747
+T + TC S G+ SC W DC +E
Sbjct: 58 QTSCCQPTCCQSSSCQ TGCGIGSCRTR CRPDCRVΞ
93
Query : 748 DGWMGAACDQRACHPRCAEHGTCRDGKCECS PGWΝGEHC 786
C C C C+ + S P + G+ C Sbj ct : 94 GTCLPPCC SCTPPTCCQPVSAQASCCRPSYCGQSC 130
The novel TEΝ-M-like protein encoded by the gene of invention has highest homology to the mouse TEΝ-M4 protein, which belongs to the ODZ/TENM family of proteins. This family was first identified in Drosophila as being a pair-rule gene affecting segmentation of the early embryo. It was the first pair-rule gene identified that was not a transcription factor, but a type II transmembrane protein. Vertebrate homologs of the TENM family have been identified in mouse and zebrafish. In the mouse, TEN-M4 expression was found to be on the cell surface, in the brain, trachea as well as developing limb and bone. Analysis of the TEN- MI protein reveals that it can bind to itself, making it likely that TEN-M4 may be a dimeric moiety as well. In cell culture experiments, fragments of the TEN-M protems can bind the Drosophila PS2 integrins. In addition, members of the TEN-M family have been identified to be downstream of the endoplasmic reticulum stress response pathway, which alters the response of cells to their environment. This suggests that the ODZ/TENM family may be involved in cell adhesion, spreading and motility. Translocations leading to the fusion of this gene with the N G1/HGL gene from chromosome 8 have been found to generate a paracrine growth factor for one mammary carcinoma cell line, termed gamma-heregulin. Therefore this novel gene may have widespread implications in development, regeneration and carcinogenesis of various tissues. Two new potential ligands of the Drosophila PS2 integrins have been characterized by functional interaction in cell culture. These potential ligands are a new Drosophila laminin alpha2 chain encoded by the wing blister locus and Ten-m, an extracellular protein known to be involved in embryonic pattern formation. As with previously identified PS2 ligands, both contain RGD sequences, and RGD-containing fragments of these two proteins (DLAM-RGD and TENM-RGD) can support PS2 integrin-mediated cell spreading. In all cases, this spreading is inhibited specifically by short RGD-containing peptides. As previously found for the PS2 ligand tiggrin (and the tiggrin fragment TIG-RGD), TENM-RGD induces maximal spreading of cells expressing integrin containing the alphaPS2C splice variant. This is in contrast to DLAM-RGD, which is the first Drosophila polypeptide shown to interact preferentially with cells expressing the alphaPS2 m8 splice variant. The betaPS integrin subunit also varies in the presumed ligand binding region as a result of alternative splicing. For TIG-RGD and TENM-RGD, the beta splice variant has little effect, but for DLAM-RGD, maximal cell spreading is supported only by the betaPS4A form of the protein. Thus, the diversity in PS2 integrins due to splicing variations, in combination with diversity of matrix ligands, can greatly enhance the functional complexity of PS2-ligand interactions in the developing animal. The data also suggest that the splice variants may alter regions of the subunits that are directly involved in ligand interactions, and this is discussed with respect to models of integrin structure.
A sequence of about thirty to forty amino-acid residues long found in the sequence of epidermal growth factor (EGF) has been shown to be present, in a more or less conserved form, in a large number of other, mostly animal proteins. The list of proteins currently known to contain one or more copies of an EGF-like pattern is large and varied. The functional significance of EGF domains in what appear to be unrelated proteins is not yet clear. However, a common feature is that these repeats are found in the extracellular domain of membrane-bound proteins or in proteins known to be secreted (exception: prostaglandin G/H synthase). The EGF domain includes six cysteine residues which have been shown (in EGF) to be involved in disulfide bonds. The main structure is a two-stranded beta-sheet followed by a loop to a C-terminal short two-stranded sheet. Subdomains between the conserved cysteines vary in length. The NHL (NCL-1, HT2A and LIN-41) repeat is found in a variety of enzymes of the copper type II, ascorbate-dependent monooxygenase family which catalyse the C- terminus alpha-amidation of biological peptides. The repeat also occurs in a human zinc finger protein that specifically interacts with the activation domain of lentiviral Tat proteins. The repeat domain that is often associated with RING finger and B-box motifs (see, Ben-Zur T, Dev Biol 2000 Jan 1;217(1): 107-20; Adelaide J, Int J Oncol 2000 Apr;16(4):683-8 ; Wang XZ, Oncogene 1999 Oct 7;18(41):5718-21; Schaefer G, Oncogene 1997 Sep 18;15(12):1385- 94 ; Wang XZ, EMBO J 1998 Jul l;17(13):3619-30; Baumgartner S, EMBO J 1994 Aug 15;13(16):3728-40; Otaki JM, Dev Biol 1999 Aug 1;212(1):165-81; Mieda M, Mech Dev 1999 Sep;87(l-2):223-7; Oohashi T, J Cell Biol 1999 May 3;145(3):563-77; Graner MW, J Biol Chem 1998 Jul 17;273(29):18235-41, incorporated herein by reference).
The protein similarity information, expression pattern, and map location for the TEN- M4-like protein and nucleic acid disclosed herein suggest that this TEN-M4-like protein may have important structural and/or physiological functions characteristic of this family. Therefore, the nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications and as a research tool. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo (vi) biological defense weapon.
The NOV4 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications implicated in various diseases and disorders described below and/or other pathologies. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from: cardiac diseases, myocardial contractility in failing heart and other diseases, disorders and conditions of the like. The disclosed NON4 nucleic acid of the invention encoding a TEΝ-M4-like protein includes the nucleic acid whose sequence is provided in Table 4A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases maybe changed from the corresponding base shown in Table 4A while still encoding a protein that maintains TEN-M4-like protein-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject, hi the mutant or variant nucleic acids, and their complements, up to about 11 percent of the bases may be so changed.
The disclosed NON4 protein of the invention includes the TEΝ-M4-like protein whose sequence is provided in Table 3B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 4B while still encoding a protein that maintains beta adrenergic receptor kinase-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 3 percent of the residues may be so changed.
The protein similarity information, expression pattern, and map location for TEN-M4- like protein and nucleic acid (NON4) disclosed herein suggest that ΝON4 may have important structural and/or physiological functions characteristic of the TEΝ-M4 protein family. Therefore, the NOV4 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo.
The NOV4 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications implicated in various diseases and disorders described below. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from: Non Hippel-Lindau (NHL) syndrome, Alzheimer's disease, stroke, tuberous sclerosis, hypocalcaemia, Parkinson's disease, Huntington's disease, cerebral palsy, epilepsy, Lesch-Νyhan syndrome, multiple sclerosis, ataxia-telangiectasia, leukodystrophies, behavioral disorders, addiction, anxiety, pain, neurodegeneration, fertility disorders, hyperparathyroidism, hypoparathyroidism, cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-N) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (NSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation disorders, diabetes, autoimmune disease, renal artery stenosis, interstitial nephritis, glomerulonephritis, polycystic kidney disease, systemic lupus erythematosus, renal tubular acidosis, IgA nephropathy, hypocalcaemia, asthma, emphysema, scleroderma, allergy, ARDS, Hirschsprung's disease, Crohn's disease, appendicitis, inflammatory bowel disease, gastric ulcers, psoriasis, actinic keratosis, acne, hair growth loss, allopecia, pigmentation disorders, endocrine disorders and cancer and other diseases, disorders and conditions of the like. The NON4 nucleic acid, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
ΝON4 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-ΝOVX Antibodies" section below. The disclosed ΝOV4 polypeptide has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV4 epitope is from about amino acids 1 to 400. In another embodiment, a contemplated NOV4 epitope is from about amino acids 450 to 520. In other specific embodiments, contemplated NOV4 epitopes are from about amino acids 750 to 850, 1100 to 1200, 1250 to 1400, 1490 to 1750, 1760 to 2300, 2400 to 2600, and 2650 to 2725.
NOV5
NOV5 includes two Out At First-like proteins disclosed below. The disclosed sequences have been named NOV5a and NOV5b.
NOV5a
A disclosed NOV5a nucleic acid of 822 nucleotides identified as SEQ ID NO: 15 (also referred to as CG55764-01) encoding an Out At First-like protein is shown in Table 5A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 1-3 and ending with a TGA codon at nucleotides 820-822.
Table 5A.
NOV5a Polynucleotide
SEQ ID NO;15
ATGCGCCTTCCCGGGGTACCCCTGGCGCGCCCTGCGCTGCTGCTGCTGCTGCCGCTGCTCGCGCCGCTGC TGGGAACGGGTGCGCCGGCCGAGCTGCGGGTCCGCGTGCGGCTGCCGGACGGCCAGGTGACCGAGGAGAG CCTGCAGGCGGACAGCGACGCGGACAGCATCAGCCTCGAGCTGCGCAAGCCCGACGGCACCCTCGTCTCC TTCACCGCCGACTTCAAGAAGGATGTGAAGGTCTTCCGGGCCCTGATCCTGGGGGAGCTGGAGAAGGGGC AGAGTCAGTTCCAGGCCCTCTGCTTTGTCACCCAGCTGCAGCACAATGAGATCATCCCCAGTGAGGCCAT GGCCAAGCTCCGGCAGAAAAATCCCCGGGCAGTGCGGCAGGCGGAGGAGGTTCGGGGTCTGGAGCATCTG CACATGGATGTCGCTGTCAACTTCAGCCAGGGGGCCCTGCTGAGCCCCCATCTCCACAACGTGTGTGCCG AGGCCGTGGATGCCATCTACACCCGCCAGGAGGATGTCCGGTTCTGGCTGGAGCAAGGTGTGGACAGTTC TGTGTTCGAGGCTCTGCCCAAGGCCTCAGAGCAGGCGGAGCTGCCTCGCTGCAGGCAGGTGGGGGACCGC GGGAAGCCCTGCGTCTGCCACTATGGCCTGAGCCTGGCCTGGTACCCCTGCATGCTCAAGTACTGCCACA GCCGCGACCGGCCCACGCCCTACAAGTGTGGCATCCGCAGCTGCCAGAAGAGCTACAGCTTTGACTTCTA CGTGCCCCAGAGGCAGCTGTGTCTCTGGGATGAGGATCCCTACCCAGGCTAG The NON5a nucleic acid was identified on chromosome 11 and has 455 of 733 bases (62%) identical to a gb:GEΝBAΝK-ID:DROOAFPR]acc:L31349.1 mRNA from D. melanogaster (mRNA for out at first (oaf)).
A disclosed NOV5a polypeptide (SEQ ID NO.T 6) encoded by SEQ ID NO: 15 is 273 amino acid residues and is presented using the one-letter code in Table 5B. Signal P, Psort and/or Hydropathy results predict that NOV5a has a signal peptide and is likely to be localized outside the cell with a certainty of 0.7523. In other embodiments, NOV5a may also be localized to the endoplasmic reticulum with a certainty of 0.1000 or microbody with a certainty of 0.1000. The most likely cleavage site is between positions 27 and 28: residues GTG-AP.
Figure imgf000072_0001
The disclosed NOV5a amino acid sequence has 106 of 274 amino acid residues (38%) identical to, and 154 of 274 amino acid residues (56%) similar to, the 487 amino acid residue ρtnr:SWISSNEW-ACC:Q9NLA6 protein from Drosophila melanogaster (fruit fly) (Out At First protein).
The Out At First Protein disclosed in this invention is expressed in at least the following tissues: Adipose, Adrenal Gland/Suprarenal gland, Amygdala, Aorta, Artery, Ascending Colon, Bone, Bone Marrow, Brain, Brown adipose, Cartilage, Cervix, Cochlea, Colon, Coronary Artery, Dermis, Duodenum, Epidermis, Hair Follicles, Heart, Hippocampus, Kidney, Kidney Cortex, Liver, Lung, Lymph node, Lymphoid tissue, Mammary gland/Breast, Myometrium, Esophagus, Ovary, Oviduct Uterine Tube/Fallopian tube, Pancreas, Parotid Salivary glands, Peripheral Blood, Pituitary Gland, Prostate, Respiratory Bronchiole, Retina, Salivary Glands, Skin, Small Intestine, Spinal Chord, Spleen, Stomach, Synovium/Synovial membrane, Thalamus, Thymus, Thyroid, Trachea, Urinary Bladder, Uterus, Vein, Vulva, Whole Organism. This information was derived by determining the tissue sources of the sequences that were included in the invention including but not limited to SeqCalling sources, Public EST sources, Literature sources, and/or RACE sources. NOV5b
A disclosed NOV5b nucleic acid of 1362 nucleotides identified as SEQ ID NO: 17 (also referred to as CG55764-02) encoding a novel serine/threonine kinase-like protein is shown in Table 5C. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 1-3 and ending with a TGA at nucleotides 820-822.
Table 5C.
NOV5b Polynucleotide
SEQ ID NO:17
ATGCGCCTTCCCGGGGTACCCCTGGCGCGCCCTGCGCTGCTGCTGCTGCTGCCGCTGCTCGCGCCGCTGC TGGGAACGGGTGCGCCGGCCGAGCTGCGGGTCCGCGTGCGGCTGCCGGACGGCCAGGTGACCGAGGAGAG CCTGCAGGCGGACAGCGACGCGGACAGCATCAGCCTCGAGCTGCGCAAGCCCGACGGCACCCTCGTCTCC TTCACCGCCGACTTCAAGAAGGATGTGAAGGTCTTCCGGGCCCTGATCCTGGGGGAGCTGGAGAAGGGGC AGAGTCAGTTCCAGGCCCTCTGCTTTGTCACCCAGCTGCAGCACAATGAGATCATCCCCAGTGAGGCCAT GGCCAAGCTCCGGCAGAAAAATCCCCGGGCAGTGCGGCAGGCGGAGGAGGCTCGGGGTCTGGAGCATCTG CACATGGATGTCGCTGTCAACTGCAGCCAGGGGGCCCTGCTGAGCCCCCATCTCCACAACGTGTGTGCCG AGGCCGTGGATGCCATCTACACCCGCCAGGAGGATGTCCGGTTCTGGCTGGAGCAAGGTGTGGACAGTTC TGTGTTCGAGGCTCTGCCCAAGGCCTCAGAGCAGGCGGAGCTGCCTCGCTGCAGGCAGGTGGGGGACCGC GGGAAGCCCTGCGTCTGCCACTATGGCCTGAGCCTGGCCTGGTACCCCTGCATGCTCAAGTACTGCCACA GCCGCGACCGGCCCACGCCCTACAAGTGTGGCATCCGCAGCTGCCAGAAGAGCTACAGCTTCGACTTCTA CGTGCCCCAGAGGCAGCTGTGTCTCTGGGATGAGGATCCCTACCCAGGCTAGGGTGGGAGCAACCTGGCG AGTGGCTGCTCTGGGCCCACTGCTCTTCACCAGCCACTAGAGGGGGTGGCAACCCCCACCTGAGGCCTTA TTTCCCTCCCTCCCCACTCCCCTGGCCCTAGAGCCTGGGCCCCTCTGGCCCCATCTCACATGACTGTGAA GGGGGTGTGGCATGGCAGGGGGTCTCATGAAGGCACCCCCATTCCCACCCTGTGCCTTCCTTGCGGGCAG AGAGGGAGAGAAGGGCTCCCCAGATCTACACCCCTCCCTCCTGCATCTCCCCTGGAGTGTTCACTTGCAA GCTGCCAAAACATGATGGCCTCTGGTTGTTCTGTTGAACTCCTTGAACGTTTAGACCCTAAAAGGAGTCT ATACCTGGACACCCACCTCCCCAGACACAACTCCCTTCCCCATGCACACATCTGGAAGGAGCTGGCCCCT CAGTCCCTTCCTACTCCCCAACAAGGGGCTCACTATCCCCAAAGAAGGAGCTGTTGGGGACCCACGACGC AGCCCCTGTACTGGATTACAGCATATTCTCAT
The NOV5b nucleic acid was identified on chromosome 11 and has 456 of 733 bases (62%) identical to a gb:GENBANK-ID:DROOAFPR]acc:L31349.1 mRNA from D. melanogaster (mRNA for out at first (oaf)).
A disclosed NOV5b polypeptide (SEQ ID NO:18) encoded by SEQ ID NO:17 is 273 amino acid residues and is presented using the one-letter code in Table 5D. Signal P, Psort and or Hydropathy results predict that NOV5b has a signal peptide and is likely to be localized outside the cell with a certainty of 0.7523. In other embodiments, NOV5b may also be localized to the endoplasmic reticulum with a certainty of 0.1000 or microbody with a certainty of 0.1000. The most likely cleavage site is between positions 27 and 28: residues GTG-AP.
Figure imgf000073_0001
EQGVDSSVFEALPKASEQAELPRCRQVGDRGKPCVCHYGLSLA YPCMLKYCHSRDRPTP 240 Y CGIRSCQKSYSFDFYVPQRQLCLWDEDPYPG
The disclosed NOV5b amino acid sequence has 106 of 274 amino acid residues (38%) identical to, and 154 of 274 amino acid residues (56%) similar to, the 487 amino acid residue ρtnr:SWISSNEW-ACC:Q9NLA6 protein from Drosophila melanogaster (fruit fly) (Out At First protein).
The NOV5b Out At First Protein disclosed in this invention is expressed in at least the following tissues: Adipose, Adrenal Gland/Suprarenal gland, Amygdala, Aorta, Artery, Ascending Colon, Bone, Bone Marrow, Brain, Brown adipose, Cartilage, Cervix, Cochlea, Colon, Coronary Artery, Dermis, Duodenum, Epidermis, Hair Follicles, Heart, Hippocampus, Kidney, Kidney Cortex, Liver, Lung, Lymph node, Lymphoid tissue, Mammary gland/Breast, Myometrium, Esophagus, Ovary, Oviduct/Uterine Tube/Fallopian tube, Pancreas, Parotid Salivary glands, Peripheral Blood, Pituitary Gland, Prostate, Respiratory Bronchiole, Retina, Salivary Glands, Skin, Small Intestine, Spinal Chord, Spleen, Stomach, Synovium/Synovial membrane, Thalamus, Thymus, Thyroid, Trachea, Urinary Bladder, Uterus, Vein, Vulva, Whole Organism. This information was derived by determining the tissue sources of the sequences that were included in the invention including but not limited to SeqCalling sources, Public EST sources, Literature sources, and/or RACE sources.
NOV5b also has homology to the amino acid sequences shown in the BLASTP data listed in Table 5E.
Figure imgf000075_0003
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 5F.
Table 5F. ClustalW Sequence Alignment l)NOV5a (SEQ ID NO:15) 2)NOV5b (SEQ ID NO: 17) 3) gi 117136996 | (SEQ ID NO: 55)
4) gi I 7321824 I (SEQ ID NO:56) 5)gi|l2643516| (SEQ ID NO: 57) 6) gi j 113869611 (SEQ ID NO: 58)
10 20 30 40 50
NOV5a RJJPG P
NOV5b MRJJPGV P gi 117136996 | MI KEEHPHQSIETgAN A^Q2gVR RMA[33KA&SRTRJ33AHGNCCGi3vBi gi I 7321824 | MILKEEHPHQSIETS NS ^Q^V RM_^3K 1SRTRJ33AHG CCG|3v2 gi 112643516 | MI KEEHPHQSIE SANS^'QSS R R AJUJ A^SRTRSSAHGNCCGSVE
Figure imgf000075_0001
60 70 80 90 100
Figure imgf000075_0002
gi| 7321824 I gi 112643516 I BκNι33Sκl!E^^*t^*-_Λ8lw^J^aτi ^ wivm ω≤ailfe
Figure imgf000076_0001
110 120 130 140 150
NOV5a bsDAf^S^Sfl^ ^p|iIS^^FTAi^Rκa^ ^FR^ipi^ ^Ka»a»J t^
NOV5b bSDAfflsHsira Rl5pHe5^SFTAiWRiatB FRMlBI^ il_ ^^g^j gi|l7136996| gi|7321824|
gi|ll38696l|
160 170 180 190 200
NOV5a SJSLQHN lSp gSggSSSi^^^Q^ffi^LiHL^V^B11^^
NOVSb S3vSgLQH^ιøp E2ϊ233SΞ33^^QΛi3__ ΞLΞHLHl!]DVJ IδS gi I 7321824 I *>a Vύ:«aiι;<c.ι)3>fcWA>ja>JC««>:W)C«^5iiύιιι»aMaaa» <e.:)3iιii < af=MMrt)*{'»:,
Figure imgf000076_0002
gi|ll38696l| w^Λ^i<a^i^ιl^w.^>jajt«aa.):<κ>aiiiigιaι.-)^^»):<0:^ιlΥ<ιiaw.Mrt)*^:i
210 220 230 240 250
NOV5a G L^PgJjH /ES^VJjglSTSQE1!^!^^-SVDS§VF[^JLP ASEQA
NOV5b GAL3idPJΪgHmSSElEEQES3VRF3]|Q-gVDSgVFgS PKASEQA
Figure imgf000076_0003
gi 1113869611 b».8Uι);liH.»lsl-
260 270 280 290 300
NOV5a E lϊHRt)BGDRGK|gSV^YGLSMBgaiy-W!a8lHSfe D--
NOV5b E PlϊSlRk^GDRGKtgSyfeYGLSa BgiBliyfflSaaHS^ D--
Figure imgf000076_0004
gi I 7321824 I gi|l2643516| gi|ll38696l|
310 320 330 340 350
NOV5a
NOV5b gi|l7136996|
Figure imgf000076_0005
Figure imgf000076_0006
gi| 12643516] MSIBM wats ai i, RRGELQLMQMRCARRRN gi| 11386961] [X ^-tήiiMdiMtai-iiaMMiwMMBWMaww iaaasisis
360 370 380 390 400
NOV5a -DPYJJS-
Figure imgf000076_0007
gi I 7321824 I GSEFGDDASATC^GETRAATTTATITGGGAGGSGKDTTAGTTTTTNKLH gi 112643516 | GSEFGDDASATcf33GETRAATTTATITGGGAGGSGKDTTAATTTTTNKLR
410 420 430 440 450
NOV5a
NOVSb
giI 7321824 ) QLLLLVQQQMPFTLWSFPVHHISQSHHQSQSQHKPSRQQKQHQHHSQVAP gi 112643516 | QLLLLVQQQMPFALWSFPVHHISQSHHQSQSQHKPSRQQKQHQHHSQVAP
460 470 480
NOV5a '
NOVSb gi I 17136996 I gi I 7321824 | TSHHQSSSSTPPTPSTSSSPPSSSSSSSSSAMAAIVA gi 112643516 | TSHHQSSSSTPPTPSTSSSPPSSSSSSSSSAMAAIVA
Tables 5G-I list the domain description from DOMAIN analysis results against NOV5a. This indicates that the NOV5a sequence has properties similar to those of other proteins known to contain this domain.
Table 5G. Domain Analysis of NOV5
qi 1171369961 ref I NP 477040.1| oaf-Pi; transcript near decapentaplegic; transcript-near-decapentaplegic; near dpp complementation group 1 [Drosophila melanogaster]
CD- Length = 332 Score = 202 bits (515), Expect = 2e-51
This sequence from human chromosome 11 encodes for a novel protein which shows some sequence similarity to the Drosophila melanogaster Out At First (OAF) protein. Out At First is expressed in clusters of cells during germband extension, throughout the developing nervous system, and in the gonads of both sexes throughout the lifecycle. Mutation of the Drosophila gene is fatal and causes nervous system defects.
The disclosed NOV5 nucleic acid of the invention encoding an Out At First-like protein includes the nucleic acid whose sequence is provided in Table 5 A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 5A while still encoding a protein that maintains its Out At First-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant NOV5a and NOV5b nucleic acids, and their complements, up to about 38 percent of the bases may be so changed.
The disclosed NOV5a protein of the invention includes the Out At First-like protein whose sequence is provided in Table 5B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 5B while still encoding a protein that maintains its Out At First-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protem, up to about 62 percent of the residues may be so changed.
The disclosed NOV5b protein of the invention includes the Out At First-like protein whose sequence is provided in Table 5D. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 5D while still encoding a protein that maintains its Out At First-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protem, up to about 62 percent of the residues may be so changed.
The NOV5 nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in various diseases, disorders and conditions. The NOV5 nucleic acid, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOV5 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-NOVX Antibodies" section below. The disclosed NOV5a polypeptide has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV5a epitope is from about amino acids 40 to 75. In another embodiment, a contemplated NOV5a epitope is from about amino acids 80 to 87. In other specific embodiments, contemplated NOV5a epitopes are from about amino acids 95 to 105, 110 to 145, 155 to 180, and 225 to 260.
The disclosed NOV5b polypeptide has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV5b epitope is from about amino acids 40 to 75. In another embodiment, a contemplated NOV5b epitope is from about amino acids 80 to 90. In other specific embodiments, contemplated NOV5b epitopes are from about amino acids 95 to 105, 110 to 145, 160 to 220, and 225 to 260.
NON6
ΝOV6 includes two EphA6/ehk-2-like proteins disclosed below. The disclosed sequences have been named NOV6a and NOV6b.
NOV6a
A disclosed NOV6a nucleic acid of 3641 nucleotides identified as SEQ ID NO: 19 (also referred to as CG55704-01) encoding an EphA6/ehk-2-like protein is shown in Table 6A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 19- 2 land ending with a TGA codon at nucleotides 3124-3126. Putative untranslated regions are indicated by underline.
Table 6A.
NOV6a Polynucleotide
SEQ ID NO:19
AGAGAACCAGCGAGAGCCATGGGGGGCTGCGAAGTCCGGGAATTTCTTTTGCAATTTGGT 60
TTCTTCTTGCCCCTGCTGACAGCTTGGACCGGCGACTGCAGTCACGTCTCCAACCAAGTT 120
GTGTTGCTTGATACATCTACAGTGATGGGAGAACTAGGATGGAAAACATATCCACTGAAT 180
GGGTGGGATGCCATTACTGAAATGGATGAACACAACAGGCCCATACATACATACCAGGTA 240
TGCAATGTCATGGAACCAAACCAGAACAACTGGCTTCGTACTAACTGGATCTCTCGTGAT 300
GCTGCTCAGAAAATCTATGTGGAAATGAAGTTCACATTGAGAGATTGTAACAGCATCCCA 360
TGGGTCTTGGGAACGTGTAAAGAAACATTTACTCTGTATTATATTGAATCTGACGAATCC 420
CACGGAACTAAATTCAAGCCAAGCCAATATATASAGATTGACACAATTGCTGCGGATGAG 480
AGTTTTACTCAGATGGATTTGGGTGATCGCATCCTTAAACTCAACACTGAAATTCGTGAG 540
GTGGGGCCTATAGAAAGGAAAGGATTTTATCTGGCTTTTCAAGACATTGGGGCGTGCATT 600
GCCCTGGTTTCAGTCCGTGTTTTCTACAAGAAATGCCCCTTCACTGTTCGTAACTTGGCC 660
ATGTTTCCTGATACCATTCCAAGGGTTGATTCCTCCTCTTTGGTTGAAGTACGGGGTTCT 720
TGTGTGAAGAGTGCTGAAGAGCGTGACACTCCTAA&CTGTATTGTGGAGCTGATGGAGAT 780
TGGCTGGTTCCTCTTGGAAGGTGCATCTGCAGTACAGGATATGAAGAAATTGAGGGTTCT 840
TGCCATGCTTGCAGACCAGGATTCTATAAAGCTTTTGCTGGGAACACAAAATGTTCTAAA 900
TGTCCTCCACACAGTTTAACATACATGGAAGCAACTTCTGTCTGTCAGTGTGAAAAGGGT 960
TATTTCCGAGCTGAAAAAGACCCACCTTCTATGGCATGTACCAGGCCACCTTCAGCTCCT 1020
AGGAATGTGGTTTTTAACATCAATGAAACAGCCCTTATTTTGGAATGGAGCCCACCAAGT 1080
GACACAGGAGGGAGAAAAGATCTCACATACAGTGTAATCTGTAAGAAATGTGGCTTAGAC 1140
ACCAGCCAGTGTGAGGACTGTGGTGGAGGACTCCGCTTCATCCCAAGACATACAGGCCTG 1200 ATCAACAATTCCGTGATAGTACTTGACTTTGTGTCTCACGTGAft.TTACACCTTTGAAATA 1 60
GAAGCAATGAATGGAGTTTCTGAGTTGAGTTTTTCTCCCAAGCCATTCACAGCTATTACA 1320
GTGACCACGGATCAAGATGCACCTTCCCTGATAGGTGTGGTAAGGAAGGACTGGGCATCC 1380
CAAAATAGCATTGCCCTATCATGGCAAGCACCTGCTTTTTCCAATGGAGCCATTCTGGAC 14 0
TACGAGATCAAGTACTATGAGAAAGAACATGAGCAGCTGACCTACTCTTCCACAAGGTCC 1500
AAAGCCCCCAGTGTCATCATCACAGGTCTTAAGCCAGCCACCAAATATGTATTTCACATC 1560
CGAGTGAGAACTGCGACAGGATACAGTGGCTACAGTCAGAAATTTGAATTTGAAACAGGA 1620
GATGAAACTTCTGACATGGCAGCAGAACAAGGACAGATTCTCGTGATAGCCACCGCCGCT 1680
GTTGGCGGATTCACTCTCCTCGTCATCCTCACTTTATTCTTCTTGATCACTGGGAGATGT 1740
CAGTGGTACATAAAAGCCAAGATGAAGTCAGAAGAGAAGAGAAGAAACCACTTACAGAAT 1800
GGGCATTTGCGCTTCCCGGGAATTAA?iACTTACATTGATCCAGATACATATGAAGACCCA 1860
TCCCTAGCAGTCCATGAATTTGCAAAGGAGATTGATCCCTCAAGAATTCGTATTGAGAGA 1920
GTCATTGGGGCAGGTGAATTTGGAGAAGTCTGTAGTGGGCGTTTGAAGACACCAGGGAAA 1980
AGAGAGATCCCAGTTGCCATTAAAACTTTGAAAGGTGGCCACATGGATCGGCAAAGAAGA 20 0
GATTTTCTAAGAGAAGCTAGTATCATGGGCCAGTTTGACCATCCAAACATCATTCGCCTA 2100
GAAGGGGTTGTCACCAAAAGATCCTTCCCGGCCATTGGGGTGGAGGCGTTTTGCCCCAGC 2160
TTCCTGAGGGCAGGGTTTTTAAATAGCATCCAGGCCCCGCATCCAGTGCCAGGGGGAGGA 220
TCTTTGCCCCCCAGGATTCCTGCTGGCAGACCAGTAATGATTGTGGTGGAATATATGGAG 2280
AATGGATCCCTAGACTCCTTTTTGCGGAAGCATGATGGCCACTTCACAGTCATCCAGTTG 2340
GTCGGAATGCTCCGAGGCATTGCATCAGGCATGAAGTATCTTTCTGATATGGGTTATGTT 2 00
CATCGAGACCTAGCGGCTCGGAATATACTGGTCAATAGCAACTTAGTATGCAAAGTTTCT 2460
GATTTTGGTCTCTCCAGAGTGCTGGAAGATGATCCAGAAGCTGCTTATACAACAACTGGT 2520
GGAAAAATCCCCATAAGGTGGACAGCCCCAGAAGCCATCGCCTACAGAAAATTCTCCTCA 2580
GCAAGCGATGCATGGAGCTATGGCATTGTCATGTGGGAGGTCATGTCCTATGGAGAGAGA 2640
CCTTATTGGGAAATGTCTAACCAAGATGTCATTCTGTCCATTGAAGAAGGGTACAGACTT 2700
CCAGCTCCCATGGGCTGTCCAGCATCTCTACACCAGCTGATGCTCCACTGCTGGCAGAAG 2760
GAGAGAAATCACAGACCAAAATTTACTGACATTGTCAGCTTCCTTGACAAACTGATCCGA 820
AATCCCAGTGCCCTTCACACCCTGGTGGAGGACATCCTTGTAATGCCAGAGTCCCCTGGT 2880
GAAGTTCCGGAATATCCTTTGTTTGTCACAGTTGGTGACTGGCTAGATTCTATAAAGATG 29 0
GGGCAATACAAGAATAACTTCGTGGCAGCAGGGTTTACAACATTTGACCTGATTTCAAGA 3000
ATGAGCATTGATGACATTAGAAGAATTGGAGTCATACTTATTGGACACCAGAGACGAATA 3060
GTCAGCAGCATACAGACTTTACGTTTACACATGATGCACATACAGGAGAAGGGATTTCAT 3120
GTATGAAAGTACCACAAGCACCTGTGTTTTGTGCCTCAGCATTTCTAAAATGAACGATAT 3180
CCTCTCTACTACTCTCTCTTCTGATTCTCCAAACATCACTTCACAAACTGCAGTCTTCTG 3240
TTCAGACTATAGGCACACACCTTATGTTTATGCTTCCAACCAGGATTTTAAARTCATGCT 3300
ACATAAATCCGTTCTGAATAACCTGCAACTAAAACCCTGGCCCACTGCAGATTATTGCTA 3360
CGCAATGCAACAGCTTTAAAACCTATCTAGGCCCATGAATGGAAAACAAATCCAAATCCG 3420
ATCCTTGAΛAAGCAAAGGCTCTAAAGAAGCTCTTCAGARGAGACGGTAAAGAATGAATTC 3480
TTTTACTTATCACCCAACCACATTTCTTA^RAATGTGTTTTGGTGTCTTTTCCTACCAAA 3540
TTTCTGCTCTACAAGGCAGTCAGTTAAATCTCTCATTTCATAATTTTCACTGTGATAGAT 3600 CCTTGCTCTCTCCTCTTTTAATAAATTTAATAAAACTTTAA
The disclosed NOV6a nucleic acid sequence, has 3028 of 3367 bases (89%) identical to a gb:GENBANK-ID:MMU58332|acc:U58332.1 mRNA from Mus musculus (Mus musculus receptor tyrosine kinase mRNA, complete eds). The EphA6/ehk-2 disclosed in this invention maps to chromosome 3
A disclosed NOV6a polypeptide (SEQ ID NO:20) encoded by SEQ ID NO:19 is 1035 amino acid residues and is presented using the one-letter amino acid code in Table 6B. Signal P, Psort and/or Hydropathy results predict that NOV6a appears to be a Type la membrane protein, contains a signal peptide, and is likely to be localized in the plasma membrane with a certainty of 0.4600. In other embodiments, NOV6a is also likely to be localized to the endoplasmic reticulum with a certainty of 0.1000, or outside the cell with a certainty of 0.1000. The most probable cleavage site is between positions 22 and 23: residues LTA-WT. Table 6B.
NON6a Polypeptide
SEQ ID ΝO;20
MGGCEVREFLLQFGFF PLLTAWTGDCSHVSΝQWLLDTSTVMGE GWKTYPLΝGWDAIT 60
E DEH RPIHTYQVCΝVMEPMQΝΝ RTΝWISRDAAQKIYVEMKFTLRDCΝSIP VGTC 120
KETFTLYYIESDESHGTKFKPSQYIKIDTIAADESFTQMDLGDRILKLΝTEIREVGPIER 180
KGFYLAFQDIGACIAVSVRVFYKKCPFTVRΝDAMFPDTIPRVDSSSLVEVRGSCVKSAE 240
ERDTPKLYCGADGD VP GRCICSTGYEEIEGSCHACRPGFYKAFAGΝT CSKCPPHSL 300
TYMEATSVCQCEKGYFRAEKDPPSMACTRPPSAPRWFΝIΝETALILEWSPPSDTGGRK 360
DLTYSVICKKCGLDTSQCEDCGGGLRFIPRHTGLIΝΝSVIVLDFVSHVΝYTFEIEAMΝGV 420
SELSFSPKPFTAITVTTDQDAPSLIGWR DWASQΝSIALS QAPAFSΝGAILDYEIKYY 480
EKEHEQLTYSSTRSKAPSVIITGLKPATKYVFHIRVRTATGYSGYSQKFEFETGDETSDM 540
AAEQGQILVIATAA.VGGFTLVILTLFFLITGRCQWYI AMKSEEKRRΝHLQΝGHI.RFP 600
GIKTYIDPDTYEDPSLAVHEFA EIDPSRIRIERVIGAGEFGEVCSGRLKTPGKREIPVA 660
IKTLKGGHMDRQRRDFLREASIMGQFDHPNIIRLEGWTKRSFPAIGVEAFCPSFLRAGF 720
LNSIQAPHPVPGGGSLPPRIPAGRPVMIWEYMENGSLDSFLRHDGHFTVIQ VGMLRG 780
IASG KYLSDMGYVHRDLAARNILVNSNLVCKVSDFG SRV EDDPEAAYTTTGGKIPIR 840 TAPEAIAYRKFSSASDA SYGIVM EVMSYGERPY EMSNQDVI SIEEGYRLPAPMGC 900
PASLHQMLHC QKERNHRPKFTDIVSFLDKLIRNPSALHTVEDILVMPESPGEVPEYP 960
LFVTVGDW DSIKMGQYKN FVAAGFTTFDLISRMSIDDIRRIGVILIGHQRRIVSSIQT 1020 LR HMMHIQEKGFHV
The disclosed NOV6a amino acid sequence has 1008 of 1035 amino acid residues (97%) identical to, and 1021 of 1035 amino acid residues (98%) similar to, the 1035 amino acid residue ρtnr:SWISSNEW-ACC:Q62413 protein from Mus musculus (Mouse) (EPHRIN TYPE-A RECEPTOR 6 PRECURSOR (EC 2.7.1.112) (TYROSINE-PROTEIN KINASE RECEPTOR EHK-2) (EPH HOMOLOGY KINASE-2)).
NOV6a is expressed at least in lung, testis, and B-cells, brain, ear, ovary, thymus, and spleen.
NOV6b
A disclosed NOV6b nucleic acid of 3692 nucleotides identified as SEQ ID NO:21 (also referred to as CG55704-03) encoding an EphA6/ehk-2-like protein is shown in Table 6C. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 19-21 and ending with a TGA codon at nucleotides 3124-3126. Putative untranslated regions are found upstream from the initiation codon and downstream from the termination codon, and are indicated by underline.
Table 6C.
NOV6b Polynucleotide
SEQ ID NO:21
AGAGAACCAGCGAGAGCCATGGGGGGCTGCGAAGTCCGGGAATTTCTTTTGCAATTTGGT 60
TTCTTCTTGCCTCTGCTGACAGCGTGGCCAGGCGACTGCAGTCACGTCTCCAACAACCAA 120
GTTGTGTTGCTTGATACAACAACTGTACTGGGAGAGCTAGGATGGAAAACATATCCATTA 180
AA.TGGGTGGGATGCCATCACTGAAATGGATGAACATAATAGGCCCATTCACACATACCAG 240
GTATGTAATGTAATGGAACCAAACCAAAACAACTGGCTTCGTACAAACTGGATCTCCCGT 300
GATGCAGCTCAGAAAATTTATGTGGAAATGAAΑTTCACACTAAGGGATTGTAACAGCATC 360
CCATGGGTCTTGGGGACTTGCAAAGAAACATTTAATCTGTTTTATATGGAATCAGATGAG 420
TCCCACGGAATTAAATTCAAGCCAAACCAGTATACAAAGATCGACACAATTGCTGCTGAT 480 GAGAGTTTTACCCAGATGGATTTGGGTGATCGCATCCTCAAACTCAACACTGAAATTCGT 540 GAGGTGGGGCCTATAGAAAGGAAAGGATTTTATCTGGCTTTTCAAGACATTGGGGCGTGC 600 ATTGCCCTGGTTTCAGTCCGTGTTTTCTACAAGAAATGCCCCTTCACTGTTCGTAACTTG 660 GCCATGTTTCCTGATACCATTCCAAGGGTTGATTCCTCCTCTTTGGTTGAAGTACGGGGT 720 TCTTGTGTGAAGAGTGCTGAAGAGCGTGACACTCCTAAACTGTATTGTGGGGCTGATGGA 780 GATTGGCTGGTTCCTCTTGGAAGGTGCATCTGCAGTACAGGATATGAAGAAATTGAGGGT 840 TCTTGCCATGCTTGCAGACCAGGATTCTATAAAGCTTTTGCTGGGAACACAAAATGTTCT 900 AAATGTCCTCCACACAGTTTAACATACATGGAAGCAACTTCTGTCTGTCAGTGTGAAAAG 960 GGTTATTTCCGAGCTGAAAAAGACCCACCTTCTATGGςATGTACCAGGCCACCTTCAGCT 1020 CCTAGGAATGTGGTTTTTAACATCAATGAAACAGCCCTTATTTTGGAATGGAGCCCACCA 1080 AGTGACACAGGAGGGAGAAAAGATCTCACATACAGTGTAATCTGTAAGAAATGTGGCTTA 1140 GACACCAGCCAGTGTGAGGACTGTGGTGGAGGACTCCGCTTCATCCCAAGACATACAGGC 1200 CTGATCAACAATTCCGTGATAGTACTTGACTTTGTGTCTCACGTGAATTACACCTTTGAA 1260 ATAGAAGCAATGAATGGAGTTTCTGAGTTGAGTTTTTCTCCCAAGCCATTCACAGCTATT 1320 ACAGTGACCACGGATCAAGATGCACCTTCCCTGATAGGTGTGGTAAGGAAGGACTGGGCA 1380 TCCCAAAATAGCATTGCCCTATCATGGCAAGCACCTGCTTTTTCCAATGGAGCCATTCTG 1440 GACTACGAGATCAAGTACTATGAGAAAGTCTACCCACGGATAGCGCCGGCATTTTGGCAC 1500 TACCTGCGGGTAGAAGAACATGAGCAGCTGACCTACTCTTCCACAAGGTCCAAAGCCCCC 1560 AGTGTCATCATCACAGGTCTTAAGCCAGCCACCAAATATGTATTTCACATCCGAGTGAGA 1620 ACTGCGACAGGATACAGTGGCTACAGTCAGAAATTTGAATTTGAAACAGGAGATGAAACT 1680 TCTGACATGGCAGCAGAACAAGGACAGATTCTCGTGATAGCCACCGCCGCTGTTGGCGGA 1740 TTCACTCTCCTCGTCATCCTCACTTTATTCTTCTTGATCACTGGGAGATGTCAGTGGTAC 1800 ATAAAAGCCAAGATGAAGTCAGAAGAGAAGAGAAGAAACCACTTACAGAATGGGCATTTG 1860 CGCTTCCCGGGAATTAAAACTTACATTGATCCAGATACATATGAAGACCCATCCCTAGCA 1920 GTCCATGAATTTGCAAAGGAGATTGATCCCTCAAGAATTCGTATTGAGAGAGTCATTGGG 1980 GCAGGTGAATTTGGAGAAGTCTGTAGTGGGCGTTTGAAGACACCAGGGAAAAGAGAGATC 2040 CCAGTTGCCATTAAAACTTTGAAAGGTGGCCACATGGATCGGCAAAGAAGAGATTTTCTA 2100 AGAGAAGCTAGTATCATGGGCCAGTTTGACCATCCAAACATCATTCGCCTAGAAGGGGTT 2160 GTCACCAAAAGATCCTTCCCGGCCATTGGGGTGGAGGCGTTTTGCCCCAGCTTCCTGAGG 2220 GCAGGGTTTTTAAATAGCATCCAGGCCCCGCATCCAGTGCCAGGGGGAGGATCTTTGCCC 2280 CCCAGGATTCCTGCTGGCAGACCAGTAATGATTGTGGTGGAATATATGGAGAATGGATCC 2340 CTAGACTCCTTTTTGCGGAAGCATGATGGCCACTTCACAGTCATCCAGTTGGTCGGAATG 2400 CTCCGAGGCATTGCATCAGGCATGAAGTATCTTTCTGATATGGGTTATGTTCATCGAGAC 2460 CTAGCGGCTCGGAATATACTGGTCAATAGCAACTTAGTATGCAAAGTTTCTGATTTTGGT 2520 CTCTCCAGAGTGCTGGAAGATGATCCAGAAGCTGCTTATACAACAACTGGTGGAAAAATC 2580 CCCATAAGGTGGACAGCCCCAGAAGCCATCGCCTACAGAAAA.TTCTCCTCAGCAAGCGAT 2640 GCATGGAGCTATGGCATTGTCATGTGGGAGGTCATGTCCTATGGAGAGAGACCTTATTGG 2700 GAAATGTCTAACCAAGATGTCATTCTGTCCATTGAAGAAGGGTACAGACTTCCAGCTCCC 2760 ATGGGCTGTCCAGCATCTCTACACCAGCTGATGCTCCACTGCTGGCAGAAGGAGAGAAAT 2820 CACAGACCAAAATTTACTGACATTGTCAGCTTCCTTGACAAA.CTGATCCGAAATCCCAGT 2880 GCCCTTCACACCCTGGTGGAGGACATCCTTGTAATGCCAGAGTCCCCTGGTGAAGTTCCG 2940 GAATATCCTTTGTTTGTCACAGTTGGTGACTGGCTAGATTCTATAAAGATGGGGCAATAC 3000 AAGAATAACTTCGTGGCAGCAGGGTTTACAACATTTGACCTGATTTCAAGAATGAGCATT 3060 GATGACATTAGAA.GAATTGGAGTCATACTTATTGGACACCAGAGACGAATAGTCAGCAGC 3120 ATACAGACTTTACGTTTACACATGATGCACATACAGGAGAAGGGATTTCATGTATGAAAG 3180 TACCACAAGCACCTGTGTTTTGTGCCTCAGCATTTCTAAAATGAACGATATCCTCTCTAC 3240 TACTCTCTCTTCTGATTCTCCAAACATGACTTCACARACTGCAGTCTTCTGTTCAGACTA 3300 TAGGCACACACCTTATGTTTATGCTTCCAACCAGGATTTTAAAATCATGCTACATAAATC 3360 CGTTCTGAATAACCTGCAACTAAAACCCTGGCCCACTGCAGATTATTGCTACGCAATGCA 3420
ACAGCTTTAAAACCTATCTAGGCCCATGAΛTGGAAAACAAATCCAAATCCGATCCTTGAA 3480 AAGCAAAGGCTCTAAAGAAGCTCTTCAGAAGAGACGGTAARGAATGAATTCTTTTACTTA 3540 TCACCCAACCACATTTCTTAAAAATGTGTTTTGGTGTCTTTTCCTACCAAATTTCTGCTC 3600 TACAAGGCAGTCAGTTAAATCTCTCATTTCATAATTTTCACTGTGATAGATCCTTGCTCT 3660 CTCCTCTTTTAATAAATTTAATAAAACTTTAA
The disclosed NOV6b nucleic acid sequence has 3028 of 3367 bases (89%) identical to a gb:GENBANK-ID:MMU58332|acc:U58332.1 mRNA from Mus musculus (Mus musculus receptor tyrosine kinase mRNA, complete eds)
A disclosed NOV6b polypeptide (SEQ ID NO:22) encoded by SEQ ID NO:21 is 1035 amino acid residues and is presented using the one-letter amino acid code in Table 6D. Signal
P, Psort and/or Hydropathy results predict that NOV6a appears to be a Type la membrane protein, contains a signal peptide, and is likely to be localized in the plasma membrane with a certainty of 0.4600. In other embodiments, NOV6b is also likely to be localized to the endoplasmic reticulum with a certainty of 0.1000, or outside the cell with a certainty of 0.1000. The most probable cleavage site is between positions 22 and 23: residues LTA-WP.
Table 6D.
NOV6b Polypeptide
SEQ ID NO:22
MGGCEVREFLLQFGFFLPLLTA PGDCSHVSNNQWLLDTTTVLGELG KTYPLNG DAI 60
TEMDEH RPIHTYQVCNVMEPNQNNWLRTN ISRDAAQKIYVEMKFTLRDCNSIP VLGT 120
CKETFNLFYMESDESHGIKFKPNQYTKIDTIAADESFTQMDLGDRI KLNTEIREVGPIE 180
RKGFYAFQDIGACIAVSVRVFYKKCPFTVRNLAMFPDTIPRVDSSS VEVRGSCVKSA 240
EERDTPKLYCGADGD VPLGRCICSTGYEEIEGSCHACRPGFYKAFAGNTKCSKCPPHS 300 TYMEATSVCQCEKGYFRAEKDPPSMACTRPPSAPRNWFNINETALILE SPPSDTGGR 360 DLTYSVICKKCGLDTSQCEDCGGGLRFIPRHTGLINNSVIVLDFVSHVWYTFEIEAMNG 420
VSELSFSPKPFTAITVTTDQDAPSLIGWR DASQNSIALSWQAPAFSNGAILDYEIKY 480
YEKVYPRIAPAF HYLRVEEHEQLTYSSTRSKAPSVIITG KPATKYVFHIRVRTATGYS 540
GYSQKFEFETGDETSDMAAEQGQILVIATAAVGGFT VILTLFF ITGRCQWYIKAKMK 600
SEEKRR H QNGHLRFPGIKTYIDPDTYEDPSLAVHEFAKEIDPSRIRIERVIGAGEFGE 660
VCSGR KTPGKREIPVAIKT KGGHMDRQRRDFLREASIMGQFDHPNIIRLEGWTKRSF 720
PAIGVEAFCPSFLRAGFLNSIQAPHPVPGGGSLPPRIPAGRPVMIWEYMENGS DSFLR 780
KHDGHFTVIQLVGMLRGIASGMKYLSDMGYVHRDLAARNILV SNLVCKVSDFG SRV E 840
DDPEAaYTTTGG IPIRWTAPEAIAYRKFSSASDAWSYGIVM EVMSYGERPY E SNQD 900
VI SIEEGYRLPAPMGCPASLHQLM HC QKERNHRPKFTDIVSFLDK IRNPSAHTLV 960
EDILVMPESPGEVPEYPLFVTVGD LDSI GQYKNNFVAAGFTTFDLISRMSIDDIRRI 1020 GVILIGHQRRIVSSIQT RLHMMHIQEKGFHV
The disclosed NO V6b amino acid sequence has 1008 of 1035 amino acid residues (91%) identical to, and 1021 of 1035 amino acid residues (98%) similar to, the 1035 amino acid residue ptnr:SWISSNEW-ACC:Q62413 protein from Mus musculus (Mouse) (EPHRIN TYPE-A RECEPTOR 6 PRECURSOR (EC 2.7.1.112) (TYROSINE-PROTEIN KINASE RECEPTOR EHK-2) (EPH HOMOLOGY KINASE-2)).
NOV6b is expressed at least in lung, testis, and B-cells, brain, ear, ovary, thymus, and spleen.
Table 6E. BLAST results for NOV6
Gene Index/ Identifier
Figure imgf000083_0001
Figure imgf000084_0001
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 6F.
Table 6F Information for the ClustalW proteins
1) NOV6a (SEQ ID NO:19)
2) NOV6b (SEQ ID NO: 21)
3) gi 4885211 (SEQ ID NO: 59)
4) gi 125340 | (SEQ ID NO: 60)
5) gi 8134439 (SEQ ID NO: 61)
6) i 1079403 (SEQ ID NO: 62)
7) gi 1706629 (SEQ ID NO: 63)
10 20 30 40 50
NOV6a MGGCEVREFLLQFGFFLPBJT
Novβb MGGCEVREFLLQFGFFLPSEJT gi I 48852111 MDCQLSILLL SCS|v|jjD gi 1125340 I MALDCLJSJF gi] 8134439| MAGIVHGI FCGLFGJJCW gi] 1079403 I MPGPERTMGPLWFCCLPjJA gi]l706629] MRGSGPRGAGRRRTQGRGGGGDTPRVPASLAGCYSAPLKGPL TCLLJJCA
60 70 80 90 100
NOV6a S^W GDC,§H gK-ySvJ5^S^MS3H TYgLNS-^AiTE^HMRgE|
Figure imgf000085_0001
gi I 48852111 ^FGE IPQP^N-B2N2Es^ϊQS3HIS'^SHS-BffiI§G 3 __3 gi ( 125340 j L ASAVAMEE yiJ^RπΑTaEπMTAI^.sS-ffifeSG!B5i^ l. )ή gi I 8134439 | &VT SRIYBka-ESτflTOsRis^ ^<eiillAsB EBGBg-SIMi3-IK Ti8H gi 11079403 | LIiP-LLj&AVEE τf^ST^TAl5ffleB!lWHl3psS-IB3^gGYl8 j NτH gi 11706629 | toRT ^SP^-BSNiffiS iyi^HllAFSra^-EI^GE^S^IYAJB
110 120 130 140 150
NOV6a H A EEII iiSSWEES^
Novβ
Figure imgf000085_0002
Figure imgf000085_0003
160 170 180 190 200
Novεa MføMdτflraιl5ESlESHGτl8 gκpsBBfetMι-.ιlwlιAMιl3felSHκ.dle.»lilιJ
NOVSb SHΞJSFΞMHESHGIE g^PNliffiTia^liili^MΛafetali'B deMaTl gi|488521l| It.Malnlalitfc'gMiaEBInnwnvtS SRBHBIli'Tia^liiliJaii. lafelalnltlMnH.tra
Figure imgf000085_0004
210 220 230 240 250
NOV6a WttM÷MH MΛTSVRVFYKKCP
Figure imgf000085_0005
gi|488521l|
Figure imgf000085_0006
gi|8134439|
Figure imgf000085_0007
gι]l706629| DEBt1s.κ|23EΞ^S^'SEIΞ-^-iSi'ΞS-S-isvΞRHϊ-'
260 270 280 290 300
NOV6a
N0V6b _^iPRvj^s[^^2__^_ERS?S^Ti3-E^GA!EDE2B_-lLSREIS gi|488521l| S^vp-^QSE^[S^^Irøs^Ei2pΞ-RMEE3 B__ϊEκEΞS gi|l2534θ| ^i®aGs^ι.^EIft&Bϊpi^3vB^3IiS!&G^SM33E^τS gi| 8134439 I S^ιτGsS^sSS^^SD^sSκ!SΞ-l_i["SSGA!ϊΞfflSΞENEIS
Figure imgf000085_0008
gi|l706629| 310 320 330 340 350
NOV6a STUBS- -[^IEGsSH^^PgFYΪ^F Gfi I^ajJ3|3^LJ3 MgA^'vgQSEi oveb sτBSsi- - iEGsSHgs^pFAG τκES331ΞLΞγMi3A^'vSQB^
Figure imgf000086_0001
gi|8134439| NAJjjjff- -[HNGGE^^ ^Y^LST AA^JSa5iasj ALR|GiS5!sgτgl gi 11079403 I RPHsV§NGT g^GSPSg ϊi^SQGpEG|SvHS3lN§J3τsEG ϊ SvgR gi|l706629| KABWSI- - KNG g gl?PSF Es PH^QτBSS_a_3_ϊ_SjγSHE[ ^!s5JsS g^
360 370 380 390 400
Novsa
Novsb
Figure imgf000086_0002
Figure imgf000086_0003
gi| 125340] τ!3ff^DF.^E'gE!ros t^ .5ϊπiISEι^M^
Figure imgf000086_0004
gi|l079403| Nlegyj^AB^fflpraTllifckld Ai^ΞyM^ gi|i706629| KDBT^_felira Jik%lιι.;lilikkl MA^
410 420 430 440 450
N0V6a ^^3S2^l|TSQgED^GG^i I^ ^I NSEl^ J3F l]liiJS3 N0V6b Ms¥tB13^ τsoBEDlffl Gfcιi^ϊI5fe^ gi|488521l| *IWIj52^|SjIKQgEPgsPNERJlS^FS3TNTT2TVTSL3E35BEii g I 8134439 I p^ιJ^SβΦLT^spΞSGBI^sS5 iiS!κτ'2sϊ?s|SLQSjSSS__i gi|l079403| ffi^tTOslMsGRAfaτRl8^ι^oi3l3^ι^TEPRZγSsBlι,ϊ.a!πnraJ3
Figure imgf000086_0005
460 470 480 490 500
NOV6a ^E^J^^^LgFSPKP^T^Tfj^gD2Sl.S|G\A?RKDAaSS
NOV6b [^^ιβE4^FSP pyτi&?*T!^i-^ DSEL?G^^^w s^JsiEE_l gi|488521l| ^ft4J^EB^sppRi8,^Bsϊβ^SSpBLT lt)RI RiS3siEEl gi|l2534θ| jJ^HgsκgPFPP HJSfNliSJ^SEτEPIP42VSAt:E'MRS3τSS gi I 8134439 I f^l|^EKQaPEQD^λ^2τl!βϊ^-SEτEτQiflPKEIΪR1^s2lSI
Figure imgf000086_0006
510 520 530 540 550
NOV6a
Figure imgf000086_0007
NOV6b QAg FSJJgAJHjSSli^H PRIAPAF H LRVEEHEQL'rB^STi^ gi 148852111 QI@BHPH∑0_SΞ3 _S_Ϊ5_3Q EQ$τsg]pLfe2eJG gi 1125340 I PθB_OPWlimny«aiI CT83gR HN[F SSMAS [QT
Figure imgf000086_0008
gi 110794031
Figure imgf000086_0009
gi 11706629 I QEJigRPHlj- fflϊJ&ffi DQJfrsggilc KE
560 570 580 590 600 NOVδa
NOV6b pfe iøTSjg^K^lJ^iS ST-Ss^QSi&iJ∞STSDMSAtS1^ gi]488521l] τκfapsRBtafe^ιg^oιi; W ^τιfa!33SHEπsbsFsιlc^ssS gi|l2534θ| K^R0DE^^Gim^^QJ^Bv^^KF|GiSMCJ^3rιφDD K^E gEg gi I 8134439 I R^0KS^ SG{SJ^^S^SEE^1GPS_EITΞNTVPSP1M:I:G^GTS
Figure imgf000087_0001
610 620 630 640 650
Figure imgf000087_0002
gi 1125340 I
Figure imgf000087_0003
--jjJKR
Figure imgf000087_0004
660 670 680 690 700
■■■■i....ι....ι....|....ι-...)....|....|....|....ι
N0V6a wHlBBB KSJBfgSRRNJlLQ^SliRFi^ilSBniHDlk'atiljls'
N0V6b ^iii^^KS l-^' NlSl-iOi^fiRFBSil ^ JJD ^tlJSL
Figure imgf000087_0005
gi 1125340 |
Figure imgf000087_0006
Figure imgf000087_0007
gi ] 1706629 I GBsj^QDPEj ^-M2FH|^Ϊ i^ ^SiJ@Hj3S 3Jrg^Hjg3Gg
710 720 730 740 750
.... I ....|....|....|....|....|....1....|.... I .... I
Figure imgf000087_0008
gi ] 1079403 ] jBτt^-πir-BSk^J«)JW^^aWM-tWHHBfe-l^i^
Figure imgf000087_0009
760 770 780 790 800
.. ■■|....i-...|....|....|.... I .... I .... I .... I .... I
NOVδa [aB|R[5fc.!^i4l!ite.«.ji ^i_-VjRiJiale i».iJRSFPATGVEAFCPSF RARFLNSIO
N0V6b [^R[attf^iJiiteltl3l»lii8-i-i-VjR)Jial ^iil.feSFPATGVEAFCPSFLRAGFLNSIO
Figure imgf000087_0010
gi I 81344391 |5tl.qliifelιJι!ilel».3t».it8-i JHtιι«WAtiMa
Figure imgf000087_0011
gi] 1706629] άάAUMMAάs akmEBasa
810 820 830 840 850
..| ....|-...l ....| ....| ....| ----l ----l ----l --- - l
N0V6a APHPVPGGGSLPPRIPAGJ)ΪΪBBBOTi3ltiilali_efet*ιιM3lΛ;iit NOVδb APH GGGSIiPP IPAG^SBiHvΛ^tbiMdsMiibtatfctiltJaaH gi]488521l| sSISBiti^a^alMel^fclbMid^AiTiaiMiWM
Figure imgf000088_0001
gi]8l34439| c^^i^lff^ιti^W^*»lai3Kπ a«|R)!iiιtf*MJiιa gi|l079403] ssl^8tfa^ l!t^i^*t-H^δκiialrιla-i^<»>->,*-
Figure imgf000088_0002
860 870 880 890 900
■■•■I -...
NOV6a leWH.Je A'SB
NOV6b gi|488521l| MKYLSDMKYVHRD AARNILfflNSNLVCKVSDFGLSRVLEDD gi|l2534θ| 5iwιg^iiaBiwaa^>τ«Biii8.isιιw»ιaw5.H3feπMaγ>ιαBi; gi|8134439| gi|l079403| s tjJ(:J>)*iM Λi**l,sωSιA««i:»M.-.BΪeifc.;JFIΛs)βl gi|l706629]
910 920 930 940 950
NOV6a
NOVδb gi|488521l| gi|l2534θ| τsDP {fflffsr.Me.^i^f^iιι^AJ^A*AtdM^ι^^AM^> i<lli *^Fgg gi|8134439|
Figure imgf000088_0003
gι]l706629| t3-I^Λ taιflR-f^e.i^4:i-.Mi.ΛJ^roi:it^^J^Λ*itHl i*il^t-4 ?gπ^
960 970 980 990 1000
NOVδa lι^^rfriMd^J lM^ J ^feh_l8ll-jg!j^1^^dd^alτ
Figure imgf000088_0004
gi|488521l|
Figure imgf000088_0005
gi I 8134439 I i8ig^κCTEratetfdΛjpiJilJ Jτb i-»ilιi.*ιttlι.Mi4tR|griJ5.i43lG gι|l079403|
Figure imgf000088_0006
1010 1020 1030 1040 1050
NOV6a DJggFJJ3SS_ΞS3S^!HT ιVEDIL MPESPGl P--^PLi|vJ2GSJj^§ NOVδb D^^FJEΞSEESS^Tft EDIL MPESPG^V - -E PL[| 5EGS33ϊi3a gi]488521l| 0πS^τH^ft^tfel3^lSτSA feRP§NLPlfc0S ΩISTi3RRτ !ϊSi!tKG
Figure imgf000088_0007
gi I 8134439 I Q^MJj»j^E3i^_ERTGLENSRT^TA3l±i SSPS^SQVAS2IβΪQa
Figure imgf000088_0008
gi|l706629] DjftMjJ3S53Ss^I^ NASgRV TljjAgHGS GSG ?R,s3G!E2J S
1060 1070 1080 1090 1100
o δa ιl^S 3VA ^FSL sRM^ιp]3iSιS:ι:^:ι:SlE?^ϊ^Ξ3^τ?i
NOVδb fl]^t EN^A^^FSLΪSi φSiiSl^ltilHκE ^ϊ ϊ|
Figure imgf000088_0009
Figure imgf000089_0001
1110
N0V6 ^LHMMHIQEKGFHV
N0V6b gliHgMHIQEKGFHV gi|488521l| ETQSKNGPVPV gi]l25340| gVQiWNQSPSVMA-- gi|8134439| ΞTQ JQQMQGRMVPV gi 11079403 I EJAQSΪNQIQSVEV- - gi | l706629 |
Tables 6G lists the domain description from DOMAIN analysis results against NON6. This indicates that the ΝON6 sequence has properties similar to those of other proteins known to contain this domain.
Table 6G. Domain Analysis of ΝON6
gnl|Pfam|pfam01404, EPH_lbd, Ephrin receptor ligand binding domain. The Eph receptors, which bind to ephrins pfam00812 are a large family of receptor tyrosine kinases. This family represents the amino terminal domain which binds the ephrin ligand.
CD-Length = 174 residues, 100.0% aligned Score = 345 bits (886), Expect = 6e-96
Query : 33 QVVLLDTSTVMGE GWKTYPLΝGWDAITEMDEHΝRPIHTYQVCΝVMEPΝQlrøYILRTΝWIS 92 +V LLDT+T GELGW TYP GW+ ++ +DE+ΝRPI TYQVCNVMEPNQNN RTN I
Sbjct: 1 EVT LDTTTATGE GWLTYPPGGWEEVSGLDENNRPIRTYQVCNVMEPNQNNWLRTNWIP 60 o o σ
Query: 93 RDAAQKIYVEMKFT RDCNSIPWVLGTCKETFTLYYIESDESHGTKFKPSQYIKIDTIAA 152 R AQ++YVE+KFT+RDCNS+P VLGTCKETF YY ESDE G ++ +QY K+DTIAA
Sbjct: 61 RRGAQRVYVE KFTVRDCNS PGVLGTCKETFN YYYESDEDVGPAWRΞNQYTKVDTIAA 120
Query: 153 DESFTQMD GDRI KLNTEIREVGPIERKGFY AFQDIGACIA VSVRVFY KC 206 DESFTQ+DLGDR++KLNTE+R VGP+ +KGFYLAFQD+GAC+A VSVRVFYKKC
Sbjct: 121 DESFTQVDLGDRVMK NTEVRSVGPLSKKGFY AFQDVGACMALVSVRVFYKKC 174
The gene of invention is an ortholog of mouse EphA6 (also known as m-ehk2) which belongs to the superfamily of receptor tyrosine kinases, which constitute the largest family of oncogenes. This family includes prominent growth factor receptors such as those for epidermal growth factor, platelet-derived growth factor etc. Members of this superfamily influence cell shape, mobility, differentiation and proliferation.
Within this superfamily, the Ephrin (Eph) receptors constitute the largest subfamily. Eph receptors and their ligands, ephrins, are known to be involved in several normal developmental processes, including formation of segmented structures, axon guidance, cell adhesion and development of vasculature. Ephrin receptors are classified into two main subtypes: EpbA receptors bind to GPI-anchored ephrin- A ligands, while EphB receptors bind to ephrin-B proteins that have a transmembrane and cytoplasmic domain. The EphA6 receptor is highly expressed in the mouse brain and inner ear, including the cochlea. This receptor is also differentially expressed relative to the other ephrin receptors in certain regions of the primate neocortex during development. In addition, it is found in the developing retina and optic tectum in the chicken. It may, therefore, be involved in the development of these structures. It shows the presence of conserved ephrin and protein kinase domains, similar to the protein of invention. The protein of invention, therefore, may be involved in the development and/or dysgenesis of a variety of tissues (see, Maisonpierre PC, et al, Oncogene 1993 Dec;8(12):3277-88); Lee AM, et al., DNA Cell Biol 1996 Oct;15(10):817-25; Dodelet VC, et al, Oncogene 2000 Nov 20;19(49):5614-9; Mellitzer G, et al., Curr Opin Neurobiol 2000 Jun;10(3):400-8; Holder N, et al., Development 1999 May;126(10):2033-44; Matsunaga T, et al., Eur J Neurosci 2000 May;12(5):1599-616; Donoghue MJ, et al., J Neurosci 1999 Jul 15;19(14):5967-79; Connor RJ, et al., Dev Biol 1998 Jan l;193(l):21-35, incorporated by reference).
The ephrin domain (IPR001090) is a feature of ephrins and ephrin receptors. IPR000719 is a catalytic domain characteristic of eukaryotic protein kinases. hi the N-terminal extremity of the catalytic domain there is a glycine-rich stretch of residues in the vicinity of a lysine residue, which has been shown to be involved in ATP binding. In the central part of the catalytic domain there is a conserved aspartic acid residue which is important for the catalytic activity of the enzyme. The fibronectin type III repeat region (IPR001777) is an approximately 100 amino acid domain, different tandem repeats of which contain binding sites for DNA, heparin and the cell surface. The superfamily of sequences believed to contain Fnlll repeats represents 45 different families, the majority of which are involved in cell surface binding in some manner, or are receptor protein tyrosine kinases, or cytokine receptors. The sterile alpha motif (SAM) domain (IPR001660) is a putative protein interaction module present in a wide variety of proteins involved in many biological processes. SAM domains have been shown to homo-and hetero-oligomerize, mediating specific protein-protein interactions. This indicates that the sequence of the invention has properties similar to those of other proteins known to contain these domains and similar to the properties of these domains.
The disclosed NON6a nucleic acid of the invention encoding an EphA6-like protein includes the nucleic acid whose sequence is provided in Table 6A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases maybe changed from the corresponding base shown in Table 6A while still encoding a protein that maintains its EphA6-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant protein, up to about 3 percent of the residues may be so changed.
The disclosed ΝOV6b protein of the invention includes the EphA6-like protein whose sequence is provided in Table 6D. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 6D while still encoding a protein that maintains its EphA6-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant nucleic acids, and their complements, up to about 11 percent of the bases may be so changed. In the mutant or variant protein, up to about 3 percent of the residues may be so changed.
The above defined information for this invention suggests that EphA6-like proteins (NOV6) may function as a member of an Ephrin receptor family. Therefore, the NOV6 nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The nucleic acids and proteins of NOV6 are useful in, for example, treatment of patients suffering from: hemophilia, hypercoagulation, idiopathic thrombocytopenic purpura, immunodeficiencies, graft versus host disease, systemic lupus erythematosus, autoimmune disease, asthma, emphysema, scleroderma, allergy, ARDS, fertility, cancer, developmental disorders and other diseases, disorders and conditions of the like.
The novel NON6 nucleic acid encoding ΝON6 protein, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. These materials are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods.
ΝON6 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno specifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-ΝOVX Antibodies" section below. The disclosed ΝON6a protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, contemplated ΝON6a epitope is from about amino acids 50 to 125. In other embodiments, ΝON6a epitope is from about amino acids 175 to 200, from about amino acids 210 to 400, or from about amino acids 420 to 675, from about 700 to 720, from about 760 to 780, from about 795 to 805, and from about 806 to 950. The disclosed ΝON6b protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, contemplated ΝON6b epitope is from about amino acids 50 to 125. In other embodiments, ΝON6b epitope is from about amino acids 175 to 200, from about amino acids 210 to 400, or from about amino acids 420 to 675, from about 720 to 740, from about 770 to 790, from about 795 to 805, and from about 806 to 950. This novel protein also has value in development of powerful assay system for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
ΝOV7
A disclosed NOV7 nucleic acid of 1607 nucleotides identified as SEQ ID NO:23 (also referred to as CG94323538) encoding a glucose transporter-like protein is shown in Table 7A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 68-70 and ending with a TAG codon at nucleotides 1517-1519.
Table 7A. NOV7 Polynucleotide SEQ ID NO:23
TGGGTTTAACTGTGTCTTATAGGTGTTAGCAGAAAAft.CCTCTCTGTACAATGACAAGTGG 60
CCACTGAGAACACTTTCTCATTTCTCATGAACTGCCCAATATTCTTAGCTGTGGATGGGG 120
CAATGTTTTCCAGGTCTTCAA3TCATTTTACAACGAAACCTACTTTGAGCGACACGCAAC 180
ATTCATGGACGGGAAGCTCATGCTGCTTCTATGGTCTTGCACCGTCTCCATGTTTCCTCT 2 0
GGGCGGCCTGTTGGGGTCATTGCTCGTGGGCCTGCTGGTTGATAGCTGCGGCAGAAAGGG 300
GACCCTGCTGATCAACAACATCTTTGCCATCATCCCCGCCATCCTGATGGGAGTCAGCAA 360
AGTGGCCAAGGCTTTTGAGCTGATCGTCTTTTCCCGAGTGGTGCTGGGAGTCTGTGCAGG 4 0
TATCTCCTACAGCGCCCTTCCCATGTACCTGGGAGAACTGGCCCCCAAGAACCTGAGAGG 480
CATGGTGGGAACAATGACCGAGGTTTTCGTCATCGTTGGAGTCTTCCTAGCACAGATCTT 5 0
CAGCCTCCAGGCCATCTTGGGCAACCCGGCAGGCTGGCCGGTGCTTCTGGCGCTCACAGG 600
GGTGCCCGCCCTGCTGCAGCTGCTGACCCTGCCCTTCTTCCCCGAAAGCCCCCGCTACTC 660
CCTGATTCAGAAAGGAGATGAAGCCACAGCGCGGCCTCTGAGGAGGCTGAGAGGCCACAC 720
GGACATGGAGGCCGAGCTGGAGGACATGCGTGCGGAGGCCCGGGCCGAGCGCGCCGAGGG 780
CCACCTGTCTGTGCTGCACCTCTGTGCCCTGCGGTCCCTGCGCTGGCAGCTCCTCTCCAT 8 0
CATCGTGCTCATGGCCGGCCAGCAGCTGTCGGGCATCAATGCGATCAACTACTATGCGGA 900
CACCATCTACACATCTGCGGGCGTGGAGGCCGCTCACTCCCAATATGTAACGGTGGGCTC 960
TGGCGTCGTCAACATAGTGATGACCATCACCTCGGTGGTCCTTGTGGAGCGGCTGGGACG 1020
GCGGCACCTCCTGCTGGCCGGCTACGGCATCTGCGGCTCTGCCTGCCTGGTGCTGACGGT 1080
CTCTCCCCCCCCACAGAACAGGGTCCCCGAGCTGTCCTACCTCGGCATCATCTGTGTCTT 1140
TGCCTACATCGCGGGACATTCCATTGGGCCCAGTCCTGTCCCCTCGGTGGTGAGGACCGA 1200
GATCTTCCTGCAGTCCTCCCGGCGGGCAGCTTTCATGGTGGACGGGGCAGTGCACTGGCT 1260
CACCAACTTCATCATAGGCTTCCTGTTCCCATCCATCCAGGAGGCCATCGGTGCCTACAG 1320
TTTCATCATCTTTGCCGGAATCTGCCTCCTCACTGCGATTTACATCTACGTGGTTATTCC 1380
GGAGACCAAGGGCAAAACATTTGTGGAGATAAACCGCATTTTTGCCAAGAGAAACAGGGT 1440
GAAGCTTCCAGAGGAGAAAGAAGAAACCATTGATGCTGGGCCTCCCACAGCCTCTCCTGC 1500
CAAGGAAACTTCCTTTTAGTGGCCCTGCATGAAGGACGGGAGCCCATATTCAAGGCTTCC 1560 TTCTATGACAATGGGCCTCCCGGCCCCAGGCTCTGGGGAGGATAATA
The disclosed NON7 nucleic acid sequence, localized to chromosome 1, has 933 of 1328 bases (70%) identity to a gb:GEΝBAΝK-ID:HUMGLUT5|acc:M55531.1 mRNA from Homo sapiens (Human glucose transport-like 5 (GLUT5) mRNA, complete eds).
A disclosed NOV7 polypeptide (SEQ ID NO:24) encoded by SEQ ID NO:23 is 483 amino acid residues and is presented using the one-letter amino acid code in Table 7B. Signal P, Psort and/or Hydropathy results predict that NON7 has a signal peptide and is likely to be localized at the plasma membrane with a certainty of 0.6000. In other embodiments, ΝON7 is also likely to be localized to the golgi body with a certainty of 0.4000, to the endoplasmic reticulum (membrane) with a certainty of 0.3000, or the mitochondrial inner membrane with a certainty of 0.3000. The most likely cleavage site for aΝON7 peptide is between amino acids 18 and 19, at: GWG-ΝN.
Table 7B.
ΝOV7 Polypeptide
SEQ ID NO;24
EHFLISHELPNI SCGWG VFQVFKSFYNETYFERHATFMDGKLMLLLWSCTVSMFPLGG 60
L GSL VGLLVDSCGRKGTLLINNIFAIIPAILMGVSKVAKAFELIVFSRW GVCAGIS 120
YSALPMY GELAPKNLRGMVGTMTEVFVIVGVFLAQIFSLQAI GNPAG PVLLALTGVP 180
ALLQLLTLPFFPESPRYSLIQKGDEATARPLRRLRGHTDMEAELEDMRAEARAERAEGHL 240
SV HLCALRSLR QLLSIIVLMAGQQLSGINAINYYADTIYTSAGVEAAHSQYVTVGSGV 300
VNIVMTITSW VERLGRRHL LAGYGICGSAC V TVSPPPQNRVPELSYLGIICVFAY 360
IAGHSIGPSPVPSWRTEIF QSSRRAAFMVDGAVHW TNFIIGFLFPSIQEAIGAYSFI 420
I FAGICLLTAI YI YWI PETKGKTFVE INRI FAKRNRVKLPEEKEETIDAGPPTASPAKE 480 TSF
The disclosed NON7 amino acid sequence has 272 of 455 amino acid residues (59%) identical to, and 348 of 455 amino acid residues (76%) similar to, the 501 amino acid residue ptnr:SWISSPROT-ACC:P22732 protein from Homo sapiens (Human) (GLUCOSE TRANSPORTER TYPE 5, SMALL INTESTINE (FRUCTOSE TRANSPORTER)).
NON7 also has homology to the amino acid sequence shown in the BLASTP data listed in Table 7C.
Figure imgf000094_0001
Figure imgf000095_0004
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 7D.
Table 7D. Information for the ClustalW proteins
1) NOV7 (SEQ ID N0.23)
2) gi117441225 I (SEQ ID NO: 64)
3) gi|ll70105| (SEQ ID NO:65)
4) gi 113929044 I (SEQ ID NO: 66)
5) gi I 204416 I (SEQ ID NO: 67)
6) gi|9789967| (SEQ ID NO:68)
10 20 30 40 50
Figure imgf000095_0001
gi 1174412 5 | B^ GT BIPSJEgl^P ^ p^LSf ^^JA' ^fcgV iTPHK gi| 11701051 BKE&S !5ιJrogilEroMMιιlFkΛitJ«fefetA *«itUM<i!ιl gi 11392904 1 Qrasg laBTOKlBiπfffe^ΛilFki^jMd^ 'i^iMfatl
Figure imgf000095_0002
gi I 9789967 I |Jjj KH|§ [^^E^lll^Mfciiillft^Je^JA' 'jii^iAliiil
60 70 80 90 100
NOV7 --LSCG G^FgVF^SJSePEi2aATF|?5GKL}LSS3JScE gi]l7441225] VG SCG G^F^VF^sH^3SfESMA F 5GKLfeiLiEEScS gi] 1170105 I L_^^^iftM l4^iιiW«Λ. κJ ,^^^^J*^ .ιt^it JJe. gi|l3929044| Jt-MΛmΛt^ wim ii^t ∞siiitnmMistΛMMMx-tiits gi I 204416] gi|9789967| -tt<aaaιaι_ι»ι«)a-«.iMiι.'^B3?iE!M>MaaιιwM«iB?j«wιtt/;.ι_ιaaaBe)
110 120 130 140 150
NOV7 liιHfegcgBRaτiffiιWroaHτitfa»ιιaι«vl35vMftl^B.^I5B gi| 17441225] ιscleHaSτ.1fflιroE^ffϊ.Jli*ιifcl l33^^ gi|H70105| JFJliB^<ri;i{ ΛM^ ^A^*Λ^Λ^M»tιk.^fejda_alτHa H3 gi|l3929044] l tmarf»t^ A*^ t4^*^A*«Md«bt{i*Λ^ata|τI^a^ gi|204416| |Ffj^tle.;^ J*<JMit ^*J.Ja»«ιaiM«w^»iKEπS|Bqn|5^ gi|9789967| S3s|
160 170 180 190 200
Figure imgf000095_0003
Figure imgf000096_0001
VGICAGISSNWPMYLGE APKNLRGA GVfflPOLFITVGILVAOLFG
MffirtttfeTrt^WABi WrtMtaj^. T^^
ii.f.trtWJe»W.-l^tJdartitt.3.<>il;j:^»B;WJ^Wia»)l.3*»i>tW«ft/i.)B31e>«
210 220 230 240 250
Figure imgf000096_0002
gi|l7441225| AifflGKPAMs|vflEføilιιfeMJi^^
Figure imgf000096_0003
gi|204416|
Figure imgf000096_0004
260 270 280 290 300
NOV7 p| RSϊ3_^τ!^i ^t# iRffi^lESHϊ^LHBCJ@SϊSΞ!i!iIB
Figure imgf000096_0005
gi| 13929044 I .AI.I^rl.W.Atø^^^^
Figure imgf000096_0006
310 320 330 340 350
NOV7 H^^S3βϊN^Ξ3'IiBτfflEBaAHS_^E3SsSH2:]Jϊ ;τ&
Figure imgf000096_0007
gi ] 13929044 I -CTBTiBWW^tk^tt^Wa'iAtMi'i-^iWi.Ji it^ ii^JeliileA^tt ^iiiiW^
Figure imgf000096_0008
360 370 380 390 400
NOV7 B 3gRg2^j|53jAg^GlgGi^S5BβP PHR^PEIISgl5G0lSiFA gi| 17441225 I Ajj 3Rj 333_H?^GIE_tø_43_&ffl_^^LE^ gi|l3929044| gi|204416| gi|9789967|
410 420 430 440 450
NOV7 QiAΞllδiΞΞ3^δ RE_Ξ_ε_^^^F!!]^I^B':clB^ΞF^3Ss
Figure imgf000096_0009
gi|l3929044| gi|204416| Jgra33a^F^^4^ι^ri_ι^»LJ^ ^fei^^«nt^ gi|9789967| M^WAM^WgMBaWBfeyMMUJd^
460 470 480 490 500
Figure imgf000097_0001
gi|9789967| π8Met<cld>Wai__h -J πaFffl^>i *i dflL daιtM^
510 520
....M..I....I....I....I.
Figure imgf000097_0002
gi 117441225 ] --lj| Eϊ^ EJτiϊlϊAGS3τASPAKETSF gi 11170105 I lt<liJa.i43.alκi3fe;DB3^TRi518[ gi]l3929044| tAW 4atøϊfl5lTOB33Sτ l5Sl gi|204416] Ji i a.!a.aiκi5tKbffl3^ITRl5B[
Figure imgf000097_0003
Table 7E lists the domain description from DOMAIN analysis results against NON7. This indicates that the ΝON7 sequence has properties similar to those of other proteins known to contain this domain.
Table 7E. Domain Analysis of ΝOV7 gnl|Pfam|pfam00083, sugar r, Sugar (and other) transporter.
CD-Length = 447 residues, 96.6% aligned Score = 246 bits (629), Expect = 2e-66
Query : 21 FQVFKSFYNETYFERHATFM DGKLMLLLWSCTVSMFPl-GGIi GSLLVGLiVDSCGR
76
V F F + + VS+F +G +GSL G L D GR
Sbjct : 16 TGVIGGFATLIDFLFFFGGLTSSGSCAESTV SGLWSIFFVGRPIGSLFAGKLGDRFGR 75
Query: 77 KGTLLIIrølFAIIPAILMGVSKVAKAFELIVFSRWLGVCAGISYSALPMYLGE APKNL 136
K +LLI + +1 ++L G++ A F ++ RV++G+ G + +PMY+ E+APK L Sbjct : 76 KKSLLIGLVLFVIGSL SGLAPGA--FYL IVGRV VGLGVGGASVLVPMYISEIAPKAL 133
Query : 137 RGMVGTMTΞVFVIVGVFLAQIFSLQAILGNPAGWPVLLA TGVPAL QLLTLPFFPESPR 196
RG +G++ ++ + +G+ +A I L N GW + L L VPAL L+ L F PΞSPR Sbjct : 134 RGALGSLYQ GITIGI VAAIIG GLNKTNN GWRIPLG QLVPALLLLIG FLPESPR 193
Query: 197 YS IQKGDEATARPLRRLRGHTDWEABiSDiWHAJSAAEiJAEGHLSVLHLCA RSLR Q 256
+ +++ E +LRG D++ E+++ +AE A + + R + L
Sbjct : 194 WLVLKGKLEEARAVLAKLRGVEDVDQEIQEE AE EAGVSSEKAGLELF--RGRTRQRLL 251
Query: 257 SIIV MAGQQLSGINAINYYADTIYTSAGVEAAHSQYVTVGSGVVNIVMTITS LVERL 316
+4-L QQL+GINAI YY+ TI+ S G+ + + VT+ GWN V T ++ V+R Sbjc : 252 MGV QIFQQ TGINAIFYYSPTIFKSVGMSDSVA LVTIIVGWNFVATFVAIF VDRF 311
Query : 317 GRRHLLLAGYGICGSAC VLTVSPPPQNRVPELSYLGIICVFAYIAGHSIGPSPVPS R 376
GRR L L G L+L V+ P + 1+ + +IA ++G P+P V+ Sbjct : 312 GRRPLLLLGAAGMAICF I GVA-LLLLNKPGAGIVAIVFILLFIAFFALG GPIPWVIL 370
Query: 377 TEIFLQSSRRAAFMVDGAVH LTNFIIGF FPSIQEAIG-AYSFIIFAGIC LTAIYIYV 435
+E+F R A + A + L NFIIGF FP I AIG Y F+ FAG+ +L +4-+Y Sbjct: 371 SELFPTGVRSKAMA ATAAN LANFIIGFLFPYITGAIGGGYVF FFAGLLV FILFVYF 430
Query: 436 VIPETKGKTFVEINRIF 452
+PETKG+T EI+ +F Sbjct: 431 FVPETKGRTLEEIDELF 447
Sugar transport is a critical feature of many cell types in the body as energy storage and metabolism or defects thereof can cause a variety of human diseases. For example, glucose tranporter 4 (GLUT4) is critical to insulin-sensitive glucose uptake. Novel sugar transporters can be important for obesity, diabetes, and cancer targets (see, Hundal HS, et al., Adv Exp Med Biol 1998;441:35-45).
Biochemical and immunocytochemical studies have revealed that, in addition to GLUT1 and GLUT4, human skeletal muscle also expresses the GLUT5 hexose transporter. The subcellular distribution of GLUT5 is distinct from that of GLUT4, being localised exclusively in the sarcolemmal membrane. The substrate selectivity of GLUT5 is also considered to be different to that of GLUT1 and GLUT4 in that it operates primarily as a fructose transporter. Consistent with this suggestion studies in isolated human sarcolemmal vesicles have shown that fructose transport obeys saturable kinetics with a Nmax of 477 +/- 37 pmol.mg protein-1 min-1 and a Km of 8.3 +/- 1.2 mM. Unlike glucose uptake, fructose transport in sarcolemmal vesicles was not inhibited by cytochalasin B suggesting that glucose and fructose are unlikely to share a common route of entry into human muscle. Muscle exercise, which, stimulates glucose uptake through the increased translocation of GLUT4 to the plasma membrane, does not increase fructose transport or sarcolemmal GLUT5 content. In contrast, muscle inactivity, induced as a result of limb immobilization, caused a significant reduction in muscle GLUT4 expression with no detectable effects on GLUT5. The presence of a fructose transporter in human muscle is compatible with studies showing that this tissue can utilise fructose for both glycolysis and glycogenesis. However, the full extent to which provision of fructose via GLUT5 is important in meeting the energy requirements of human muscle during both physiological and pathophysiological circumstances remains an issue requiring further investigation.
The disclosed NON7 nucleic acid of the invention encoding a glucose transporter-like protein includes the nucleic acid whose sequence is provided in Table 7A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 7 A while still encoding a protein that maintains its glucose transporter-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 30 percent of the bases maybe so changed.
The disclosed ΝON7 protein of the invention includes glucose transporter-like protem whose sequence is provided in Table 7B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 7B while still encoding a protein that maintains its glucose transporter-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 41 percent of the residues may be so changed.
The protein similarity information, expression pattern, and map location for the glucose transporter-like protein and nucleic acid (ΝON7) disclosed herein suggest that ΝON7 may have important structural and/or physiological functions characteristic of glucose transporter family. Therefore, the ΝON7 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications. These include serving as a specific or selective nucleic acid or protein diagnostic and/or prognostic marker, wherein the presence or amount of the nucleic acid or the protein are to be assessed, as well as potential therapeutic applications such as the following: (i) a protein therapeutic, (ii) a small molecule drug target, (iii) an antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), (iv) a nucleic acid useful in gene therapy (gene delivery/gene ablation), and (v) a composition promoting tissue regeneration in vitro and in vivo.
91 The NON7 nucleic acids and proteins of the invention are useful in potential diagnostic and therapeutic applications implicated in various diseases and disorders described below and/or other pathologies. For example, the compositions of the present invention will have efficacy for treatment of patients suffering from obesity, diabetes, cancer, inflammation, CΝS diseases and other diseases, disorders and conditions of the like. The ΝON7 nucleic acid, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
ΝON7 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immunospecifically to the novel substances of the invention for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-ΝOVX Antibodies" section below. For example the disclosed ΝOV7 protein have multiple hydrophilic regions, each of which can be used as an immunogen. hi one embodiment, contemplated NON7 epitope is from about amino acids 20 to 40. In other embodiments, contemplated ΝON7 epitopes are from amino acids 200 to 250, from amino acids 260 to 265, from amino acids 360 to 365, or from amino acids 440 to 460. This novel protein also has value in development of powerful assay system for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
ΝOV8
A disclosed NON8 nucleic acid of 3270 nucleotides identified as SEQ ID ΝO:25 (also designated as Ace. No. CG95545-01) encoding a novel Type la Membrane Sushi-Containing Domain-like protein is shown in Table 8 A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 309-311 and ending with a TGA codon at nucleotides 2550-2552.
Table 8A.
NOV8 Polynucleotide
SEQ ID NO:25
CGGGGCTCTGCGTCAGCTGTGTCATTATCCGATGAGTGTCTGTCCCCCTTTGCGAATGTGAGCGGCGAGA GGGCAGCAAGTGCGGAGCCAGAGACGGACGCGGAACGGGCGTGTCCTAAGCCCAGGCCCCGACAGGAGGA AGGACCCGCGCTCTGCGGCCTCCCGGGGACCCCGCAGCGCCCCCCGCTTCCCTCGGCGGCGCCGGAAGCC GCCGGCTGGTCCCCTCCCCGCGGCGCCTGTAGCCTTATCTCTGCACCCTGAGGGCCCCGGGAGGAGGCGC GGGCGCGCCGGGAGGGACCGGCGGCGGCATGGGCCGGGGGCCCTGGGATGCGGGCCCGTCTCGCCGCCTG CTGCCGCTGTTGCTGCTGCTCGGCCTGGCCCGCGGCGCCGCGGGAGCGCCGGGCCCCGACGGTTTAGACG TCTGTGCCACTTGCCATGAACATGCCACATGCCAGCAAAGAGAAGGGAAGAAGATCTGTATTTGCAACTA TGGATTTGTAGGGA?s.CGGGAGFGACTCAGTGTGTTGATAAAAATGAGTGCCAGTTTGGAGCCACTCTTGT CTGTGGGAACCACACATCTTGCCACAACACCCCCGGGGGCTTCTATTGCATTTGCCTGGAAGGATATCGA GCCACAAACAACAACAAGACATTCATTCCCAACGATGGCACCTTTTGTACAGACATAGATGAGTGTGAAG TTTCTGGCCTGTGCAGGCATGGAGGGCGATGCGTGAACACTCATGGGAGCTTTGAATGCTACTGTATGGA TGGATACTTGCCAAGGAATGGACCTGAACCTTTCCACCCGACCACCGATGCCACATCATGCACAGAAATA GACTGTGGTACCCCTCCTGAGGTTCCAGATGGCTATATCATAGGAAATTATACGTCTAGTCTGGGCAGCC AGGTTCGTTATGCTTGCAGAGAAGGATTCTTCAGTGTTCCAGAAGATACAGTTTCAAGCTGCACAGGCCT GGGCACATGGGAGTCCCCAAAATTACATTGCCAAGAGATCAACTGTGGCAACCCTCCAGAAATGCGGCAC GCCATCTTGGTAGGAAATCACAGCTCCAGGCTGGGCGGTGTGGCTCGCTATGTCTGTCAAGAGGGCTTTG AGAGCCCTGGAGGAAAGATCACTTCTGTTTGCACAGAGAAAGGCACCTGGAGAGAAAGTACTTTAACATG CACAGAAATTCTGACAAAGATTAATGATGTATCACTGTTTAATGATACCTGTGTGAGATGGCAAATAAAC TCAAGAAGAATAAACCCCAAGATCTCATATGTGATATCCATAAAAGGACAACGGTTGGACCCTATGGAAT CAGTTCGTGAGGAGACAGTCAACTTGACCACAGACAGCAGGACCCCAGAAGTGTGCCTAGCCCTGTACCC AGGCACCAACTACACCGTGAACATCTCCACAGCACCTCCCAGGCGCTCGATGCCAGCCGTCATCGGTTTC CAGACAGCTGAAGTTGATCTCTTAGAAGATGATGGAAGTTTCAATATTTCAATATTTAATGAAACTTGTT TGAAATTGAACAGGCGTTCTAGGAAAGTTGGATCAGAACACATGTACCAATTTACCGTTCTGGGTCAGAG GTGGTATCTGGCTAACTTTTCTCATGCAACATCGTTTAACTTCACAACGAGGGAACAAGTGCCTGTAGTG TGTTTGGATCTGTACCCTACGACTGATTATACGGTGAATGTGACCCTGCTGAGATCTCCTAAGCGGCACT CAGTGCAAATAACAATAGCAACTCCCCCAGCAGTAAAACAGACCATCAGTAACATTTCAGGATTTAATGA AACCTGCTTGAGATGGAGAAGCATCAAGACAGCTGATATGGAGGAGATGTATTTATTCCACATTTGGGGC CAGAGATGGTATCAGAAGGAATTTGCCCAGGAAATGACCTTTAATATCAGTAGCAGCAGCCGAGATCCCG AGGTGTGCTTGGACCTACGTCCGGGTACCAACTACAATGTCAGTCTCCGGGCTCTGTCTTCGGAACTTCC TGTGGTCATCTCCCTGACAACCCAGATAACAGAGCCTCCCCTCCCGGAAGTAGAATTTTTTACGGTGCAC AGAGGACCTCTACCACGCCTCAGACTGAGGARAGCCAΑGGAGAAAAATGGACCAATCAGTTCATATCAGG TGTTAGTGCTTCCCCTGGCCCTCCAAAGCACATTTTCTTGTGATTCTGAAGGCGCTTCCTCCTTCTTTAG CAACGCCTCTGATGCTGATGGATACGTGGCTGCAGAACTACTGGCCAAAGATGTTCCAGATGATGCCATG GAGATACCTATAGGAGACAGGCTGTACTATGGGGAATATTATAATGCACCCTTGAAAAGAGGGAGTGATT ACTGCATTATATTACGAATCACAAGTGAATGGAATAAGGTGAGAAGACACTCCTGTGCAGTTTGGGCTCA GGTGAAAGATTCGTCACTCATGCTGCTGCAGATGGCGGGTGTTGGACTGGGTTCCCTGGCTGTTGTGATC ATTCTCACATTCCTCTCCTTCTCAGCGGTGTGATGGCAGATGGACACTGAGTGGGGAGGATGCACTGCTG CTGGGCAGGTGTTCTGGCAGCTTCTCAGGTGCCCGCACAGAGGCTCCGTGTGACTTCCGTCCAGGGAGCA TGTGGGCCTGCAACTTTCTCCATTCCCAGCTGGTCCCCATTCCTGGATTTAAGATGGTGGCTATCCCTGA GGAGTCACCATAAGGAGAAAA.CTCAGGAATTCTGAGTCTTCCCTGCTACAGGACCAGTTCTGTGCAATGA ACTTGAGACTCCTGATGTACACTGTGATATTGACCGAAGGCTACATACAGATCTGTGAATCTTGGCTGGG ACTTCCTCTGAGTGATGCCTGAGGGTCAGCTCCTCTAGACATTGACTGCAAGAGAATCTCTGCAACCTCC TATATAAAAGCATTTCTGTTAATTCATTCAGAATCCATTCTTTACAATATGCAGTGAGATGGGCTTAAGT TTGGGCTAGAGTTTGACTTTATGAAGGAGGTCATTGAAAAAGAGAACAGTGACGTAGGCAAATGTTTCAA GCACTTTAGAAACAGTACTTTTCCTATAATTAGTTGATATACTAATGAGAAAATATACTAGCCTGGCCAT GCCAATAAGTTTCCTGCTGTGTCTGTTAGGCAGCATTGCTTTGATGCAATTTCTATTGTCCTATATATTC AAAAGTAATGTCTACATTCCAGTAAAAATATCCCGTAATTAAGAAAAAAAA.
In a search of sequence databases, it was found, for example, that the nucleic acid sequence of this invention has 2428 of 2431 bases (99%) identical to a gb:GENBANK- ID:HSM802135|acc:AL137432.1 mRNA from Homo sapiens (Homo sapiens mRNA; cDNA DKFZp761E1824 (from clone DKFZp761E1824); partial eds).
The disclosed NOV8 polypeptide (SEQ ID NO:26) encoded by SEQ ID NO:25 has 747 amino acid residues and is presented in Table 8B using the one-letter amino acid code. Signal P, Psort and/or Hydropathy results predict that NON8 is a Type la membrane protein, has a signal peptide, and is likely to be localized at the plasma membrane with a certainty of 0.9190. In other embodiments, ΝON8 may also be localized to the endoplasmic reticulum (membrane) with a certainty of 0.100, and lysosomes with a certainty of 0.2000. The most likely cleavage site for ΝON8 is between positions 29 and 30, AAG-AP. Table 8B.
NON8 Polypeptide
SEQ ID ΝO-.26
MGRGPWDAGPSRRLLPLLLLLGLARGAAGAPGPDGLDVCATCHEHATCQQREGKKICICNYGFVGNGRTQ CVDKNECQFGATLVCGNHTSCHNTPGGFYCICLEGYRATN NKTFIPNDGTFCTDIDECEVSGLCRHGGR CVNTHGSFECYCMDGYLPRNGPEPFHPTTDATSCTEIDCGTPPEVPDGYIIGNYTSSLGSQVRYACREGF FSVPEDTVSSCTGLGTWESPKLHCQEINCGNPPE RHAILVGNHSSRLGGVARYVCQEGFESPGGKITSV CTEKGTWRESTLTCTEILTKINDVSLF DTCVR QINSRRINPKISYVISIKGQRLDPMESVREETVNLT TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGSFNISIFNETCL LNRRSRKV GSEHMYQFTVLGQRWYLANFSHATSFNFTTREQVPVVCLDLYPTTDYTVNVTI.LRSPKRHSVQITIATPP AVKQTISNISGFNETCLRWRSIKTADMEEMYLFHI GQR YQKEFAQEMTFNISSSSRDPEVCLDLRPGT NYNVSLRALSSELPWISLTTQITEPPLPEVEFFTVHRGPLPRDRLRKAKEKNGPISSYQVLVLPLALQS TFSCDSEGASSFFSNASDADGYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCII RITSE NKVRRHSCA AQVKDSSLMLLQMAGVGLGSLAVVIILTFLSFSAV
A search of sequence databases reveals that the NON8 amino acid sequence has 570 of 570 amino acid residues (100%) identical to, and 570 of 570 amino acid residues (100%) similar to, the 570 amino acid residue ptnr:SPTREMBL-ACC:Q9ΝTA7 protein from Homo sapiens (Human) (HYPOTHETICAL 63.7 KDA PROTEIN)(Fig. 3B).
NON8 maps to chromosome 1 lpl5.3, and is found in at least Adrenal Gland/Suprarenal gland, Amygdala, Aorta, Bone Marrow, Brain, Colon, Dermis, Duodenum, Heart, Hippocampus, Hypothalamus, Kidney, Liver, Lung, Lymph node, Lymphoid tissue, Pancreas, Pituitary Gland, Placenta, Retina, Small Intestine, Spinal Chord, Spleen, Substantia Νigra, Synovium/Synovial membrane, Testis, Thalamus, Urinary Bladder, Uterus. This information was derived by determining the tissue sources of the sequences that were included in the invention including but not limited to SeqCalling sources, Public EST sources, Literature sources, and or RACE sources.
ΝON8 also has homology to the amino acid sequence shown in the BLASTP data listed in Table 8C.
Figure imgf000102_0001
Figure imgf000103_0004
The homology of these sequences is shown graphically in the ClustalW analysis shown in Table 8D.
Table 8D. Information for the ClustalW proteins
1) NOV8 (SEQ ID NO:25)
2) NOV9 (SEQ ID N0.27)
3) gi|ll360234) (SEQ ID N0.69)
4) gi|l740222θ| (SEQ ID NO:70)
5) gi I 16552183 I (SEQ ID NO: 71)
6) gi|l4740162| (SEQ ID NO:72)
7) gi|l0438017| (SEQ ID NO:73)
10 20 30 40 50
NOV8 MGRGPWDAGPSRR LPLL G ARGAAGAPGPDGLDVCATCHEHATCQQ
NOV9 MGRGPWDAGPSRR LPL LL GLARGAAGAPGPDGLDVCATCHEHATCQQ gi| 11360234 I gi| 17402220 I
Figure imgf000103_0001
60 70 80 90 100
NOV8 REGKKICICNYGFVGNGRTQCVDKNECQFGATLVCGNHTSCHNTPGGFYC
NOV9 REGKKICICNYGFVGNGRTQCVDKNECQFGAT VCGNHTSCHNTPGGFYC
Figure imgf000103_0002
gi| 14740162 I
110 120 130 140 150
NOV8 ICLEGYRATNNNKTFIPNDGTFCTDIDECEVSGLCRHGGRCVNTHGSFEC
NOV9 ICLEGYRATNNNKTFIPNDGTFCTDIDECEVSGLCRHGGRCVNTHGSFEC
gi 117402220 I ECEVSGLCRHGGRCVNTHGSFEC
Figure imgf000103_0003
160 170 180 190 200 NOV8 YCMDGYLPRNGPEPFHPTTDATSCTEI M«t!ιιt2ά-ΛU-Mtv* tmvii*Α κtte
NOV9 YCMDGYLPRNGPEPFHPTTDATSCTEI gi 111360234 I PCGTPPEVPDGYIIGNYTSSLGS gi 117402220 ] YCMDGYLPRNGPEPFHPTTDATSCTEI itϋttXtKtHϊiΦiftfi gi 116552183 | --MDGYLPRNGPEPFHPTTDATSCTEI THTeraaaawaiia'
Figure imgf000104_0001
210 220 230 240 250
NOV8 1VRYACREGFFSVPEDTVSSCTGLGT ESPKLHCOEINCGNPPEMRHAII
Figure imgf000104_0002
gi|ll360234| gi|l740222θ| gi|l6552183| WRYACREGFFSVPEDTVSSCTGLGTWESPKLHCOEINCGNPPEMRHAII gi] 14740162 I
260 270 280 290 300
NOV8 Meι^ι:>aaa>««wjf*,»u«CMMMaa:<atιt>M>iMι«4iMa:<WM'jι;T3fc« wtWMg>»w»;'
Figure imgf000104_0003
gi I 11360234 I rGNHSSRLGGVARYVCOEGFESPGGKITSVCTEKGTWRESTLTCTEILTI gi|l7402220| 3NHSSRLGGVARYVCOEGFESPGGKITSVCTEKGTWRESTLTCTEILTI gi| 16552183 I gi 114740162 I 3NHSSRLGGVARYVCOEGFESPGGKITSVCTEKGTWRESTLTCTEILTI gi|l0438017|
310 320 330 340 350
NOV8
NOV9 gi|ll360234| WjapOT<aιaar*iMiaMi^ iι iacr<raflwarata gmr<aMm gi|l7402220| JDVSLFNDTCVRWQINSRRINPKISYVISIKGQRLDPMESVREETVNL1 gi|l6552183| NDVSLFNDTCVRWOINSRRINPKISYVISIKGORLDPMESVREETVNL1 gi 114740162 I NDVSLFNDTCVRWOINSRRINPKI! IWdiSlaftitiaatalnMiSIBI gi|l0438017| BSSΞ
360 370 380 390 400
NOV8 ΓDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS
NOV9 TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi 111360234 I TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l740222θ| TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l6552183| TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l4740162| ΓDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l0438017| TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS
10 420 430 440 450
NOV8 FNISIFNETCLKLNRRSRKVGSEHMYQFTVLGQRWYLANFSHATSFNFTT NOV9 aswaiasiaww nrn atfatsimamam iH
Figure imgf000105_0001
gi|l740222θ| gi 116552183 I NISIFNETCLKL RRSRKVGSEHMYOFTVLGORWYLANFSHATSFNFT
gi|l0438017| FNISIFNETCLKLNRRSRKVGSEHMYQFTVLGQR YLANFSHATSFNFTT
460 470 480 490 500
NOV8 REOVPWCLDLYPTTDYTVNVTLLRSPKRHSVOITIATPPAVKOTISNI
NOV9 :^aM^^a'*'iw«l^ιn^«awB»-.'^ι^^i^^'Jan^lr^aaa;^^Bt^«^wι-it-^ι»aaiV^:<»jι>♦a l»a gi| 11360234 I IEOVPWCLDLYPTTDYTVNVTLLRΞPKRHSVQITIATPPAVKOTISNI! gi]l740222θ| gi] 16552183 | tEQVPVVCLD YPTTDYTVNVTLLRSPKRHSVQITIATEPAVKQTISNI gi|l4740162| iOVPWCLDLYPTTDYTVNVTLLRSPKRHSVQITIATPPAVKOTISNI! gi|l0438017|
510 520 530 540 550
NOV8 -FNETCLRWRSIKTADMEEMYLFHIWGORWYQKEFAQEMTFNISSSSRD
NOV9 & mιiwmM^t>AιM>m#MMΛMtmtm M>r &iτ w»m Λ&kUάM gi| 11360234 I gi|l740222θ|
Figure imgf000105_0002
gi] 16552183] gi] 14740162] gi|l0438017|
560 570 580 590 600
NOV8 VC D RPGTNYNVSLRALSSELPWISLTTC H MMύAHMύMHMΪ
NOV9 IVCLDLRPGTNYNVSLRALSSELPWISLTTOITEPPLPEVEFFTVHR
Figure imgf000105_0003
gi|l740222θ| VCLDLRPGTNYNVSLRALSSELPWISLTTOITEPPLPEVEFFTVHRG
Figure imgf000105_0004
gi|l0438017| 3tWa.)l).-tatiι.li.>'<i'ttfct<t:tJ<ftM^Bat*t*fc)fttHf.ln) a)aai.!)attig_^aιttj.l.f.i
610 620 630 640 650
NOV8 ιi.a.iBaft: .*j;«^^Ma«;{A'tftfiw_f.aι»jirt^g^{w.M^t>Wt.^aWitWι>j»
Figure imgf000105_0005
gi|ll360234| aammwΛ« t^tΛΛ »mΛUΛ-j ssmsis^ssΛ^msf^s^ ^ » gi|l7402220| 1iltiaf&Α'i*\VΛUnU*ϊ*f*KVIlXWtK1ΑCtXKiaa MX1tMt gi 116552183 I PRLRLRKAKEKNGPISSYOVLVLPLALOSTFΞCDSEGASSFFSNASDA gi 114740162 I .PRLRLRKAKEKNGPISSYOVLVLPLALOSTFSCDSEGASSFFSNASDA
Figure imgf000105_0006
660 670 680 690 700
NOV8 w m ϊ iΩ N βs i &mamfϊ NOV9 GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE gi|ll360234| GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE
Figure imgf000106_0001
gi]l6552183] gi|l4740162| t'4Vi.w^Λ^;<ι\i-jι)iMai5««a<nrta<vvt.sVϋtaafli«Je<;.iyrtt*««ι:«»ιfc< gi|l0438017|
710 720 730 740
NOV8 ismsw i!
NOV9 mt'Avnni^wms»vim^mmwmΛtm*nmmgβBm*fiiΛtιs^x gi|ll360234| gijl740222θ| gi|l6552183| IWSJjg-lϊEIilcc tmffi B vpwiWI- gi|l4740162| mvmaaaϊwm m iasmiMtmΛtiVi Ga gi|l0438017|
Table 8E lists the domain description from DOMAIN analysis results against NON8. This indicates that the ΝON8 sequence has properties similar to those of other proteins known to contain this domain.
Table 8E. Domain Analysis of ΝOV8 gnl ] Smart | smar 00179 , EGF_CA, Calcium-binding EGF-like domain
CD-Length = 41 residues, 80.5% aligned Score = 52.8 bits (125), Expect = 7e-08
Query: 125 DIDECEVSG CRHGGRCVNTHGSFECY-CMDGY 156
DIDEC C++GG CVNT GS+ C C GY Sbjct: 1 DIDECASGNPCQNGGTCWTVGSYRCEECPPGY 33
The polynucleotide encoding a disclosed NON8 Type la Membrane Sushi-Containing Domain-like protein is identified by the comparative sequencing of human chromosome 1 lpl5 and mouse chromosome 7. This gene contains two very important domains associated with developmental proteins- the CUB domain and the domain first found in Clr, Cls, uEGF, and bone morpho genetic protein. The CUB domain is found in 16 functionally diverse proteins such as the dorso-ventral patterning protein tolloid, bone morphogenetic protein 1, a family of spermadhesins, complement subcomponents Cls/Clr and the neuronal recognition molecule A5. Most of these proteins are known to be involved in developmental processes. The second domain is found mostly among developmentally-regulated proteins and spermadhesins.
The disclosed ΝON8 nucleic acid of the invention encoding a Type la Membrane
Sushi-Containing Domain-like protein includes the nucleic acid whose sequence is provided in
Table 8 A, or a fragment thereof. The invention also includes a mutant or variant nucleic that enhances the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 1% percent of the bases may be so changed.
The disclosed NON8 protein of the invention includes Type la Membrane Sushi- Containing Domain-like protein whose sequence is provided in Table 8B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 8B while still encoding a protein that maintains its Type la Membrane Sushi-Containing Domain-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 0% percent of the residues may be so changed.
The invention further encompasses antibodies and antibody fragments, such as Fa or (Fat>)2, that bind immuno specifically to any of the proteins of the invention.
The above defined information for this invention suggests that this Type la Membrane Sushi-Containing Domain-like protein (ΝON8) may function as a member of a glucose transporter family. Therefore, the ΝON8 nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The ΝON8 nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in cancer including but not limited to Inflamation, Autoimmune disorders, Aging and Cancer. For example, a cDΝA encoding the Type la Membrane Sushi-Containing Domain-like protein (ΝON8) may be useful in gene therapy, and the Type la Membrane Sushi-Containing Domain-like protein (ΝON8) may be useful when administered to a subject in need thereof. By way of nonlimiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from cancer, trauma, regeneration (in vitro and in vivo), viral/bacterial/parasitic infections, Atherosclerosis, Aneurysm, Hypertension, Fibromuscular dysplasia, Stroke, Scleroderma, Obesity, Transplantation, Myocardial infarction, Embolism, Cardiovascular disorders, Bypass surgery, Adrenoleukodystrophy, Congenital Adrenal Hyperplasia, Diabetes, Non Hippel-Lindau (NHL) syndrome, Pancreatitis, Cirrhosis, Hemophilia, Hypercoagulation, Idiopathic thrombocytopenic purpura, Immunodeficiencies, Graft vesus host disease (GNHD), Lymphedema, Allergies, autoimmume disease, Alzheimer's disease, Tuberous sclerosis, hypercalceimia, Parkinson's disease, Huntington's disease, Cerebral palsy, Epilepsy, Lesch- Νyhan syndrome, Multiple sclerosis, Ataxia-telangiectasia, Leukodystrophies, Behavioral disorders, Addiction, Anxiety, Pain, Neuroprotection, Systemic lupus erythematosus, Asthma, Emphysema, Scleroderma, ARDS, Renal artery stenosis, Interstitial nephritis, Glomerulonephritis, Polycystic kidney disease, Systemic lupus erythematosus, Renal tubular acidosis, IgA nephropathy and other diseases, disorders and conditions of the like.
The NOV8 nucleic acid encoding Type la Membrane Sushi-Containing Domain-like protein, and the Type la Membrane Sushi-Containing Domain-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOV8 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel NON8 substances for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-ΝOVX Antibodies" section below. The disclosed ΝOV8 protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV8 epitope is from about amino acids 40 to 300. In another embodiment, a NON8 epitope is from about amino acids 305 to 360, from about 400 to 450, from about 500 to 560, from about 580 to 610, and from about 620 to 680. These novel proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
ΝON9
A disclosed ΝON9 nucleic acid of 2507 nucleotides identified as SEQ ID ΝO:27 (designated CuraGen Ace. No. CG95545-02) encoding a novel Type la Membrane-Sushi Domain Containing Protein-like protein is shown in Table 9A. An open reading frame was identified beginning at nucleotides 309-311 and ending at nucleotides 2469-2471. Putative untranslated regions are indicated by underline.
Table 9A.
NON9 Polynucleotide
SEQ ID ΝO:27
CGGGGCTCTGCGTCAGCTGTGTCATTATCCGATGAGTGTCTGTCCCCCTTTGCGAATGTG 60 AGCGGCGAGAGGGCAGCAAGTGCGGAGCCAGAGACGGACGCGGAACGGGCGTGTCCTAAG 120 CCCAGGCCCCGACAGGAGGAAGGACCCGCGCTCTGCGGCCTCCCGGGGACCCCGCAGCGC 180
CCCCCGCTTCCCTCGGCGGCGCCGGAAGCCGCCGGCTGGTCCCCTCCCCGCGGCGCCTGT 240
AGCCTTATCTCTGCACCCTGAGGGCCCCGGGAGGAGGCGCGGGCGCGCCGGGAGGGACCG 300
GCGGCGGCATGGGCCGGGGGCCCTGGGATGCGGGCCCGTCTCGCCGCCTGCTGCCGCTGT 360
TGCTGCTGCTCGGCCTGGCCCGCGGCGCCGCGGGAGCGCCGGGCCCCGACGGTTTAGACG 20
TCTGTGCCACTTGCCATGAACATGCCACATGCCAGCAAAGAGAAGGGAAGAAGATCTGTA 80
TTTGCAACTATGGATTTGTAGGGAACGGGAGGACTCAGTGTGTTGATAAAAATGAGTGCC 5 0
AGTTTGGAGCCACTCTTGTCTGTGGGAACCACACATCTTGCCACAACACCCCCGGGGGCT 600
TCTATTGCATTTGCCTGGAAGGATATCGAGCCACAAACAACAA.CAAGACATTCATTCCCA 660
ACGATGGCACCTTTTGTACAGACATAGATGAGTGTGAAGTTTCTGGCCTGTGCAGGCATG 720
GAGGGCGATGCGTGAACACTCATGGGAGCTTTGAATGCTACTGTATGGATGGATACTTGC 780
CAAGGAATGGACCTGAACCTTTCCACCCGACCACCGATGCCACATCATGCACAGAAATAG 8 0
ACTGTGGTACCCCTCCTGAGGTTCCAGATGGCTATATCATAGGAAATTATACGTCTAGTC 900
TGGGCAGCCAGGTTCGTTATGCTTGCAGAGAAGGATTCTTCAGTGTTCCAGAAGATACAG 960
TTTCAAGCTGCACAGGCCTGGGCACATGGGAGTCCCCAAAATTACATTGCCAAGAGATCA 1020
ACTGTGGCAACCCTCCAGAAATGCGGCACGCCATCTTGGTAGGAAATCACAGCTCCAGGC 1080
TGGGCGGTGTGGCTCGCTATGTCTGTCAAGAGGGCTTTGAGAGCCCTGGAGGAAAGATCA 1140
CTTCTGTTTGCACAGAGAAAGGCACCTGGAGAGAAAGTACTTTAACATGCACAGAAATTC 1200
TGACAAAGATTARTGATGTATCACTGTTTAATGATACCTGTGTGAGATGGCAAATA5ACT 1260
CAAGAAGAATAAACCCCAAGATCTCATATGTGATATCCATAAAAGGACAACGGTTGGACC 1320
CTATGGAATCAGTTCGTGAGGAGACAGTCAACTTGACCACAGACAGCAGGACCCCAGAAG 1380
TGTGCCTAGCCCTGTACCCAGGCACCAACTACACCGTGAACATCTCCACAGCACCTCCCA 1440
GGCGCTCGATGCCAGCCGTCATCGGTTTCCAGACAGCTGAAGTTGATCTCTTAGAAGATG 1500
ATGGAAGTTTCAATATTTCAATATTTAATGAAACTTGTTTGAAATTGAACAGGCGTTCTA 1560
GGAAAGTTGGATCAGAACACATGTACCAATTTACCGTTCTGGGTCAGAGGTGGTATCTGG 1620
CTAACTTTTCTCATGCAACATCGTTTAACTTCACAACGAGGGAACAAGTGCCTGTAGTGT 1680
GTTTGGATCTGTACCCTACGACTGATTATACGGTGAATGTGACCCTGCTGAGATCTCCTA 1740
AGCGGCACTCAGTGCAAATAACAATAGCAACTCCCCCAGCAGTAAAACAGACCATCAGTA 1800
ACATTTCAGGATTTAATGAAACCTGCTTGAGATGGAGAAGCATCAAGACAGCTGATATGG 1860
AGGAGATGTATTTATTCCACATTTGGGGCCAGAGATGGTATCAGAAGGAATTTGCCCAGG 1920
AAATGACCTTTAATATCAGTAGCAGCAGCCGAGATCCCGAGGTGTGCTTGGACCTACGTC 1980
CGGGTACCAACTACAATGTCAGTCTCCGGGCTCTGTCTTCGGAACTTCCTGTGGTCATCT 2040
CCCTGACAACCCAGATAACAGAGCCTCCCCTCCCGGAAGTAGAATTTTTTACGGTGCACA 2100
GAGGACCTCTACCACGCCTCAGACTGAGGAAAGCCAAGGAGAAAAATGGACCAATCAGCA 2160
ACGCCTCTGATGCTGATGGATACGTGGCTGCAGAACTACTGGCCAAAGATGTTCCAGATG 2220
ATGCCATGGAGATACCTATAGGAGACAGGCTGTACTATGGGGAATATTATAATGCACCCT 2 80
TGAAAAGAGGGAGTGATTACTGCATTATATTACGAATCACAAGTGAATGGAATAAGGTGA 2340
GAAGACACTCCTGTGCAGTTTGGGCTCAGGTGAAAGATTCGTCACTCATGCTGCTGCAGA 400
TGGCGGGTGTTGGACTGGGTTCCCTGGCTGTTGTGATCATTCTCACATTCCTCTCCTTCT 2460 CAGCGGTGTGATGGCAGATGGACACTGAGTGGGGAGGATGCACTGCT
The disclosed NON9 nucleic acid sequence, localized to chromosome 9, has 1747 of 1747 bases (100%) identical to a gb:GEΝBAΝK-ID:AX050019|acc:AX050019.1 mRNA from Homo sapiens (Sequence 32 from Patent WO0071710) (Fig. 3A). The full amino acid sequence of the protein of the invention was found to have 440 of 441 amino acid residues (99%) identical to, and 441 of 441 amino acid residues (100%) similar to, the 570 amino acid residue ptnr:SPTREMBL-ACC:Q9NTA7 protein from Homo sapiens (Human) (HYPOTHETICAL 63.7 KDA PROTEIN).
The disclosed NOV9 polypeptide (SEQ ID NO.28) encoded by SEQ ID NO:27 has 720 amino acid residues and is presented in Table 9B using the one-letter amino acid code. Signal P, Psort and/or Hydropathy results predict that NON9 is a Type la membrane protein, has a signal peptide, and is likely to be localized at the plasma membrane with a certainty of 0.9190. In other embodiments, ΝON9 may also be localized to the endoplasmic reticulum (membrane) with a certainty of 0.100, and lysosomes with a certainty of 0.2000. The most likely cleavage site for NON9 is between positions 29 and 30, AAG-AP.
Table 9B.
ΝOV9 Polypeptide
SEQ ID NO:28
MGRGPWDAGPSRRLLPLLLLLGLARGAAGAPGPDGLDVCATCHEHATCQQREGKKICICN 60
YGFVGNGRTQCVD NECQFGATLVCGNHTSCH TPGGFYCICLEGYRATNNNKTFIPNDG 120
TFCTDIDECEVSGLCRHGGRCVNTHGSFECYCMDGYLPRNGPEPFHPTTDATSCTEIDCG 180
TPPEVPDGYIIGNYTSSLGSQVRYACREGFFSVPEDTVSSCTGLGT ESPKLHCQEINCG 240
NPPEMRHAILVGNHSSRLGGVARYVCQEGFESPGGKITSVCTEKGTWRESTLTCTEILTK 300
INDVSLFNDTCVR QINSRRINPKISYVISIKGQRLDPMESVREETVNLTTDSRTPEVCL 360
ALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGSFNISIFNETCLKLNRRSRKV 420
GSEHMYQFTVLGQR YLAMFSHATSFNFTTREQVPWCLDLYPTTDYTVNVTLLRSP RH 480
SVQITIATPPAVKQTISNISGFNETCLRWRSIKTADMEEMYLFHIWGQRWYQKEFAQEMT 540
FNISSSSRDPEVCLDLRPGTNYNVSLRALSSELPWISLTTQITEPPLPEVEFFTVHRGP 600
LPRLRLRKAKEKNGPISNASDADGYVAAELLAKDVPDDA EIPIGDRLYYGEYYNAPLKR 660
GSDYCIILRITSE NKVRRHSCAV AQVKDSSLMLLQMAGVGLGSLAWIILTFLSFSAV 720
A search of sequence databases reveals that the NON9 amino acid sequence has 440 of 441 amino acid residues (99%) identical to, and 441 of 441 amino acid residues (100%) similar to, the 570 amino acid residue ρtnr:SPTREMBL-ACC:Q9ΝTA7 protein from Homo sapiens (Human) (HYPOTHETICAL 63.7 KDA PROTEIN).
NON9 is expressed in at least the pancreas, placenta, nervous system, tumor tissues, brain and the hypothalamus.
The disclosed ΝON9 polypeptide has homology to the amino acid sequences shown in the BLASTP data listed in Table 9C.
Figure imgf000110_0001
Figure imgf000111_0003
The homology between these and other sequences is shown graphically in the ClustalW analysis shown in Table 9D. hi the ClustalW alignment of the NOV9 protein, as well as all other ClustalW analyses herein, the black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties), whereas non-highlighted amino acid residues are less conserved and can potentially be altered to a much broader extent without altering protein structure or function.
Table 9D. ClustalW Analysis of NOV9
1) NOV8 (SEQ ID NO: 5)
2) NOV9 (SEQ ID N0.27)
3) gi|ll360234| (SEQ ID NO:69)
4) gi|l7402220| (SEQ ID NO.70)
5) gi|l6552183| (SEQ ID NO: 71)
6) gi|l4740162| (SEQ ID NO: 72)
7) gl|l0438017| (SEQ ID Nθ:73)
10 20 30 40 50
NOV8 MGRGPWDAGPSRR LPL LLLGLARGAAGAPGPDG DVCATCHEHATCQQ
NOV9 MGRGPWDAGPSRRLLP LL LGLARGAAGAPGPDG DVCATCHEHATCQQ
Figure imgf000111_0001
gi|l0438017|
60 70 80 90 100
NOV8 REGKKICICNYGFVGNGRTQCVDKNECQFGATLVCGNHTSCHNTPGGFYC
NOV9 REGKKICICNYGFVGNGRTQCVD NECQFGATLVCGNHTSCHNTPGGFYC
Figure imgf000111_0002
gi] 10438017 I
110 120 130 140 150
NOV8 ICLEGYRATNNNKTFIPNDGTFCTDIDECEVSGLCRHGGRCVNTHGSFEC NOV9 ICLEGYRATNNNKTFIPNDGTFCTDIDECEVSGLCRHGGRCVNTHGSFEC gi|ll360234| gi 117402220 I ECEVSGLCRHGGRCVNTHGSFEC
gi] 14740162 I
160 170 180 190 200
NOV8 YCMDGYLPRNGPEPFHPTTDATSCTEI
NOV9 YCMDGYLPRNGPEPFHPTTDATSCTEIg gi 111360234 I DCGTPPEVPDGYIIGNYTSSLGS gi ] 17402220 I YCMDGYT.,PRNrGPEPFHPTTDATSC FlllJ».ell>.JJak*i5.i!ri-'i*fe.i lbfct<cfeJ
Figure imgf000112_0001
gi ] 10438017 I
210 220 230 240 250
.■ ■.| ....|....]----l--.-l ----l----l----]..--l ----l
Figure imgf000112_0002
NOV9 gi|ll3602 4| gi|l740222θ] gi|l6552183] gi|l4740162| gi] 10438017]
260 270 280 290 300
NOV8
N0V9 tfrii.i^fI<e.tVJrW «tJtt^^Jt.tiiJι^Wιl^i^ι.i?^^JAJ ιj^^^iJ gi|ll360234| ΓGNHSSRLGGVARYVCOEGFEΞPGGKITΞVCTEKGTWRESTLTCTEΪLT
Figure imgf000112_0003
gi|l4740162| ΓGNHSSRLGGVARYVCOEGFESPGGKITSVCTEKGTWREST TCTEΪLTI gi| 10438017 I
310 320 330 340 350
NOV8 ι^ι.tf/ -iaii.iιlfwti;wfrt<^-.:j:*^>id:*^--t««.-**:w.)atiiiaairta*iaDtBtι>-H^ftι
N0V9 gi|ll360234] NDVSLFNDTCVRWOINSRRINPKISYVIΞIKGORLDPMESVREETV LT gi 117402220 I u BtΛiιaan>mwaι.»MiiB-af«i!.ai««8t8tf«-BMS XMWMIKiflMsHAUatn, gi|l6552183| NDVSLFNDTCVRWOINSRRINPKISYVISIKGQRLDPMESVREETVNL gi|l4740162| ■<i)ιrtt<.-««la>ι.lι^tj:lW.ll*ik.i:*ti.>di*t--'«<ι«J.«iW.)a»lMafll^t'<:^^ιtt<{l|ftι gi|l0438017| -Ws-tøda.ali.kfai.l.J
360 370 380 390 400
NOV8 ΓDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS
N0V9 ΓDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi| 11360234 I TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l7402220| TDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi|l6552183| ΓDSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDGS gi 114740162 I .DSRTPEVCLALYPGTNYTVNISTAPPRRSMPAVIGFQTAEVDLLEDDG gi|l0438017|
410 420 430 440 450
NOV8
NOV9 ιaMta«aMaιιι>t>c«fiMa:ιaaa<waaiisrtfi5gBiiw«κMir«ιrg gi| 11360234 I NISI FNETC KLNRRSRKVGSEHMYQFTVLGOR YLANFSHATSFNFT gi|l740222θ| Mj«-B«-wjiMiι«ιιi!«ιiiiarMacwieιaa:ιi3i_ι«ιaιitfjιiH»ι w<ιra^ιaMii»viBta ιaiMi gi| 16552183 I 'NISIFNETCLK NRRSRKVGSEHM rirtaW ftfltlaMifeViMamtalnln
gi|l0438017]
460 470 480 490 500
NOV8 S^Ξ^raRπra3SSiraro 3Ξ^SΞ3!^SS3^33^^El^iE
NOV9 i.wmawflffl »3gaBaaπwai5M8 gi|ll360234| lEOVPWCLDLYPTTDYTVNVTLLRSPKRHSVOITIATPPAVKOTISNIi gi|l7402220| irwmawrti EMagasaaiwa -ft gi|l6552183| gi| 14740162 I irtaftMa Mf mtoaaaj mnnHHiiMs gi|l0438017| l:l^.l 1t,^^*.^'<_,U.:'^ .^^I,:I: I<^l:itJ.l,>L^. -J-^ ..^J:^ :
510 520 530 540 550
NOV8 .FNETCLRWRSIKTADMEEMYLFHIWGORWYOKEFAQEMTFNISSSSRDI
NOV9 MaaιMι«ι.ara.M»!«tt»]fli«:Ha«n «^^^^^
Figure imgf000113_0001
gi|l740222θ| .FNETCLRWRSIKTADMEEMYLFHH* gi| 16552183 I gi| 14740162 j .W^^IMdJ_UJUi «iW^ gi|10438017| 5FNETC RWRSIKTADMEEMY FHIWGQRWYQKEFAQE TFNISSSSRDI
560 570 580 590 600
NOV8 /C DLRPGTNYNVSLRALS SE PVi waaιιiιιftMn-aaaι.3 NOV9 gi|H360234| VCLDLRPGTNYNVSLRALSSELPWTΞLTTOITEPPLPEVEFFTVHRG gi| 17402220 I tw<).m;jatD»^-^tt/.oM<>.-Ma<.aw<<Hfturtiι<, ^aanaatJ5.^aι>.t<ilart5
gi 114740162 I iatfttιiiπιagei w«ttfjrti)fei<waaιιaw<»aιιι ι»ι»ι aιaaw wjaiaaiιιttiiaeii gi| 10438017 j iaf«**rt«t:ta^ι.^'?^frt«ι»iWW;.a.<.at.t*«;tftιι.W«ιιJD_aafl;>aia.3J^ιtiiι;W;
610 620 630 640 650
NOV8 LPRLRLRKAKEKNGPISSYQVLVLPLALQSTFSCDSEGASSFFSNASDAD
NOV9 LPRLRLRKAKEKNGPIS gi 111360234 I LPRLRLRKAKEKNGPISSYQVLVLPLALQSTFSCDSEGASSFFSNASDAD gi|l7402220| LPRLRLRKAKEKNGPISSYQVLVLPLALQSTFSCDSEGASSFFSNASDAD gi| 16552183 I .PRLRLRKAKEKNGPISSYQVLVLP ALQSTFSCDSEGASSFFSNASDAD gi|l4740162| LPRLRLRKAKEKNGPISSYQVLVLPLALQSTFSCDSEGASSFFSNASDAD gi|l0438017| LPRLRLRKAKEKNGPISSYQVLVLPLALQSTFSCDSEGASSFFSNASDAD 660 670 680 690 700
NOV8 GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE NOV9 GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE gi|ll360234| GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE gi 117402220 I 3YVAAELLAKDVPDDAMEIPIGDR YYGEYYNAPLKRGSDYCIILRITSE gi 116552183 I GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIIIiRITSE gi| 14740162 I GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE gi| 10438017 I GYVAAELLAKDVPDDAMEIPIGDRLYYGEYYNAPLKRGSDYCIILRITSE
710 720 730 740
NOV8 WNKVRRHSCAVWAQVKDSSLMLLQMAGVGLGSLAWIILTFLSFSAλ
NOV9 MNKVRRHSCAVWAQVKDSS MLLQMAGVGLGSLAWIILTFLSFSA\ gi| 11360234 I WNKVRRHSCAVWAQVKDSS MLLQMAGVGLGSLAWIILTFLSFSA gi|l7402220| WNKVRRHSCAVWAQVKDSS MLLQMAGVGLGSLAWIILTFLSFSAV gi|l6552183| [W5ϊlϊl-l35BlccRV^KLfi8 vpwflBI- gi] 14740162 I WNKVRRHSCAVWAQVKDSS MLLQMAGVGLGSLAWIILTFLSFSA gi|l0438017| WNKVRRHSCAVWAQVKDSSLMLLQ AGVGLGSLAWIILTFLSFSA
Table 9E lists the domain description from DOMAIN analysis results against NOV9. This indicates that the NON9 sequence has properties similar to those of other proteins known to contain this domain.
Table 9E. Domain Analysis of ΝON9 gnl|Smart|smart00179, EGF_CA, Calcium-binding EGF-like domain
CD-Length = 41 residues, 80.5% aligned Score = 52.8 bits (125), Expect = 7e-08
Query: 125 DIDECEVSGLCRHGGRCWTHGSFΞCY-CMDGY 156
DIDEC C++GG CVKTT GS+ C C GY Sbjct: 1 DIDECASGΝPCQΝGGTCWTVGSYRCEECPPGY 33
The disclosed ΝON9 polynucleotide encodes a Type la Membrane Sushi-Containing Domain-like protein, identified by the comparative sequencing of human chromosome 1 lpl5 and mouse chromosome 7. This gene contains two very important domains associated with developmental proteins- the CUB domain and the domain first found in Clr, Cls, uEGF, and bone morpho genetic protein. The CUB domain is found in 16 functionally diverse proteins such as the dorso-venfral patterning protein tolloid, bone moφhogenetic protein 1, a family of spermadhesins, complement subcomponents Cls/Clr and the neuronal recognition molecule A5. Most of these proteins are known to be involved in developmental processes. The second domain is found mostly among developmentally-regulated proteins and spermadhesins. The disclosed NON9 nucleic acid of the invention encoding a Type la membrane sushi-containing domain-like protein includes the nucleic acid whose sequence is provided in Table 9A, or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 9A while still encoding a protein that maintains its a Type la membrane sushi-containing domain-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonUmiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
The disclosed ΝON9 protein of the invention includes the Type la membrane sushi- containing domain-like protein whose sequence is provided in Table 9B. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 2 while still encoding a protein that maintains its a Type la membrane sushi-containing domain-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 1 percent of the residues may be so changed.
The invention further encompasses antibodies and antibody fragments, such as Fab or (Fab)2, that bind immuno specifically to any of the proteins of the invention.
The above defined information for this invention suggests that this Type la membrane sushi-containing domain- like protein (ΝON9) may function as a member of a family. Therefore, the ΝON9 nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The ΝON9 nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in liver toxicity and damage such as in cancer, cirrhosis, or troglitazone treatment for diabetes; brain and CNS disorders including cancer, Parkinson's, Alzheimer's, epilepsy, schizophrenia and other diseases, disorders and conditions of the like. For example, a cDNA encoding a Type la membrane sushi-containing domain-like protein (NON9) may be useful in gene therapy, and the Type la membrane sushi-containing domainlike protein (ΝON9) may be useful when administered to a subject in need thereof. By way of nonlimiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from cancer, diabetes, obesity, fertility as well as other diseases, disorders and conditions. The ΝON9 nucleic acid encoding a Type la membrane sushi- containing domain-like protein, and the a Type la membrane sushi-containing domain-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
ΝON9 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel ΝON9 substances for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti-ΝOVX Antibodies" section below. The disclosed ΝOV9 protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOV9 epitope is from about amino acids 40 to 300. In another embodiment, a NOV9 epitope is from about amino acids 305 to 360, from about 400 to 450, from about 500 to 560, from about 580 to 610, and from about 620 to 680. These novel proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
NOV10
NON 10 includes two butyrophilin-like proteins disclosed below. The disclosed sequences have been named ΝONlOa and ΝONlOb.
NOVlOa
A disclosed ΝONlOa nucleic acid of 861 nucleotides identified as SEQ ID ΝO:29 (designated CuraGen Ace. No. CG55746-01 ) encoding a novel butyrophilin-like protein is shown in Table 10A. An open reading frame was identified beginning with an ATG initiation codon at nucleotides 46-48 and ending with a TGA codon at nucleotides 793-795. Putative untranslated regions, if any, are found upstream from the initiation codon and downstream from the termination codon. Table 10A
NONlOa Polynucleotide
SEQ ID ΝO:29
CAGGTTACACTTCGTAAGAACTGGAATGTAAAGTAAAGGCAGACAATGACAAAATATCTTGTTTTCTTTT CAGCTTTATTCACAGTGACAGTCCCTAAGCACCTGTACATAATAAAGCACCCCAGCAATGTGACCCTGGA ATGCAACTTTGACACTGGTAGTCATGTGAACCTTGGAGCAATAACAGTCAGTTTGCAAAAGGTGGAAAAT GATACATCCCCACACCGTGAAAGAGCCACTTTGCTGGAGGAGCAGCTGCCCCTAGGGAAGGCCTCGTTCC ACATACCTCAAGTCCAAGTGAGGGACGAAGGACAGTACCAATGCATAATCATCTATGGGGTCGCCTGGGA CTACAAGTACCTGACTCTGAAAGTCAAAGGTGCTTCCTACAGGAAAATAAACACTCACATCCTAAAGGTT CCAGAAACAGATGAGGTAGAGCTCACCTGCCAGGCTACAGGTTATCCTCTGGCAGAAGTATCCTGGCCAA ACGTCAGCGTTCCTGCCAACACCAGCCACTCCAGGACCCCTGAAGGCCTCTACCAGGTCACCAGTGTTCT GCGCCTAAAGCCACCCCCTGGCAGAAACTTCAGCTGTGTGTTCTGGAATACTCACGTGAGGGAACTTACT TTGGCCAGCATTGACCTTCAAAGTAAGATGGAACCCAGGACCCATCCAACTTGGCTGCTTCACATTTTCA TCCCCTTCTGCATCATTGCTTTCATTTTCATAGCCACAGTGATAGCCCTAAGAAAA.CAACTCTGTCAAAA GCTGTATTCTTCAAAAGGTAAGTGAGTTTTATTCATGGTAACCCAATGCACTGGGTGTCTGCAGCATGAG CCACTGCTTTGCACTGCAGGC
In a search of public sequence databases, the NOVlOa nucleic acid sequence, which maps to chromosome 9, and has 467 of 473 bases (98%) identical to a gb:GENBANK- ID:AK001872|acc:AK001872.1 mRNA from Homo sapiens (Homo sapiens cDNA FLJllOlO fis, clonePLACE1003145).
The disclosed NONlOa polypeptide (SEQ ID ΝO.30) encoded by SEQ ID NO:29 has 249 amino acid residues and is presented in Table 10B using the one-letter amino acid code. Signal P, Psort and/or Hydropathy results predict that NONlOa is a Type la membrane protein, has a signal peptide, and is likely to be localized at the plasma membrane with a certainty of 0.4600. In other embodiments, ΝONlOa may also be localized to the endoplasmic reticulum (membrane) with a certainty of 0.3700, and lysosomes with a certainty of 0.3000. The most likely cleavage site for ΝON8 is between positions 17 and 18, TNP-KH.
Figure imgf000117_0001
A search of sequence databases reveals that the NOVlOa amino acid sequence has 159 of 231 amino acid residues (68%) identical to, and 182 of 231 amino acid residues (78%) similar to, the 247 amino acid residue ptnr: SPTREMBL-ACC.Q9WUL5 protein from Mus musculus (Mouse) (BUTYROPHILIN-LIKE PROTEIN)
NONlOa is expressed in at least Bone Marrow, Lung, Testis, Thymus, Uterus, Whole Organism. NONlOb
A disclosed ΝON1 Ob nucleic acid of 660 nucleotides identified as SEQ ID ΝO:31 (designated CuraGen Ace. No. CG55746-05) encoding a novel BUTYROPHILIN PRECURSOR B7-DC-like protein is shown in Table IOC. An open reading frame was identified beginning at nucleotides 34-36 and ending at nucleotides 583-585. Putative untranslated regions are indicated by underline.
Table IOC
NOVl Ob Polynucleotide
SEQ ID NO;31
AGCTGTGGCAAGTCCTCATATCAAATACAGAACATGATCTTCCTCCTGCTAATGTTGAGC 60
CTGGAATTGCAGCTTCACCAGATAGCAGCTTTATTCACAGTGACAGTCCCTAAGGAACTG 120
TACATAATAGAGCATGGCAGCAATGTGACCCTGGAATGCAACTTTGACACTGGAAGTCAT 180
GTGAACCTTGGAGCAATAACAACCAGTTTGCAAAAGGTGGAAAATGATACATCCCCACAC 240
CGTGAAAGAGCCACTTTGCTGGAGGAGCAGCTGCCCCTAGGGAAGGCCTCGTTCCACATA 300
CCTCAAGTCCAAGTGAGGGACGAAGGACAGTACCAATGCATAATCATCTATGGGGTCGCC 360
TGGGACTACAAGTACCTGACTCTGAAAGTCAAAGGTCAGATGGAACCCAGGACCCATCCA 420
ACTTGGCTGCTTCACATTTTCATCCCCTCCTGCATCATTGCTTTCATTTTCATAGCCACA 80
GTGATAGCCCTAAGAAAACAACTCTGTCAAAAGCTGTATTCTTCAAAAGACACAACAAAA. 540
AGACCTGTCACCACAACAAAGAGGGARGTGAACAGTGCTATCTGAACCTGTGGTCTTGGG 600
AGCCAGGGTGACCTGATATGACATTTAAAGAAGCTTCTGGACTCTGAACAAGAATTCGGT 660
In a search of public sequence databases, the NON 10b nucleic acid sequence, which maps to chromosome 9, has 394 of 396 bases (99%) identical to a gb.GEΝBAΝEC- ID:AF329193|acc:AF329193.1 mRΝA from Homo sapiens (Homo sapiens butyrophilin precursor B7-DC mRΝA, complete eds).
The disclosed ΝONlOb polypeptide (SEQ ID ΝO.32) encoded by SEQ ID NO:31 has 183 amino acid residues and is presented in Table 10D using the one-letter amino acid code. Signal P, Psort and/or Hydropathy results predict that NON 10b is a Type II membrane protein, has a signal peptide, and is likely to be localized to the mitochondrial inner membrane with a certainty of 0.8463. In other embodiments, ΝOVlOb may also be localized to the plasma membrane with a certainty of 0.4400, mitochondrial intermembrane space with a certainty of 0.3008, and mitochondrial matrix space with a certainty of 0.2317. The most likely cleavage site for ΝOVlOb is between positions 19 and 20, IAA-LF.
Table 10D
ΝONlOb Polypeptide
SEQ ID ΝO:32
MIFLLLMLSLELQLHQIAALFTVTVPKELYIIEHGSNVTLECNFDTGSHVNLGAITTSLQ 60
KVENDTSPHRERATLLEEQLPLGKASFHIPQVQVRDEGQYQCIIIYGVAWDYKYLTL VK 120
GQ EPRTHPTWLLHIFIPSCIIAFIFIATVIALRKQLCQKLYSSKDTTKRPVTTTKREV 180 SAI A search of sequence databases reveals that the NOVl Ob amino acid sequence has 121 of 129 amino acid residues (93%) identical to, and 121 of 129 amino acid residues (93%) similar to, the 273 amino acid residue ptnr:SPTREMBL-ACC:Q9BQ51 protein from Homo sapiens (Human) (butyrophilin precursor B7-DC (PD-1 -ligand 2 protein)).
NOVl OB, the butyrophilin precursor B7-DC -like gene disclosed in this invention is expressed in at least the following tissues: Bone Marrow, Lung, Testis, Thymus, Uterus, Whole Organism.
The disclosed NOVl Ob polypeptide has homology to the amino acid sequences shown in the BLASTP data listed in Table 10E.
Figure imgf000119_0001
The homology between these and other sequences is shown graphically in the ClustalW analysis shown in Table 10F. In the ClustalW alignment of the NOVlOa and NOVl 0b proteins, as well as all other ClustalW analyses herein, the black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties), whereas non-highlighted amino acid residues are less conserved and can potentially be altered to a much broader extent without altering protein structure or function.
Table 10F. ClustalW Analysis of NOVl OA
1 ) NOVlOa (SEQ ID NO : 29) 2) NOVlOb (SEQ ID NO: 31)
3) gi|l3640665| (SEQ ID NO:74)
4) gi|l337685θ| (SEQ ID NO:75)
5) gi|l0946740] (SEQ ID NO:76)
6) gi|7661534| (SEQ ID Nθ:77)
7) gi|ll230798| (SEQ ID NO:78)
10 20 30 40 50
NOVl0 BrκϊJt Ε ^JI^ιtfι iJifeHHιικapli.!ι^iιi-Jla_«lNi5BlτGSHivi
NOVlOb |]EiFi Λ&s ELQi-HQ _^3iiS_B3 lEι]i^SS3^β'rGSH gi|l3640665| |Jj]lFL!ϊ»έϊiE QLHQ$ g3iΞ-SSβΣ?]iβS!_S®ffl,I'Gstξi gi|l3376850] J5JlF J!|^tt.SIJELQLHQΪ SSiΞ^SE2Sll|K^J^Ϊ[^3J33 GSI^
Figure imgf000120_0001
gil 11230798] UJ] IF ΪFT - - r.c Aτ, !^ι^i35pilBv^5MgϊBϊBit^SpiR|^ vEREiJ
60 70 80 90 100
NOVlOa
Figure imgf000120_0002
WfcMJ*l*ιtlidBUMfcMM
NOVlOb UjGg3ττaSSH
Figure imgf000120_0003
gi|l3640665| tsMJάΛ UΛΛΛMMimti
Figure imgf000120_0004
gi] 11230798 I ^Ij fή V ESSE^EQ IQFV GEEDLKPQHg FgG^SJΪPfcC^ΪLKg-φ.
110 120 130 140 150
NOVlOa mtVii' iiHtaBSG sSSjRiaBiTHBi- κv§
NOVlOb sMEM gi|l3640665| 3QBSSSSESE Mt&ΨMhMmSS 3s- | H3J-K @ gi|l3376850| JTHgjJ-K JS
Figure imgf000120_0005
gi]7661534| ιsB88-flftliBSR--5M8fe]3 - iQ SSwDg gi|l!230798| LQS DSκtoJ3AggcHi3sE-GAS^Riϊ3EB S -E BJQRØS - VD§
160 170 180 190 200
■■■■i....|....ι....i-...|....|....|....|....ι....ι
Novioa
NOVlOb gi 113640665 |
Figure imgf000120_0006
Figure imgf000120_0007
Figure imgf000120_0008
210 220 230 240 250
Figure imgf000120_0009
gi | l337685θ | Pf^l pP GRNf^sgS f^THV T-^S_rDI_OSθ ^RTH]S^I^Ii3πiaS gi ] 10946740 ]
Figure imgf000121_0001
gi | 7661534 | [JErNTTTNEIJ3γSτ§RR DPE|NHT|EIjV p -JSI.PLAHPgNERTJ$tjV0LG gi 1 11230798 ] |J^ATA D ]3τHRSQPGQNHTgEl ipp-SljPATHPgQNRTB! f G
260 270 280 290
NOVlOa lιlι-V_V-lalι-.3lιltIgiaB3aoι1co!ιtϊ-Ysst5Gκ
NOVl Ob lil-li^J-.all TBBA(1l350LiC0kliYSSigDTTKRPVTMTKREVNSAI - - gi 1 13640665 | ! *^alιj3l.igτCTIaHI35θI.COK:≤YSSl!SDTTK PVτgτK EVNSAI - - gi 1 13376850 | («lιJiW^ ^EM^HI3i^CO ≤ΥSsBbτTKR Vlft',TKRE NSAI - - gi 1 10946740 I STELjEsilESlQSiK* gi I 7661534 | A0I_LCΪιGyf LTFIFJ^LJSlKG-jaMMDViSKCGIQDTN^KKjgSDTHLEET gi 1 11230798 | sgϊ, i ϊ.l|vVgτVL3FLrfKQ iEHLD ^KCGVEDTS^K ΪΪNDTQFEET
Tables 10G lists the domain description from DOMAIN analysis results against NOV10. This indicates that the NOV10 sequence has properties similar to those of other proteins known to contain this domain.
Table 10G Domain Analysis of NOV10 gnllSmart|smart00409, IG, Immunoglobulin
CD-Length = 86 residues, 89.5% aligned Score = 37.0 bits (84), Expect = 0.001
Query: 27 SNVTLECNFDTGSHVNLGAITVSLQKVENDTSPHRERATLLEEQLPLGKΑSFHIPQVQVR 86
+VTL C TV+ K R ++ G ++ I V Sbjct: 10 ESVTLSCEASGNPPP TVTWYKQGGKLLAESGRFSVSRSG GNSTLTISNVTPE 61
Query: 87 DEGQYQCIIIYGVAWDYKY TLKVK 111
D G Y C TL V
Sbjct: 62 DSGTYTCAATNSSGSASSGTTLTV 86
The gene sequence of invention described herein encodes for a novel member of the B7-Immunoglobulin family of enzymes. Specifically, the sequence encodes a novel BUTYROPHILIN-like protein. BUTYROPHILIN molecules play crucial roles in T-cell activation making them plausible targets for cancer, ADDS, and inflammation therapies. The protein described here is known to be expressed in spleen, and liver which may indicate roles in lupus, endocrine disorders, inflammation, autoimmune disorders, and cancers including liver, bone, and leukemia.
The disclosed NOV10 nucleic acid of the invention encoding a butyrophilin-like protein includes the nucleic acid whose sequence is provided in Table 10 or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 10 while still encoding a protein that maintains its butyrophilin-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject, hi the mutant or variant nucleic acids, and their complements, up to about 2 percent (NOVlOa) or 1% (NOVlOb) of the bases may be so changed.
The disclosed NOVIO protein of the invention includes the butyrophilin-like protein whose sequence is provided in Table 10A and IOC. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 10B while still encoding a protein that maintains its butyrophilin-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 32 percent (NOVlOa) or (NOVlOb) of the residues maybe so changed.
The invention further encompasses antibodies and antibody fragments, such as Fa or (Fab that bind immunospecifically to any of the proteins of the invention.
The above defined information for this invention suggests that this butyrophilin-like protein (NOVIO) may function as a member of a butyrophilin family. Therefore, the NOVIO nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The NOVIO nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in cancer including but not limited to various pathologies and disorders as indicated below. For example, a cDNA encoding the butyrophilin-like protein (NOVIO) may be useful in gene therapy, and the butyrophilin-like protein (NOVIO) may be useful when administered to a subject in need thereof. By way of nonlimiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from : brain disorders including epilepsy, eating disorders, schizophrenia, ADD, and cancer; heart disease; inflammation and autoimmune disorders including Crohn's disease, IBD, lupus, allergies, rheumatoid and osteoarthritis, inflammatory skin disorders, blood disorders; psoriasis colon cancer, leukemia AIDS; thalamus disorders; metabolic disorders including diabetes and obesity; lung diseases such as asthma, emphysema, cystic fibrosis, and cancer; multiple sclerosis, pancreatic disorders including pancreatic insufficiency and cancer; and prostate disorders including prostate cancer and other diseases, disorders and conditions of the like.
The NOVIO nucleic acid encoding the butyrophilin-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOVIO nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel NOVIO substances for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti- NOVX Antibodies" section below. The disclosed NOVlOa protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOVlOa epitope is from about amino acids 25 to 40. In another embodiment, a NOVlOa epitope is from about amino acids 50 to 70, from about 60 to 100, from about 110 to 140, from about 145 to 185, and from about 190 to 210. The disclosed NOVlOb protein has multiple hydrophilic regions, each of which can be used as an immunogen. In one embodiment, a contemplated NOVlOb epitope is from about amino acids 50 to 80. In another embodiment, a NOVlOb epitope is from about amino acids 80 to 110, from about 111 to 130, and from about 150 to 175.
These novel proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
NOV11
A disclosed NOVl 1 nucleic acid of 1115 nucleotides identified as SEQ ID NO:33 (also designated as Ace. No. CG50329-01) encoding a novel BUTYROPHILIN-LIKE PROTEIN-like protein is shown in Table 11 A. An open reading frame was identified begimiing with an ATG initiation codon at nucleotides 46-48 and ending with a TAA codon at 931-933.
Table 11 A.
NOV11 Polynucleotide
SEQ ID NO:33
AACCTGCTCTGAGGGGTGGGGAGAAAGACCCCATCACCTGCTAGGATGAGCAGAGCGTGGGGCGATGCAG TCATTCCCTCACTGTCCGTGCTCCGCTCATTCATTCATCTCCTTGAACTCCTGACCTCAGGCAATGGGAA AGCTGACTTTGATGTCACTGGGCCTCATGCCCCTATTCTGGCTATGGCTGGGGGACACGTGGAGTTACAG TGCCAGCTGTTCCCCAATATCAGTGCCGAGGACATGGAGCTGAGGTGGTACAGGTGCCAGCCCTCCCTAG CTGTGCACATGCATGAGAGAGGGATGGACATGGATGGAGAGCAAAAGTGGCAGTACAGAGGAAGGACCAC CTTCATGAGTGACCACGTGGCCAGGGGCAAGGCCATGGTGAGGAGTCACAGGGTCACCACCTTTGACAAC AGGACATACTGCTGCCGCTTCAAGGATGGTGTAAAGTTCGGCGAGGCCACTGTGCAGGTGCAGGTGGCAG GTAAGTCAGGGCTGGGCAGAGAGCCCAGAATCCAGGTGACAGACCAGCAGGATGGAGTCAGGGCGGAGTG CACATCAGCAGGCTGTTTCCCCAAGTCCTGGGTGGAACGGAGAGACTTCAGGGGCCAGGCTAGGCCTGCT GTGACCAATCTATCAGCCTCAGCCACCACCAGGCTCTGGGCTGTGGCATCCAGCTTGACGCTCTGGGACA GGGCTGTGGAGGGTCTCTCCTGCTCCATCTCCAGCCCCCTCCTCCCTGAAAGGTCAGTTTCAGGCATCCA CTGGGGGTCATGGAATGTATCCCCCAAGGACAAGGGGGGCTTATTAGAGTCACACTCTGAGGTCCTGGGG TTAGAACTTCAACAGATGACTGGGGGGCAGGGGATACAAAATGGAACCCATAACAATTCTCAAAATGCTT TTTCCTCAAACCTGAAAGTGTAAAACCTGCTCTGAGGGGTGGGGAGAAAGACCCCATCACCTGCTAGGAT GAGCAGAGCGTGGGGCGATGCAGTCATTCCCTCACTGAAGACATTTATGGGGCACCTCCCTATGCACGAG ACAGGAAGGAAGGAATTACAGAAACAAAACCTCACAAATATATACAATTATTACGTGTTAATTAA
In a search of public sequence databases, the NOVl 1 nucleic acid sequence, located on chromosome 1 has 508 of 780 bases (65%) identical to a gb.GENBANK- ID:AF269232|acc:AF269232.1 mRNA from Mus musculus (Mus musculus butyrophilin-like protein BUTR-1 (Butrl) mRNA, complete eds) .
The NOV 11 protein (SEQ ID NO:34), encoded by SEQ ID NO:33, has 295 amino acids. Signal P, Psort and/or Hydropathy results predict that NOVl 1 is a Type la membrane protein, has a signal peptide, and is likely to be localized to the mitochondrial matrix space with a certainty of 0.6797. In other embodiments, NOVl 1 may also be localized to the microbodies with a certainty of 0.4023, mitochondrial inner membrane with a certainty of 0.3682, mitochondrial inner membrane space with a certainity of 0.3682. The most likely cleavage site for NOVl 1 is between positions 31 and 32, GNG-KA.
Figure imgf000124_0001
A search of sequence databases reveals that the NOVl 1 amino acid sequence has 140 of 274 amino acid residues (51%) identical to, and 185 of 274 amino acid residues (67%) similar to, the 275 amino acid residue ptnr:SPTREMBL-ACC:Q9JK39 protein from Mus musculus (Mouse) (BUTYROPHILIN-LIKE PROTEIN BUTR-1)
The disclosed NOVl 1 polypeptide has homology to the amino acid sequences shown in the BLASTP data listed in Table 1 lC.
Figure imgf000125_0001
The homology between these and other sequences is shown graphically in the ClustalW analysis shown in Table 1 ID. In the ClustalW alignment of the NOVl 1 protein, as well as all other ClustalW analyses herein, the black outlined amino acid residues indicate regions of conserved sequence (i.e., regions that may be required to preserve structural or functional properties), whereas non-highlighted amino acid residues are less conserved and can potentially be altered to a much broader extent without altering protein structure or function. Table 11D. ClustalW Analysis of NON11
1) ΝOV11 (SEQ ID Ν0.33)
2) gi|8101125| (SEQ ID N0.79)
3) gi|l674173θ| (SEQ ID NO.80)
4) gi I 59214611 (SEQ ID NO: 81)
5) gi|l4751898| (SEQ ID NO:82)
6) gi| 17028375 I (SEQ ID NO: 83)
10 20 30 40 50
NOV11 EsRS -GDAVIJ3si^V RSFΪH[gEjJ]_,TSGNGK DJ|D2τ^HAajJ^MaS gi| 8101125] |ARSHPGDAglS--ilLVSFj-F3Q2 TSGNG §Di 2ΪSΞPHSl5S!IE gi|l6741730|
Figure imgf000126_0001
Figure imgf000126_0002
gi 114751898 I gi|l7028375|
60 70 80 90 100
NOVll
Figure imgf000126_0003
gi|8101125| g KEJJpgKJl LNIS^GBlSKSD^PS5' E3. gi|l6741730| -NTTLRCHLSPEKNAEDMEVRWFRSQFSPAVFVYKGGRERTEEQMEEYRG
Figure imgf000126_0004
gi 114751898 I NTTLRCHLSPEKNAEDMEVRWFRSOFSPAVFVYKGGRERTE ftlflMaWsB gi|l7028375] ENTTLRCHLSPEKNAEDMEVRWFRSOFSPAVFVYKGGRERTI
110 120 130 140 150
NOVll gi) 8101125 I ^SSNGSOTJ^EA yi^iSS^FJS^lS^^T^sblTB^K^E gi]l674173θ) iMM am^ιιιsw«HWiiwjιt;iιxιifeτ«i5iMιιιw;wi3fB_ gi|592146l|
gi)l7028375) ιaτU^4.jκiia«H^^ι^ιtιara8π^τL*; 'iaι. «tri.^.^A*iRi^
160 170 180 190 200
NOVll KSGJEESR0QSTDQ^-SvS?^τS^CTiEs,WEIβFRSQARiS?TN gi I 8101125 I R — lffisBRH pτdBκSTOfi!5felϊifi{ro5E|3κvR[Sι_Bτ,κiaopvsgESH gi)l674173θ| gi|592146l) gi|l4751898| G- <eMi^*JsteGHl^.l«I^^^^^Rlei^iJid.^jamgy_l^^ gi|l7028375| — |>^^ftJEJAoi5TOs^<Ji<dGia!B3E^iyt;i»^yeiEB^!HiS3
210 220 230 240 250
NOVll ^SATTRJ^AE^Sϊj ^ JjRI^ΞGϊJiJ^S^ JHpp vSGE^GS N; gi] 8101125) F^SAS B^ -L itTfrpyRr^ GlggCTsϊSpBπ SODT-GFI-SA lCtf gi)l6741730| gi|592146l| gi 114751898 I
Figure imgf000127_0001
260 270 280 290 300
....|....|....|....|....)....|....|....|....|....|
NOVll SPKDK
Figure imgf000127_0002
Figure imgf000127_0003
gi I 5921461] RHvHpCAVA PIIV VI MIPI_fflcHγwHNlSlBlκla.MSM gi 114751898 I |33vEl PFfSficHYwtMBSk i gi ] 17028375 | tg3Α^P IWA AVI TASPWMVS TVIl^VFII M^S^CC|M|l S S! !515Bήl
310 320 330 340 350
NOVll
Figure imgf000127_0004
gi|8101125| H-G IIK!sf S [|SFS K^P
Figure imgf000127_0005
gi|592146l| [^SS^Fli^jRilELKELEKERV iς^EELQVKlKjEliSSβSSSΪ^V
Figure imgf000127_0006
gi 117028375 I fflSgBBJKvlSf alEidara bθtø»tø:>H.;Md dnlaHil- -
360 370 380 390 400
NOVll gi I 8101125] gi 116741730 I AE QFFSN gi I 59214611 D LDPDTAHPD F SEDRRSVRRCPFRH GESVPDNPERFDSQPCV GR gi ] 14751898 ] DW DPDTAHPD FLSEDRRSVRRCPFRHLGESVPDNPERFDSQPCVLGR gi 117028375 I ADVN TGLRNT
410 420 430 440 450
NOVll gi] 8101125 I
gi I 59214611 ESFASGKHYWEVEVENVIE TVGVCRDSVERKGEVL IPQNGF TLEMHK gi 114751898 | ESFASGKHY EVEVENVIEWTVGVCRDSVERKGEVL IPQNGF T EMHK
460 470 480 490 500
NOVll gi I 8101125 I
gi I 5921461 ] GQYRAVSSPDRILP KESLCRVGVF DYEAGDVSFYNMRDRSHIYTCPRS gi 114751898 | GQYRAVSSPDRILPLKES CRVGVFLDYEAGDVSFYNMRDRSHIYTCPRS
510 520 530 540 550
NOVll gi I 8101125 I gi | 5921461 ] AFSVP VRPFFR GCEDSPIFI-CPA TGANGVTVPEEGLT H gi ] 14751898 | AFSGPDTSQSGDPPEPIESIPWSHSHVDKPWSSQQPPHNTHLPAASFTPT
560
NOVll gi)8101125| gi|l6741730| gi I 59214611 RVGTHQSL gi I 14751898 I TDLSPSFLL TRLCF gi|l7028375|
Tables 1 IE lists the domain description from DOMAIN analysis results against NOVl 1. This indicates that the NOVl 1 sequence has properties similar to those of other proteins known to contain this domain.
Table HE. Domain Analysis of NOVll gnl|Smart|smart00406, IGv, Immunoglobulin V-Type
CD-Length = 80 residues, 96.2% aligned Score = 34.7 bits (78), Expect = 0.008
Query: 52 VELQCQLFPNISAEDMELRWYRCQPS AVHMHERGMDMDGEQKWQYRGRTTFMSDHVARG 111 V C+ + + W R P + + y+GR T D+ ++
Sbj ct : 2 VTLSCKASGF-TFSSYYVSWVRQPPGKG EW GYIGSDVSYSEASY GRVTISKDN-S N 59
Query: 112 KAMVRSHRVTTFDNRTYCC 130
+ + D TY C
Sb ct : 60 DVS TISN RVEDTGTYYC 78
The gene sequence of invention described herein encodes for a novel member of the B7-Immunoglobulin family of enzymes. Specifically, the sequence encodes a novel BUTYROPHILIN-like protein. BUTYROPHILIN molecules play crucial roles in T-cell activation making them plausible targets for cancer, ADDS, and inflammation therapies. The protein described here is known to be expressed in spleen, and liver which may indicate roles in lupus, endocrine disorders, inflammation, autoimmune disorders, and cancers including liver, bone, and leukemia.
Despite the fact that many tumors express MHC class I molecules presenting "foreign" peptide antigens, a vigorous rumor-destructing immune response is seldom detected. A possible explanation is that tumors cannot provide adequate costimulatory signals as provided by professional antigen presenting cells. CD28, upon interacting with B7, triggers costimulatory signals critical for the T-cell response. Transfection of tumor cells with B7 augments the immunogenicity of the tumor so that an anti-tumor immune response can be amplified. When B7-CD28 costimulation is provided CTL specific for otherwise silent epitopes can be activated. Therefore, unresponsiveness of T cells to many tumor antigens should be considered as ignorance rather than tolerance. Immunological ignorance may thus contribute to the failure of the immune system to respond against the tumor antigens.
There is considerable evidence to support an important role for co-stimulatory molecules in regulating the proliferation and activation of T cells in the immune response. Of particular relevance is the interaction between CD28 on T cells and B7 expressed on the surface of antigen presenting cells (APCs). CTLA-4, another molecule present on activated T cells may downregulate T cell activity, but its role remains uncertain. CTLA4-Ig, a fusion protein consisting of the extracellular domain of CTLA4 and the Fc portion of human immunoglobulin Gl (IgGl), has been useful for studying the role of CD28/B7 interactions in immune responses. A number of studies have shown that CTLA4-Ig can switch off T cell activation. In an ovalbumin sensitive murine model of asthma, CTLA4-Ig treatment suppressed the response to inhaled allergen (increased airway hyperresponsiveness [AHR], IgE production, recruitment of eosinophils into the lungs, production of IL-4, IL-5, and IL-10 and increased IFNgamma production from CD3-TCR-activated T cells). Anti B7-2 treatment has similar effects suggesting that interaction of B7-2 with CD28 is important in the development of a Th-2 type inflammatory response in mice. Recent observations have been of relevance to human allergic disease. In vitro studies have shown that CTLA4-Ig or anti-B7-2 antibody can inhibit allergen-induced proliferation and cytokine production by peripheral blood mononuclear cells from atopic subjects. The role of co-stimulation has been studied in a human bronchial explant model of asthma. CTLA4-Ig fusion protein effectively blocked allergen-induced production of IL-5 and IL-13 in bronchial explants from atopic asthmatics. These studies confirm the requirement for interaction between co-stimulatory molecules in cytokine production and allergic inflammation, and point to the CD28-B7 pathway as being important to the allergen-induced inflammation in asthma. Studies of organ transplantation in primates suggest that CTLA4-Ig is extremely effective in preventing organ rejection. While phase 1 clinical trials have shown CTLA-4-Ig treatment of patients with psoriasis vulgaris to be well tolerated and to result in clinical improvement, its role in asthma management merits further investigation.
The initiation and progression of autoimmune diseases, such as insulin-dependent diabetes mellitus (IDDM), are complex processes that depend on autoantigen exposure, genetic susceptibility, and secondary events that promote autoaggression. T-cell costimulation, largely mediated by CD28/B7 interactions, is a major regulatory pathway in the activation and differentiation of T-cells that cause IDDM in murine models. In this article, we summarize our results in two models of IDDM: the non obese diabetic (NOD) mouse and diabetes induced with multiple low doses of streptozotocin (MDSDM). In both of these models, blockade of CD28/B7 costimulation regulates the development of disease. The effects of blockade vary with the intensity of cognate signal delivered to the T-cells, the timing of the costimulatory signal, and perhaps even the CD28 ligand expressed on antigen-presenting cells (APCs). Our results suggest that targeting CD28/B7 signals is a feasible approach for treatment and prevention of recurrence of autoimmune diabetes. However, the dynamic nature of these interactions highlights the importance of a clear understanding of their role in regulation of the disease. PMID: 9048209, UI: 97200274
The disclosed NOVl 1 nucleic acid of the invention encoding a butyrophilin-like protein includes the nucleic acid whose sequence is provided in Table 11 A or a fragment thereof. The invention also includes a mutant or variant nucleic acid any of whose bases may be changed from the corresponding base shown in Table 11 A while still encoding a protein that maintains its butyrophilin-like activities and physiological functions, or a fragment of such a nucleic acid. The invention further includes nucleic acids whose sequences are complementary to those just described, including nucleic acid fragments that are complementary to any of the nucleic acids just described. The invention additionally includes nucleic acids or nucleic acid fragments, or complements thereto, whose structures include chemical modifications. Such modifications include, by way of nonlimiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject. In the mutant or variant nucleic acids, and their complements, up to about 1 percent of the bases may be so changed.
The disclosed NOVl 1 protein of the invention includes butyrophilin-like protein whose sequence is provided in Table 1 IB. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residue shown in Table 1 IB while still encoding a protein that maintains its butyrophilin-like activities and physiological functions, or a functional fragment thereof. In the mutant or variant protein, up to about 7 percent of the residues may be so changed. The invention further encompasses antibodies and antibody fragments, such as Fa or (Fab)2, that bind immunospecifically to any of the proteins of the invention.
The above defined information for this invention suggests that this butyrophilin-like protein (NOVl 1) may function as a member of a butyrophilin family. Therefore, the NOVl 1 nucleic acids and proteins identified here may be useful in potential therapeutic applications implicated in (but not limited to) various pathologies and disorders as indicated below. The potential therapeutic applications for this invention include, but are not limited to: protein therapeutic, small molecule drug target, antibody target (therapeutic, diagnostic, drug targeting/cytotoxic antibody), diagnostic and/or prognostic marker, gene therapy (gene delivery/gene ablation), research tools, tissue regeneration in vivo and in vitro of all tissues and cell types composing (but not limited to) those defined here.
The NOVl 1 nucleic acids and proteins of the invention are useful in potential therapeutic applications implicated in cancer including but not limited to various pathologies and disorders as indicated below. For example, a cDNA encoding butyrophilin-like protein (NOVl 1) may be useful in gene therapy, and the butyrophilin-like protein (NOVl 1) may be useful when administered to a subject in need thereof. By way of nonlimiting example, the compositions of the present invention will have efficacy for treatment of patients suffering from brain disorders including epilepsy, eating disorders, schizophrenia, ADD, and cancer; heart disease; inflammation and autoimmune disorders including Crohn's disease, IBD, lupus, allergies, rheumatoid and osteoarthritis, inflammatory skin disorders, blood disorders; psoriasis colon cancer, leukemia AIDS; thalamus disorders; metabolic disorders including diabetes and obesity; lung diseases such as asthma, emphysema, cystic fibrosis, and cancer; multiple sclerosis, pancreatic disorders including pancreatic insufficiency and cancer; and prostate disorders including prostate cancer as well as other diseases, disorders and conditions. The NOVl 1 nucleic acid encoding the butyrophilin-like protein of the invention, or fragments thereof, may further be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed.
NOVl 1 nucleic acids and polypeptides are further useful in the generation of antibodies that bind immuno-specifically to the novel NOVl 1 substances for use in therapeutic or diagnostic methods. These antibodies may be generated according to methods known in the art, using prediction from hydrophobicity charts, as described in the "Anti- NOVX Antibodies" section below. The disclosed NOVl 1 protein has multiple hydrophilic regions, each of which can be used as an immunogen. h one embodiment, a contemplated NOVll epitope is from about amino acids 25 to 50. In another embodiment, a NOVll epitope is from about amino acids 60 to 140. In additional embodiments, a NOVl 1 epitope is from about amino acids 150 to 200, and from about amino acids 240 to 280. These novel proteins can be used in assay systems for functional analysis of various human disorders, which will help in understanding of pathology of the disease and development of new drug targets for various disorders.
NOVX Nucleic Acids and Polypeptides
One aspect of the invention pertains to isolated nucleic acid molecules that encode NOVX polypeptides or biologically active portions thereof. Also included in the invention are nucleic acid fragments sufficient for use as hybridization probes to identify NOVX-encoding nucleic acids (e.g., NOVX mRNAs) and fragments for use as PCR primers for the amplification and/or mutation of NOVX nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA), RNA molecules (e.g., mRNA), analogs of the DNA or RNA generated using nucleotide analogs, and derivatives, fragments and homologs thereof. The nucleic acid molecule may be single-stranded or double-stranded, but preferably is comprised double- stranded DNA.
An NOVX nucleic acid can encode a mature NOVX polypeptide. As used herein, a "mature" form of a polypeptide or protein disclosed in the present invention is the product of a naturally occurring polypeptide or precursor form or proprotein. The naturally occurring polypeptide, precursor or proprotein includes, by way of nonlimiting example, the full-length gene product, encoded by the corresponding gene. Alternatively, it may be defined as the polypeptide, precursor or proprotein encoded by an ORF described herein. The product "mature" form arises, again by way of nonlimiting example, as a result of one or more naturally occurring processing steps as they may take place within the cell, or host cell, in which the gene product arises. Examples of such processing steps leading to a "mature" form of a polypeptide or protein include the cleavage of the N-terminal methionine residue encoded by the initiation codon of an ORF, or the proteolytic cleavage of a signal peptide or leader sequence. Thus a mature form arising from a precursor polypeptide or protem that has residues 1 to N, where residue 1 is the N-terminal methionine, would have residues 2 through N remaining after removal of the N-terminal methionine. Alternatively, a mature form arising from a precursor polypeptide or protein having residues 1 to N, in which an N-terminal signal sequence from residue 1 to residue M is cleaved, would have the residues from residue M+1 to residue N remaining. Further as used herein, a "mature" form of a polypeptide or protein may arise from a step of post-translational modification other than a proteolytic cleavage event. Such additional processes include, by way of non-limiting example, glycosylation, myristoylation or phosphorylation. In general, a mature polypeptide or protein may result from the operation of only one of these processes, or a combination of any of them.
The term "probes", as utilized herein, refers to nucleic acid sequences of variable length, preferably between at least about 10 nucleotides (nt), 100 nt, or as many as approximately, e.g., 6,000 nt, depending upon the specific use. Probes are used in the detection of identical, similar, or complementary nucleic acid sequences. Longer length probes are generally obtained from a natural or recombinant source, are highly specific, and much slower to hybridize than shorter-length oligomer probes. Probes may be single- or double-stranded and designed to have specificity in PCR, membrane-based hybridization technologies, or ELISA-like technologies.
The term "isolated" nucleic acid molecule, as utilized herein, is one, which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5'- and 3 '-termini of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, in various embodiments, the isolated NOVX nucleic acid molecules can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell/tissue from which the nucleic acid is derived (e.g., brain, heart, liver, spleen, etc.). Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material or culture medium when produced by recombinant techniques, or of chemical precursors or other chemicals when chemically synthesized.
A nucleic acid molecule of the invention, e.g., a nucleic acid molecule having the nucleotide sequence SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a complement of this aforementioned nucleotide sequence, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or a portion ofthe nucleic acid sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, as a hybridization probe, NOVX molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, et αl., (eds.), MOLECULAR CLONING: A LABORATORY MANUAL 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; and Ausubel, et αl., (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993.) A nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to NOVX nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
As used herein, the term "oligonucleotide" refers to a series of linked nucleotide residues, which oligonucleotide has a sufficient number of nucleotide bases to be used in a PCR reaction. A short oligonucleotide sequence may be based on, or designed from, a genomic or cDNA sequence and is used to amplify, confirm, or reveal the presence of an identical, similar or complementary DNA or RNA in a particular cell or tissue. Oligonucleotides comprise portions of a nucleic acid sequence having about 10 nt, 50 nt, or 100 nt in length, preferably about 15 nt to 30 nt in length. In one embodiment of the invention, an oligonucleotide comprising a nucleic acid molecule less than 100 nt in length would further comprise at least 6 contiguous nucleotides SEQ ID NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a complement thereof. Oligonucleotides may be chemically synthesized and may also be used as probes.
In another embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule that is a complement of the nucleotide sequence shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a portion of this nucleotide sequence (e.g., a fragment that can be used as a probe or primer or a fragment encoding a biologically-active portion of an NOVX polypeptide). A nucleic acid molecule that is complementary to the nucleotide sequence shown SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 3 and 33 is one that is sufficiently complementary to the nucleotide sequence shown SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 that it can hydrogen bond with little or no mismatches to the nucleotide sequence shown SEQ ID NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, thereby forming a stable duplex.
As used herein, the term "complementary" refers to Watson-Crick or Hoogsteen base pairing between nucleotides units of a nucleic acid molecule, and the term "binding" means the physical or chemical interaction between two polypeptides or compounds or associated polypeptides or compounds or combinations thereof. Binding includes ionic, non-ionic, van der Waals, hydrophobic interactions, and the like. A physical interaction can be either direct or indirect. Indirect interactions may be through or due to the effects of another polypeptide or compound. Direct binding refers to interactions that do not take place through, or due to, the effect of another polypeptide or compound, but instead are without other substantial chemical intermediates.
Fragments provided herein are defined as sequences of at least 6 (contiguous) nucleic acids or at least 4 (contiguous) amino acids, a length sufficient to allow for specific hybridization in the case of nucleic acids or for specific recognition of an epitope in the case of amino acids, respectively, and are at most some portion less than a full length sequence. Fragments may be derived from any contiguous portion of a nucleic acid or amino acid sequence of choice. Derivatives are nucleic acid sequences or amino acid sequences formed from the native compounds either directly or by modification or partial substitution. Analogs are nucleic acid sequences or amino acid sequences that have a structure similar to, but not identical to, the native compound but differs from it in respect to certain components or side chains. Analogs may be synthetic or from a different evolutionary origin and may have a similar or opposite metabolic activity compared to wild type. Homologs are nucleic acid sequences or amino acid sequences of a particular gene that are derived from different species.
Derivatives and analogs may be full length or other than full length, if the derivative or analog contains a modified nucleic acid or amino acid, as described below. Derivatives or analogs of the nucleic acids or proteins of the invention include, but are not limited to, molecules comprising regions that are substantially homologous to the nucleic acids or proteins of the invention, in various embodiments, by at least about 70%, 80%, or 95% identity (with a preferred identity of 80-95%) over a nucleic acid or amino acid sequence of identical size or when compared to an aligned sequence in which the alignment is done by a computer homology program known in the art, or whose encoding nucleic acid is capable of hybridizing to the complement of a sequence encoding the aforementioned proteins under stringent, moderately stringent, or low stringent conditions. See e.g. Ausubel, et al., CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, New York, NY, 1993, and below.
A "homologous nucleic acid sequence" or "homologous amino acid sequence," or variations thereof, refer to sequences characterized by a homology at the nucleotide level or amino acid level as discussed above. Homologous nucleotide sequences encode those sequences coding for isoforms of NOVX polypeptides. Isoforms can be expressed in different tissues of the same organism as a result of, for example, alternative splicing of RNA. Alternatively, isoforms can be encoded by different genes. In the invention, homologous nucleotide sequences include nucleotide sequences encoding for an NOVX polypeptide of species other than humans, including, but not limited to: vertebrates, and thus can include, e.g., frog, mouse, rat, rabbit, dog, cat cow, horse, and other organisms. Homologous nucleotide sequences also include, but are not limited to, naturally occurring allelic variations and mutations of the nucleotide sequences set forth herein. A homologous nucleotide sequence does not, however, include the exact nucleotide sequence encoding human NOVX protein. Homologous nucleic acid sequences include those nucleic acid sequences that encode conservative amino acid substitutions (see below) in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, as well as a polypeptide possessing NOVX biological activity. Various biological activities of the NOVX proteins are described below.
An NOVX polypeptide is encoded by the open reading frame ("ORF") of an NOVX nucleic acid. An ORF corresponds to a nucleotide sequence that could potentially be translated into a polypeptide. A stretch of nucleic acids comprising an ORF is uninterrupted by a stop codon. An ORF that represents the coding sequence for a full protein begins with an ATG "start" codon and terminates with one of the three "stop" codons, namely, TAA, TAG, or TGA. For the purposes of this invention, an ORF may be any part of a coding sequence, with or without a start codon, a stop codon, or both. For an ORF to be considered as a good candidate for coding for a bonafide cellular protein, a minimum size requirement is often set, e.g., a stretch of DNA that would encode a protein of 50 amino acids or more.
The nucleotide sequences determined from the cloning of the human NOVX genes allows for the generation of probes and primers designed for use in identifying and/or cloning NOVX homologues in other cell types, e.g. from other tissues, as well as NOVX homologues from other vertebrates. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, 25, 50, 100, 150, 200, 250, 300, 350 or 400 consecutive sense strand nucleotide sequence SEQ ID NOS:l, 3, 5, 1, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33; or an anti-sense strand nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33; or of a naturally occurring mutant of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
Probes based on the human NOVX nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In various embodiments, the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissues which mis- express an NOVX protein, such as by measuring a level of an NOVX-encoding nucleic acid in a sample of cells from a subject e.g., detecting NOVX mRNA levels or determining whether a genomic NOVX gene has been mutated or deleted.
"A polypeptide having a biologically-active portion of an NOVX polypeptide" refers to polypeptides exhibiting activity similar, but not necessarily identical to, an activity of a polypeptide of the invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. A nucleic acid fragment encoding a "biologically- active portion of NOVX" can be prepared by isolating a portion SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, that encodes a polypeptide having an NOVX biological activity (the biological activities of the NOVX proteins are described below), expressing the encoded portion of NOVX protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of NOVX.
NOVX Nucleic Acid and Polypeptide Variants
The invention further encompasses nucleic acid molecules that differ from the nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 due to degeneracy of the genetic code and thus encode the same NOVX proteins as that encoded by the nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 33.
In addition to the human NOVX nucleotide sequences shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 33, it will be appreciated by those skilled in the art that DNA sequence polymorphisms that lead to changes in the amino acid sequences of the NOVX polypeptides may exist within a population (e.g., the human population). Such genetic polymorphism in the NOVX genes may exist among individuals within a population due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules comprising an open reading frame (ORF) encoding an NOVX protein, preferably a vertebrate NOVX protein. Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the NOVX genes. Any and all such nucleotide variations and resulting amino acid polymoφhisms in the NOVX polypeptides, which are the result of natural allelic variation and that do not alter the functional activity of the NOVX polypeptides, are intended to be within the scope of the invention. Moreover, nucleic acid molecules encoding NOVX proteins from other species, and thus that have a nucleotide sequence that differs from the human SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 are intended to be within the scope of the invention. Nucleic acid molecules corresponding to natural allelic variants and homologues of the NOVX cDNAs of the invention can be isolated based on their homology to the human NOVX nucleic acids disclosed herein using the human cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 6 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence ofSEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33. In another embodiment, the nucleic acid is at least 10, 25, 50, 100, 250, 500, 750, 1000, 1500, or 2000 or more nucleotides in length. In yet another embodiment, an isolated nucleic acid molecule of the invention hybridizes to the coding region. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.
Homologs (i.e., nucleic acids encoding NOVX proteins derived from species other than human) or other related sequences (e.g., paralogs) can be obtained by low, moderate or high stringency hybridization with all or a portion of the particular human sequence as a probe using methods well known in the art for nucleic acid hybridization and cloning.
As used herein, the phrase "stringent hybridization conditions" refers to conditions under which a probe, primer or oligonucleotide will hybridize to its target sequence, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures than shorter sequences. Generally, stringent conditions are selected to be about 5 °C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength and pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target sequence hybridize to the target sequence at equilibrium. Since the target sequences are generally present at excess, at Tm, 50% of the probes are occupied at equilibrium. Typically, stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30°C for short probes, primers or oligonucleotides (e.g., 10 nt to 50 nt) and at least about 60°C for longer probes, primers and oligonucleotides. Stringent conditions may also be achieved with the addition of destabilizing agents, such as formamide.
Stringent conditions are known to those skilled in the art and can be found in Ausubel, et al, (eds.), CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. Preferably, the conditions are such that sequences at least about 65%, 70%, 75%, 85%, 90%, 95%, 98%, or 99% homologous to each other typically remain hybridized to each other. A non-limiting example of stringent hybridization conditions are hybridization in a high salt buffer comprising 6X SSC, 50 mM Tris-HCl (pH 7.5), 1 mM EDTA, 0.02% PVP, 0.02% FicoU, 0.02% BSA, and 500 mg/ml denatured salmon sperm DNA at 65°C, followed by one or more washes in 0.2X SSC, 0.01% BSA at 50°C. An isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
In a second embodiment, a nucleic acid sequence that is hybridizable to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof, under conditions of moderate stringency is provided. A non-limiting example of moderate stringency hybridization conditions are hybridization in 6X SSC, 5X Denhardt's solution, 0.5% SDS and 100 mg/ml denatured salmon sperm DNA at 55°C, followed by one or more washes in IX SSC, 0.1% SDS at 37°C. Other conditions of moderate stringency that may be used are well-known within the art. See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990; GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY.
In a third embodiment, a nucleic acid that is hybridizable to the nucleic acid molecule comprising the nucleotide sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof, under conditions of low stringency, is provided. A non-limiting example of low stringency hybridization conditions are hybridization in 35% formamide, 5X SSC, 50 mM Tris-HCl (pH 7.5), 5 mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 mg/ml denatured salmon sperm DNA, 10% (wt/vol) dextran sulfate at 40°C, followed by one or more washes in 2X SSC, 25 mM Tris-HCl (pH 7.4), 5 mM EDTA, and 0.1% SDS at 50°C. Other conditions of low stringency that may be used are well known in the art (e.g., as employed for cross-species hybridizations). See, e.g., Ausubel, et al. (eds.), 1993, CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, NY, and Kriegler, 1990, GENE TRANSFER AND EXPRESSION, A LABORATORY MANUAL, Stockton Press, NY; Shilo and Weinberg, 1981. Proc Natl Acad Sci USA 78 : 6789-6792.
Conservative Mutations
In addition to naturally-occurring allelic variants of NOVX sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, thereby leading to changes in the amino acid sequences of the encoded NOVX proteins, without altering the functional ability of said NOVX proteins. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence SEQ ID NOS:2, 4, , 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, and 34. A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequences of the NOVX proteins without altering their biological activity, whereas an "essential" amino acid residue is required for such biological activity. For example, amino acid residues that are conserved among the NOVX proteins of the invention are predicted to be particularly non-amenable to alteration. Amino acids for which conservative substitutions can be made are well-known within the art.
Another aspect of the invention pertains to nucleic acid molecules encoding NOVX proteins that contain changes in amino acid residues that are not essential for activity. Such NOVX proteins differ in amino acid sequence from SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 45% homologous to the amino acid sequences SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. Preferably, the protein encoded by the nucleic acid molecule is at least about 60% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; more preferably at least about 70% homologous SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; still more preferably at least about 80% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; even more preferably at least about 90% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; and most preferably at least about 95% homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. An isolated nucleic acid molecule encoding an NOVX protein homologous to the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOSrl, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
Mutations can be introduced into SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted, non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined within the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted non-essential amino acid residue in the NOVX protein is replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of an NOVX coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for NOVX biological activity to identify mutants that retain activity. Following mutagenesis SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, the encoded protein can be expressed by any recombinant technology known in the art and the activity of the protein can be determined.
The relatedness of amino acid families may also be determined based on side chain interactions. Substituted amino acids may be fully conserved "strong" residues or fully conserved "weak" residues. The "strong" group of conserved amino acid residues may be any one of the following groups: STA, NEQK, NHQK, NDEQ, QHRK, MILV, MILF, HY, FYW, wherein the single letter amino acid codes are grouped by those amino acids that may be substituted for each other. Likewise, the "weak" group of conserved residues may be any one of the following: CSA, ATV, SAG, STNK, STPA, SGND, SNDEQK, NDEQHK, NEQHRK, VLIM, HFY, wherein the letters within each group represent the single letter amino acid code. In one embodiment, a mutant NOVX protein can be assayed for (i) the ability to form proteimprotein interactions with other NOVX proteins, other cell-surface proteins, or biologically-active portions thereof, (ii) complex formation between a mutant NOVX protein and an NOVX ligand; or (iii) the ability of a mutant NOVX protein to bind to an intracellular target protein or biologically-active portion thereof; (e.g. avidin proteins). h yet another embodiment, a mutant NOVX protein can be assayed for the ability to regulate a specific biological function (e.g., regulation of insulin release).
Antisense Nucleic Acids
Another aspect of the invention pertains to isolated antisense nucleic acid molecules that are hybridizable to or complementary to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments, analogs or derivatives thereof. An "antisense" nucleic acid comprises a nucleotide sequence that is complementary to a "sense" nucleic acid encoding a protein (e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence), specific aspects, antisense nucleic acid molecules are provided that comprise a sequence complementary to at least about 10, 25, 50, 100, 250 or 500 nucleotides or an entire NOVX coding strand, or to only a portion thereof. Nucleic acid molecules encoding fragments, homologs, derivatives and analogs of an NOVX protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, or antisense nucleic acids complementary to an NOVX nucleic acid sequence of SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, are additionally provided.
In one embodiment, an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding an NOVX protein. The term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues. In another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding the NOVX protein. The term "noncoding region" refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions).
Given the coding strand sequences encoding the NOVX protein disclosed herein, antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick or Hoogsteen base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of NOVX mRNA, but more preferably is an oligonucleotide that is antisense to only a portion of the coding or noncoding region of NOVX mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of NOVX mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis or enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally-occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids (e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used).
Examples of modified nucleotides that can be used to generate the antisense nucleic acid include: 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl- 2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2-methylguanine, 3-methylcytosine, 5-methylcytosine, Nό-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5'-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3-amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminoρurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an NOVX protein to thereby inhibit expression of the protein (e.g., by inhibiting transcription and/or translation). The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule that binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention includes direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface (e.g., by linking the antisense nucleic acid molecules to peptides or antibodies that bind to cell surface receptors or antigens). The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient nucleic acid molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other. See, e.g., Gaultier, et al, 1987. Nucl. Acids Res. 15: 6625-6641. The antisense nucleic acid molecule can also comprise a 2'-o-methylribonucleotide (See, e.g., fr oue, et al. 1987. Nucl Acids Res. 15: 6131-6148) or a chimeric RNA-DNA analogue (See, e.g., Inoue, et al, 1987. FEBSLett. 215: 327-330.
Ribozymes and PNA Moieties
Nucleic acid modifications include, by way of non-limiting example, modified bases, and nucleic acids whose sugar phosphate backbones are modified or derivatized. These modifications are carried out at least in part to enhance the chemical stability of the modified nucleic acid, such that they may be used, for example, as antisense binding nucleic acids in therapeutic applications in a subject.
In one embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity that are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes as described in Haselhoff and Gerlach 1988. Nature 334: 585-591) can be used to catalytically cleave NOVX mRNA transcripts to thereby inhibit translation of NOVX mRNA. A ribozyme having specificity for an NOVX-encoding nucleic acid can be designed based upon the nucleotide sequence of an NOVX cDNA disclosed herein (i.e., SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an NOVX-encoding mRNA. See, e.g., U.S. Patent 4,987,071 to Cech, et al. and U.S. Patent 5,116,742 to Cech, et al. NOVX mRNA can also be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel et al, (1993) Science 261:1411-1418.
Alternatively, NOVX gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the NOVX nucleic acid (e.g., the NOVX promoter and/or enhancers) to form triple helical structures that prevent transcription of the NOVX gene in target cells. See, e.g., Helene, 1991. Anticancer Drug Des. 6: 569-84; Helene, et al. 1992. Ann. NY. Acad. Sci. 660: 27-36; Maher, 1992. Bioassays 14: 807-15.
In various embodiments, the NOVX nucleic acids can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acids can be modified to generate peptide nucleic acids. See, e.g., Hyrup, et al, 1996. BioorgMed Chem 4: 5-23. As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics (e.g., DNA mimics) in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup, et al, 1996. supra; Perry-O'Keefe, et al, 1996. Proc. Natl. Acad. Sci. USA 93: 14670-14675.
PNAs of NOVX can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, e.g., inducing transcription or translation arrest or inhibiting replication. PNAs of NOVX can also be used, for example, in the analysis of single base pair mutations in a gene (e.g., PNA directed PCR clamping; as artificial restriction enzymes when used in combination with other enzymes, e.g., Si nucleases (See, Hyrup, et al, 1996.supra); or as probes or primers for DNA sequence and hybridization (See, Hyrup, et al, 1996, supra; Perry-O'Keefe, et al, 1996. supra).
In another embodiment, PNAs of NOVX can be modified, e.g., to enhance their stability or cellular uptake, by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of NOVX can be generated that may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes (e.g., RNase H and DNA polymerases) to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (see, Hyrup, et al., 1996. supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup, et al, 1996. supra and Finn, et ah, 1996. Nucl Acids Res 24: 3357-3363. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry, and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used between the PNA and the 5' end of DNA. See, e.g., Mag, et al, 1989. Nucl Acid Res 17: 5973-5988. PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment. See, e.g., Finn, et al, 1996. supra. Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment. See, e.g., Petersen, et al, 1975. Bioorg. Med. Chem. Lett. 5: 1119-11124.
In other embodiments, the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger, et al, 1989. Proc. Natl. Acad. Sci. U.S.A. 86: 6553-6556; Lemaitre, et al, 1987. Proc. Natl. Acad. Sci. 84: 648-652; PCT Publication No. , WO88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134). hi addition, oligonucleotides can be modified with hybridization triggered cleavage agents (see, e.g., Krol, et al, 1988. BioTechniques 6:958-976) or intercalating agents (see, e.g., Zon, 1988. Pharm. Res. 5: 539-549). To this end, the oligonucleotide may be conjugated to another molecule, e.g., a peptide, a hybridization triggered cross-linking agent, a transport agent, a hybridization-triggered cleavage agent, and the like.
NOVX Polypeptides
A polypeptide according to the invention includes a polypeptide including the amino acid sequence of NOVX polypeptides whose sequences are provided in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. The invention also includes a mutant or variant protein any of whose residues may be changed from the corresponding residues shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34 while still encoding a protein that maintains its NOVX activities and physiological functions, or a functional fragment thereof.
In general, an NOVX variant that preserves NOVX-like function includes any variant in which residues at a particular position in the sequence have been substituted by other amino acids, and further include the possibility of inserting an additional residue or residues between two residues of the parent protein as well as the possibility of deleting one or more residues from the parent sequence. Any amino acid substitution, insertion, or deletion is encompassed by the invention. In favorable circumstances, the substitution is a conservative substitution as defined above.
One aspect of the invention pertains to isolated NOVX proteins, and biologically- active portions thereof, or derivatives, fragments, analogs or homologs thereof. Also provided are polypeptide fragments suitable for use as immunogens to raise anti-NOVX antibodies, hi one embodiment, native NOVX proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques, hi another embodiment, NOVX proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, an NOVX protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
An "isolated" or "purified" polypeptide or protein or biologically-active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the NOVX protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of NOVX proteins in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly-produced. In one embodiment, the language "substantially free of cellular material" includes preparations of NOVX proteins having less than about 30% (by dry weight) of non-NOVX proteins (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non-NOVX proteins, still more preferably less than about 10% of non-NOVX proteins, and most preferably less than about 5% of non-NOVX proteins. When the NOVX protein or biologically-active portion thereof is recombinantly-produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the NOVX protein preparation.
The language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins in which the protein is separated from chemical precursors or other chemicals that are involved in the synthesis of the protein, hi one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of NOVX proteins having less than about 30% (by dry weight) of chemical precursors or non-NOVX chemicals, more preferably less than about 20% chemical precursors or non-NOVX chemicals, still more preferably less than about 10% chemical precursors or non-NOVX chemicals, and most preferably less than about 5% chemical precursors or non-NOVX chemicals.
Biologically-active portions of NOVX proteins include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequences of the NOVX proteins (e.g., the amino acid sequence shown in SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34) that include fewer amino acids than the full-length NOVX proteins, and exhibit at least one activity of an NOVX protein. Typically, biologically- active portions comprise a domain or motif with at least one activity of the NOVX protein. A biologically-active portion of an NOVX protein can be a polypeptide which is, for example, 10, 25, 50, 100 or more amino acid residues in length.
Moreover, other biologically-active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native NOVX protein.
In an embodiment, the NOVX protein has an amino acid sequence shown SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34. In other embodiments, the NOVX protein is substantially homologous to SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, and retains the functional activity of the protein of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail, below. Accordingly, in another embodiment, the NOVX protein is a protein that comprises an amino acid sequence at least about 45% homologous to the amino acid sequence SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, and retains the functional activity of the NOVX proteins of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34.
Determining Homology Between Two or More Sequences
To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity").
The nucleic acid sequence homology may be determined as the degree of identity between two sequences. The homology may be determined using computer programs known in the art, such as GAP software provided in the GCG program package. See, Needleman and Wunsch, 1970. J Mol Biol 48: 443-453. Using GCG GAP software with the following settings for nucleic acid sequence comparison: GAP creation penalty of 5.0 and GAP extension penalty of 0.3, the coding region of the analogous nucleic acid sequences referred to above exhibits a degree of identity preferably of at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99%, with the CDS (encoding) part of the DNA sequence shown in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33.
The term "sequence identity" refers to the degree to which two polynucleotide or polypeptide sequences are identical on a residue-by-residue basis over a particular region of comparison. The term "percentage of sequence identity" is calculated by comparing two optimally aligned sequences over that region of comparison, determining the number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I, in the case of nucleic acids) occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the region of comparison (i.e., the window size), and multiplying the result by 100 to yield the percentage of sequence identity. The term "substantial identity" as used herein denotes a characteristic of a polynucleotide sequence, wherein the polynucleotide comprises a sequence that has at least 80 perpent sequence identity, preferably at least 85 percent identity and often 90 to 95 percent sequence identity, more usually at least 99 percent sequence identity as compared to a reference sequence over a comparison region.
Chimeric and Fusion Proteins
The invention also provides NOVX chimeric or fusion proteins. As used herein, an NOVX "chimeric protein" or "fusion protein" comprises an NOVX polypeptide operatively- linked to a non-NOVX polypeptide. An "NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to an NOVX protein SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, whereas a "non-NOVX polypeptide" refers to a polypeptide having an amino acid sequence corresponding to a protein that is not substantially homologous to the NOVX protein, e.g., a protein that is different from the NOVX protein and that is derived from the same or a different organism. Within an NOVX fusion protein the NOVX polypeptide can correspond to all or a portion of an NOVX protein. In one embodiment, an NOVX fusion protein comprises at least one biologically-active portion of an NOVX protein. In another embodiment, an NOVX fusion protein comprises at least two biologically-active portions of an NOVX protein, hi yet another embodiment, an NOVX fusion protein comprises at least three biologically-active portions of an NOVX protein. Within the fusion protein, the term "operatively-linked" is intended to indicate that the NOVX polypeptide and the non-NOVX polypeptide are fused in-frame with one another. The non-NOVX polypeptide can be fused to the N-terminus or C-terminus of the NOVX polypeptide.
In one embodiment, the fusion protein is a GST-NO VX fusion protein in which the NOVX sequences are fused to the C-terminus of the GST (glutathione S-transferase) sequences. Such fusion proteins can facilitate the purification of recombinant NOVX polypeptides.
In another embodiment, the fusion protein is an NOVX protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of NOVX can be increased through use of a heterologous signal sequence.
In yet another embodiment, the fusion protein is an NOVX-immunoglobulin fusion protein in which the NOVX sequences are fused to sequences derived from a member of the immunoglobulin protein family. The NOVX-immunoglobulin fusion proteins of the invention can be incoφorated into pharmaceutical compositions and administered to a subject to inhibit an interaction between an NOVX ligand and an NOVX protein on the surface of a cell, to thereby suppress NOVX-mediated signal transduction in vivo. The NOVX-immunoglobulin fusion proteins can be used to affect the bioavailability of an NOVX cognate ligand. Inhibition of the NOVX ligand/NOVX interaction may be useful therapeutically for both the treatment of proliferative and differentiative disorders, as well as modulating (e.g. promoting or inhibiting) cell survival. Moreover, the NOVX-immunoglobulin fusion proteins of the invention can be used as immunogens to produce anti-NOVX antibodies in a subject, to purify NOVX ligands, and in screening assays to identify molecules that inhibit the interaction of NOVX with an NOVX ligand.
An NOVX chimeric or fusion protein of the invention can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, e.g., by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be caπied out using anchor primers that give rise to complementary overhangs between two consecutive gene fragments that can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Ausubel, et al (eds.) CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, John Wiley & Sons, 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). An NOVX-encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the NOVX protein.
NOVX Agonists and Antagonists
The invention also pertains to variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists. Variants of the NOVX protein can be generated by mutagenesis (e.g., discrete point mutation or truncation of the NOVX protein). An agonist of the NOVX protein can retain substantially the same, or a subset of, the biological activities of the naturally occurring form of the NOVX protein. An antagonist of the NOVX protein can inhibit one or more of the activities of the naturally occurring form of the NOVX protein by, for example, competitively binding to a downstream or upstream member of a cellular signaling cascade which includes the NOVX protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the NOVX proteins.
Variants of the NOVX proteins that function as either NOVX agonists (i.e., mimetics) or as NOVX antagonists can be identified by screening combinatorial libraries of mutants (e.g., truncation mutants) of the NOVX proteins for NOVX protein agonist or antagonist activity. In one embodiment, a variegated library of NOVX variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of NOVX variants carl be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential NOVX sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of NOVX sequences therein. There are a variety of methods which can be used to produce libraries of potential NOVX variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be perfoπned in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired, set of potential NOVX sequences. Methods for synthesizing degenerate oligonucleotides are well-known within the art. See, e.g., Narang, 1983. Tetrahedron 39: 3; Itakura, et al, 1984. Annu. Rev. Biochem. 53: 323; Itakura, et al, 1984. Science 198: 1056; Ike, et al, 1983. Nucl. Acids Res. 11 : 477.
Polypeptide Libraries
In addition, libraries of fragments of the NONX protein coding sequences can be used to generate a variegated population of ΝOVX fragments for screening and subsequent selection of variants of an ΝOVX protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an ΝOVX coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DΝA, renaturing the DΝA to form double-stranded DΝA that can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with Si nuclease, and ligating the resulting fragment library into an expression vector. By this method, expression libraries can be derived which encodes Ν-terminal and internal fragments of various sizes of the ΝOVX proteins.
Various techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDΝA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of ΝOVX proteins. The most widely used techniques, which are amenable to high throughput analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recursive ensemble mutagenesis (REM), a new technique that enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify ΝOVX variants. See, e.g., Arkin and Yourvan, 1992. Proc. Natl. Acad. Sci. USA 89: 7811-7815; Delgrave, et al, 1993. Protein Engineering 6:327-331.
Anti-NOVX Antibodies
Also included in the invention are antibodies to NOVX proteins, or fragments of NOVX proteins. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen. Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab, Fab' and F(ab')2 fragments, and an Fab expression library. In general, an antibody molecule obtained from humans relates to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule. Certain classes have subclasses as well, such as IgGi, IgG2, and others. Furthermore, in humans, the light chain may be a kappa chain or a lambda chain. Reference herein to antibodies includes a reference to all such classes, subclasses and types of human antibody species.
An isolated NOVX-related protein of the invention may be intended to serve as an antigen, or a portion or fragment thereof, and additionally can be used as an immunogen to generate antibodies that immunospecifically bind the antigen, using standard techniques for polyclonal and monoclonal antibody preparation. The full-length protein can be used or, alternatively, the invention provides antigenic peptide fragments of the antigen for use as immunogens. An antigenic peptide fragment comprises at least 6 amino acid residues of the amino acid sequence of the full length protein and encompasses an epitope thereof such that an antibody raised against the peptide forms a specific immune complex with the full length protein or with any fragment that contains the epitope. Preferably, the antigenic peptide comprises at least 10 amino acid residues, or at least 15 amino acid residues, or at least 20 amino acid residues, or at least 30 amino acid residues. Preferred epitopes encompassed by the antigenic peptide are regions of the protein that are located on its surface; commonly these are hydrophilic regions.
In certain embodiments of the invention, at least one epitope encompassed by the antigenic peptide is a region of NOVX-related protein that is located on the surface of the protein, e.g., a hydrophilic region. A hydrophobicity analysis of the human NOVX-related protein sequence will indicate which regions of a NOVX-related protein are particularly hydrophilic and, therefore, are likely to encode surface residues useful for targeting antibody production. As a means for targeting antibody production, hydropathy plots showing regions of hydrophilicity and hydrophobicity may be generated by any method well known in the art, including, for example, the Kyte Doolittle or the Hopp Woods methods, either with or without Fourier transformation. See, e.g., Hopp and Woods, 1981, Proc. Nat. Acad. Sci. USA 78: 3824-3828; Kyte and Doolittle 1982, J. Mol. Biol. 157: 105-142, each of which is incoφorated herein by reference in its entirety. Antibodies that are specific for one or more domains within an antigenic protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.
A protein of the invention, or a derivative, fragment, analog, homolog or ortholog thereof, may be utilized as an immunogen in the generation of antibodies that immunospecifically bind these protein components.
Various procedures known within the art may be used for the production of polyclonal or monoclonal antibodies directed against a protein of the invention, or against derivatives, fragments, analogs homologs or orthologs thereof (see, for example, Antibodies: A Laboratory Manual, Harlow and Lane, 1988, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, incoφorated herein by reference). Some of these antibodies are discussed below.
Polyclonal Antibodies
For the production of polyclonal antibodies, various suitable host animals (e.g., rabbit, goat, mouse or other mammal) may be immunized by one or more injections with the native protein, a synthetic variant thereof, or a derivative of the foregoing. An appropriate immunogenic preparation can contain, for example, the naturally occurring immunogenic protein, a chemically synthesized polypeptide representing the immunogenic protein, or a recombinantly expressed immunogenic protein. Furthermore, the protein may be conjugated to a second protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include but are not limited to keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. The preparation can further include an adjuvant. Various adjuvants used to increase the immunological response include, but are not limited to, Freund's (complete and incomplete), mineral gels (e.g., aluminum hydroxide), surface active substances (e.g., lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, dinitrophenol, etc.), adjuvants usable in humans such as Bacille Calmette-Guerin and Corynebacterium parvum, or similar immunostimulatory agents. Additional examples of adjuvants which can be employed include MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
The polyclonal antibody molecules directed against the immunogenic protein can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as affinity chromatography using protein A or protein G, which provide primarily the IgG fraction of immune serum. Subsequently, or alternatively, the specific antigen which is the target of the immunoglobulin sought, or an epitope thereof, may be immobilized on a column to purify the immune specific antibody by immunoaffmity chromatography. Purification of immunoglobulins is discussed, for example, by D. Wilkinson (The Scientist, published by The Scientist, Inc., Philadelphia PA, Vol. 14, No. 8 (April 17, 2000), pp. 25-28).
Monoclonal Antibodies
The term "monoclonal antibody" (MAb) or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one molecular species of antibody molecule consisting of a unique light chain gene product and a unique heavy chain gene product. In particular, the complementarity determining regions (CDRs) of the monoclonal antibody are identical in all the molecules of the population. MAbs thus contain an antigen binding site capable of immunoreacting with a particular epitope of the antigen characterized by a unique binding affinity for it.
Monoclonal antibodies can be prepared using hybridoma methods, such as those described by Kohler and Milstein, Nature, 256:495 (1975). In a hybridoma method, a mouse, hamster, or other appropriate host animal, is typically immunized with an immunizing agent to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the immunizing agent. Alternatively, the lymphocytes can be immunized in vitro.
The immunizing agent will typically include the protein antigen, a fragment thereof or a fusion protein thereof. Generally, either peripheral blood lymphocytes are used if cells of human origin are desired, or spleen cells or lymph node cells are used if non-human mammalian sources are desired. The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, MONOCLONAL ANTIBODIES: PRINCIPLES AND PRACTICE, Academic Press, (1986) pp. 59-103). Immortalized cell lines are usually transformed mammalian cells, particularly myeloma cells of rodent, bovine and human origin. Usually, rat or mouse myeloma cell lines are employed. The hybridoma cells can be cultured in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances prevent the growth of HGPRT-deficient cells.
Prefeπed immortalized cell lines are those that fuse efficiently, support stable high level expression of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. More preferred immortalized cell lines are murine myeloma lines, which can be obtained, for instance, from the Salk Institute Cell Distribution Center, San Diego, California and the American Type Culture Collection, Manassas, Virginia. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol, 133:3001 (1984); Brodeur et al., MONOCLONAL ANTIBODY PRODUCTION TECHNIQUES AND APPLICATIONS, Marcel Dekker, Inc., New York, (1987) pp. 51-63).
The culture medium in which the hybridoma cells are cultured can then be assayed for the presence of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by the hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA). Such techniques and assays are known in the art. The binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson and Pollard, Anal. Biochem., 107:220 (1980). Preferably, antibodies having a high degree of specificity and a high binding affinity for the target antigen are isolated.
After the desired hybridoma cells are identified, the clones can be subcloned by limiting dilution procedures and grown by standard methods. Suitable culture media for this puφose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 medium. Alternatively, the hybridoma cells can be grown in vivo as ascites in a mammal.
The monoclonal antibodies secreted by the subclones can be isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
The monoclonal antibodies can also be made by recombinant DNA methods, such as those described in U.S. Patent No. 4,816,567. DNA encoding the monoclonal antibodies of the invention can be readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies). The hybridoma cells of the invention serve as a preferred source of such DNA. Once isolated, the DNA can be placed into expression vectors, which are then transfected into host cells such as simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. The DNA also can be modified, for example, by substituting the coding sequence for human heavy and light chain constant domains in place of the homologous murine sequences (U.S. Patent No. 4,816,567; Morrison, Nature 368, 812-13 (1994)) or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide. Such a non-immunoglobulin polypeptide can be substituted for the constant domains of an antibody of the invention, or can be substituted for the variable domains of one antigen-combining site of an antibody of the invention to create a chimeric bivalent antibody.
Humanized Antibodies
The antibodies directed against the protein antigens of the invention can further comprise humanized antibodies or human antibodies. These antibodies are suitable for administration to humans without engendering an immune response by the human against the admimstered immunoglobulin. Humanized forms of antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen- binding subsequences of antibodies) that are principally comprised of the sequence of a human immunoglobulin, and contain minimal sequence derived from a non-human immunoglobulin. Humanization can be performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Natur , 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the coπesponding sequences of a human antibody. (See also U.S. Patent No. 5,225,539.) In some instances, Fv framework residues of the human immunoglobulin are replaced by coπesponding non-human residues. Humanized antibodies can also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions coπespond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin consensus sequence. The humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin (Jones et al, 1986; Riechmann et al., 1988; and Presta, Curr. Op. Struct. Biol, 2:593-596 (1992)). Human Antibodies
Fully human antibodies relate to antibody molecules in which essentially the entire sequences of both the light chain and the heavy chain, including the CDRs, arise from human genes. Such antibodies are termed "human antibodies", or "fully human antibodies" herein. Human monoclonal antibodies can be prepared by the trioma technique; the human B-cell hybridoma technique (see Kozbor, et al., 1983 Immunol Today 4: 72) and the EBN hybridoma technique to produce human monoclonal antibodies (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96). Human monoclonal antibodies may be utilized in the practice of the present invention and may be produced by using human hybridomas (see Cote, et al., 1983. Proc Natl Acad Sci USA 80: 2026-2030) or by transforming human B-cells with Epstein Ban Virus in vitro (see Cole, et al., 1985 In: MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc., pp. 77-96).
In addition, human antibodies can also be produced using additional techniques, including phage display libraries (Hoogenboom and Winter, J. Mol. Biol, 227:381 (1991); Marks et al., J. Mol. Biol, 222:581 (1991)). Similarly, human antibodies can be made by introducing human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon challenge, human antibody production is observed, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly, and antibody repertoire. This approach is described, for example, in U.S. Patent Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; 5,661,016, and in Marks et al. (Bio/Technology 10, 779-783 (1992)); Lonberg et al. (Nature 368 856-859 (1994)); Moπison ( Nαtwre 368, 812-13 (1994)); Fishwild et al,( Nature Biotechnology 14, 845-51 (1996)); Νeuberger (Nature Biotechnology 14, 826 (1996)); and Lonberg and Huszar (Intern. Rev. Immunol 13 65-93 (1995)).
Human antibodies may additionally be produced using transgenic nonhuman animals which are modified so as to produce fully human antibodies rather than the animal's endogenous antibodies in response to challenge by an antigen. (See PCT publication WO94/02602). The endogenous genes encoding the heavy and light immunoglobulin chains in the nonhuman host have been incapacitated, and active loci encoding human heavy and light chain immunoglobulins are inserted into the host's genome. The human genes are incoφorated, for example, using yeast artificial chromosomes containing the requisite human DΝA segments. An animal which provides all the desired modifications is then obtained as progeny by crossbreeding intermediate transgenic animals containing fewer than the full complement of the modifications. The prefeπed embodiment of such a nonhuman animal is a mouse, and is termed the Xenomouse™ as disclosed in PCT publications WO 96/33735 and WO 96/34096. This animal produces B cells which secrete fully human immunoglobulins. The antibodies can be obtained directly from the animal after immunization with an immunogen of interest, as, for example, a preparation of a polyclonal antibody, or alternatively from immortalized B cells derived from the animal, such as hybridomas producing monoclonal antibodies. Additionally, the genes encoding the immunoglobulins with human variable regions can be recovered and expressed to obtain the antibodies directly, or can be further modified to obtain analogs of antibodies such as, for example, single chain Fv molecules.
An example of a method of producing a nonhuman host, exemplified as a mouse, lacking expression of an endogenous immunoglobulin heavy chain is disclosed in U.S. Patent No. 5,939,598. It can be obtained by a method including deleting the J segment genes from at least one endogenous heavy chain locus in an embryonic stem cell to prevent reaπangement of the locus and to prevent formation of a transcript of a reaπanged immunoglobulin heavy chain locus, the deletion being effected by a targeting vector containing a gene encoding a selectable marker; and producing from the embryonic stem cell a transgenic mouse whose somatic and germ cells contain the gene encoding the selectable marker.
A method for producing an antibody of interest, such as a human antibody, is disclosed in U.S. Patent No. 5,916,771. It includes introducing an expression vector that contains a nucleotide sequence encoding a heavy chain into one mammalian host cell in culture, introducing an expression vector containing a nucleotide sequence encoding a light chain into another mammalian host cell, and fusing the two cells to form a hybrid cell. The hybrid cell expresses an antibody containing the heavy chain and the light chain.
In a further improvement on this procedure, a method for identifying a clinically relevant epitope on an immunogen, and a coπelative method for selecting an antibody that binds immunospecifically to the relevant epitope with high affinity, are disclosed in PCT publication WO 99/53049.
Fab Fragments and Single Chain Antibodies
According to the invention, techniques can be adapted for the production of single-chain antibodies specific to an antigenic protein of the invention (see e.g., U.S. Patent No. 4,946,778). In addition, methods can be adapted for the construction of Fa expression libraries (see e.g., Huse, et al., 1989 Science 246: 1275-1281) to allow rapid and effective identification of monoclonal Fa fragments with the desired specificity for a protein or derivatives, fragments, analogs or homologs thereof. Antibody fragments that contain the idiotypes to a protein antigen may be produced by techniques known in the art including, but not limited to: (i) an F(ab-)2 fragment produced by pepsin digestion of an antibody molecule; (ii) an Fa fragment generated by reducing the disulfide bridges of an F(ab')2 fragment; (iii) an Fab fragment generated by the treatment of the antibody molecule with papain and a reducing agent and (iv) Fv fragments.
Bispecific Antibodies
Bispecific antibodies are monoclonal, preferably human or humanized, antibodies that have binding specificities for at least two different antigens. In the present case, one of the binding specificities is for an antigenic protein of the invention. The second binding target is any other antigen, and advantageously is a cell-surface protein or receptor or receptor subunit.
Methods for making bispecific antibodies are known in the art. Traditionally, the recombinant production of bispecific antibodies is based on the co-expression of two immunoglobulin heavy-chain/light-chain pairs, where the two heavy chains have different specificities (Milstein and Cuello, Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture often different antibody molecules, of which only one has the coπect bispecific structure. The purification of the coπect molecule is usually accomplished by affinity chromatography steps. Similar procedures are disclosed in WO 93/08829, published 13 May 1993, and in Traunecker et al, 1991 EMBO J, 10:3655-3659.
Antibody variable domains with the desired binding specificities (antibody-antigen combining sites) can be fused to immunoglobulin constant domain sequences. The fusion preferably is with an immunoglobulin heavy-chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is prefeπed to have the first heavy-chain constant region (CHI) containing the site necessary for light-chain binding present in at least one of the fusions. DNAs encoding the immunoglobulin heavy-chain fusions and, if desired, the immunoglobulin light chain, are inserted into separate expression vectors, and are co- transfected into a suitable host organism. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121:210 (1986).
According to another approach described in WO 96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The prefeπed interface comprises at least a part of the CH3 region of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory "cavities" of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab')2 bispecific antibodies). Techniques for generating bispecific antibodies from antibody fragments have been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science 229:81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab')2 fragments. These fragments are reduced in the presence of the dithiol comRho-Interacting Proteing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab' fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab'-TNB derivatives is then reconverted to the Fab' -thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
Additionally, Fab' fragments can be directly recovered from E. coli and chemically coupled to form bispecific antibodies. Shalaby et al., J. Exp. Med. 175:217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab')2 molecule. Each Fab' fragment was separately secreted from E. coli and subjected to directed chemical coupling in- vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol. 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The "diabody" technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See, Gruber et al, J. Immunol. 152:5368 (1994).
Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al., J. Immunol. 147:60 (1991).
Exemplary bispecific antibodies can bind to two different epitopes, at least one of which originates in the protein antigen of the invention. Alternatively, an anti-antigenic arm of an immunoglobulin molecule can be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2, CD3, CD28, or B7), or Fc receptors for IgG (FcγR), such as FcγRI (CD64), FcγRII (CD32) and FcγRIII (CD16) so as to focus cellular defense mechanisms to the cell expressing the particular antigen. Bispecific antibodies can also be used to direct cytotoxic agents to cells which express a particular antigen. These antibodies possess an antigen-binding arm and an arm which binds a cytotoxic agent or a radionuclide chelator, such as EOTUBE, DPTA, DOTA, or TETA. Another bispecific antibody of interest binds the protein antigen described herein and further binds tissue factor (TF).
Heteroconjugate Antibodies
Heteroconjugate antibodies are also within the scope of the present invention. Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Patent No. 4,676,980), and for treatment of HIV infection (WO 91/00360; WO 92/200373; EP 03089). It is contemplated that the antibodies can be prepared in vitro using known methods in synthetic protein chemistry, including those, involving crosslinking agents. For example, immunotoxins can be constructed using a disulfide exchange reaction or by forming a thioether bond. Examples of suitable reagents for this p npose include iminothiolate and methyl-4-mercaptobutyrimidate and those disclosed, for example, in U.S. Patent No. 4,676,980. Effector Function Engineering
It can be desirable to modify the antibody of the invention with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating cancer. For example, cysteine residue(s) can be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region. The homodimeric antibody thus generated can have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med., 176: 1191- 1195 (1992) and Shopes, J. hnmunol., 148: 2918-2922 (1992). Homodimeric antibodies with enhanced anti-tumor activity can also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research, 53: 2560-2565 (1993). Alternatively, an antibody can be engineered that has dual Fc regions and can thereby have enhanced complement lysis and ADCC capabilities. See Stevenson et al., Anti-Cancer Drug Design, 3: 219-230 (1989).
Immunoconjugates
The invention also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a chemotherapeutic agent, toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
Chemotherapeutic agents useful in the generation of such immunoconjugates have been described above. Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes. A variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212Bi, 1311, 131hι, 90Y, and 186Re.
Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis- diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro- 2,4-dinitrobenzene). For example, a ricin immunotoxin can be prepared as described in Vitetta et al., Science, 238: 1098 (1987). Carbon- 14-labeled l-isothiocyanatobenzyl-3- methyldiethylene uiaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. See WO94/11026.
In another embodiment, the antibody can be conjugated to a "receptor" (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a "ligand" (e.g., avidin) that is in turn conjugated to a cytotoxic agent.
In one embodiment, methods for the screening of antibodies that possess the desired specificity include, but are not limited to, enzyme-linked immunosorbent assay (ELISA) and other immunologically-mediated techniques known within the art. In a specific embodiment, selection of antibodies that are specific to a particular domain of an NOVX protein is facilitated by generation of hybridomas that bind to the fragment of an NOVX protein possessing such a domain. Thus, antibodies that are specific for a desired domain within an NOVX protein, or derivatives, fragments, analogs or homologs thereof, are also provided herein.
Anti-NOVX antibodies may be used in methods known within the art relating to the localization and/or quantisation of an NOVX protein (e.g., for use in measuring levels of the NOVX protein within appropriate physiological samples, for use in diagnostic methods, for use in imaging the protein, and the like). In a given embodiment, antibodies for NOVX proteins, or derivatives, fragments, analogs or homologs thereof, that contain the antibody derived binding domain, are utilized as pharmacologically-active compounds (hereinafter "Therapeutics").
An anti-NOVX antibody (e.g., monoclonal antibody) can be used to isolate an NOVX polypeptide by standard techniques, such as affinity chromatography or immunoprecipitation. An anti-NOVX antibody can facilitate the purification of natural NOVX polypeptide from cells and of recombinantly-produced NOVX polypeptide expressed in host cells. Moreover, an anti-NOVX antibody can be used to detect NOVX protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the NOVX protein. Anti-NOVX antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include I, 131I, 35S or 3H.
NOVX Recombinant Expression Vectors and Host Cells
Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding an NOVX protein, or derivatives, fragments, analogs or homologs thereof. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively-linked. Such vectors are refeπed to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno-associated viruses), which serve equivalent functions.
The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, that is operatively-linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably-linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner that allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
The term "regulatory sequence" is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Regulatory sequences include those that direct constitutive expression of a nucleotide sequence in many types of host cell and those that direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., NOVX proteins, mutant forms of NOVX protems, fusion proteins, etc.).
The recombinant expression vectors of the invention can be designed for expression of NOVX proteins in prokaryotic or eukaryotic cells. For example, NOVX proteins can be expressed in bacterial cells such as Escherichia coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in Escherichia coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three puφoses: (i) to increase expression of recombinant protein; (ii) to increase the solubility of the recombinant protein; and (iii) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith and Johnson, 1988. Gene 67: 31-40), pMAL (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N. J.) that fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amrann et al, (1988) Gene 69:301-315) and pET 1 Id (Studier et al, GENE EXPRESSION TECHNOLOGY: METHODS πsr ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 60-89).
One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein. See, e.g., Gottesman, GENE EXPRESSION TECHNOLOGY: METHODS IN ENZYMOLOGY 185, Academic Press, San Diego, Calif. (1990) 119-128. Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (see, e.g., Wada, et al, 1992. Nucl. Acids Res. 20: 2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
In another embodiment, the NOVX expression vector is a yeast expression vector. Examples of vectors for expression in yeast Saccharomyces cerivisae include pYepSecl (Baldari, et al, 1987. EMBOJ. 6: 229-234), pMFa (Kurjan and Herskowitz, 1982. Cell 30: 933-943), pJRY88 (Schultz et al, 1987. Gene 54: 113-123), pYES2 (hivifrogen Coφoration, San Diego, Calif), and picZ (InVitrogen Coφ, San Diego, Calif).
Alternatively, NOVX can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., SF9 cells) include the pAc series (Smith, et al, 1983. Mol. Cell. Biol. 3: 2156-2165) and the pVL series (Lucklow and Summers, 1989. Virology 170: 31-39).
In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, 1987. Nature 329: 840) and pMT2PC (Kaufinan, et al, 1987. EMBO J. 6: 187-195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, adenovirus 2, cytomegalovirus, and simian virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see, e.g., Chapters 16 and 17 of Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989.
In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue-specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert, et al, 1987. Genes Dev. 1: 268-277), lymphoid-specific promoters (Calame and Eaton, 1988. Adv. Immunol. 43: 235-275), in particular promoters of T cell receptors (Winoto and Baltimore, 1989. EMBO J. 8: 729-733) and immunoglobulins (Banerji, et al, 1983. Cell 33: 729-740; Queen and Baltimore, 1983. Cell 33: 741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle, 1989. Proc. Natl. Acad. Sci. USA 86: 5473-5477), pancreas-specific promoters (Edlund, et al, 1985. Science 230: 912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Pat. No. 4,873,316 and European Application Publication No. 264,166). Developmentally-regulated promoters are also encompassed, e.g., the murine hox promoters (Kessel and Gruss, 1990. Science 249: 374-379) and the -fetoprotein promoter (Campes and Tilghman, 1989. Genes Dev. 3: 537-546).
The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively-linked to a regulatory sequence in a manner that allows for expression (by transcription of the DNA molecule) of an RNA molecule that is antisense to NOVX mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen that direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen that direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see, e.g., Weintraub, et al., "Antisense RNA as a molecular tool for genetic analysis," Reviews-Trends in Genetics, Vol. 1(1) 1986.
Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, NONX protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
Vector DΝA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DΝA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DΕAΕ-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (MOLECULAR CLONING: A LABORATORY MANUAL. 2nd ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Various selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding NOVX or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incoφorated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) NOVX protein. Accordingly, the invention further provides methods for producing NOVX protein using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding NOVX protein has been introduced) in a suitable medium such that NOVX protein is produced. In another embodiment, the method further comprises isolating NOVX protein from the medium or the host cell. Transgenic NOVX Animals
The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which NOVX protein-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous NOVX sequences have been introduced into their genome or homologous recombinant animals in which endogenous NOVX sequences have been altered. Such animals are useful for studying the function and or activity of NOVX protein and for identifying and/or evaluating modulators of NOVX protein activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, etc. A transgene is exogenous DNA that is integrated into the genome of a cell from which a transgenic animal develops and that remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous NOVX gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
A transgenic animal of the invention can be created by introducing NOVX-encoding nucleic acid into the male pronuclei of a fertilized oocyte (e.g., by micromjection, retroviral infection) and allowing the oocyte to develop in a pseudopregnant female foster animal. The human NOVX cDNA sequences SEQ ID NOS.l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27,
29, 31, 33, and 35 can be introduced as a transgene into the genome of a non-human animal.
Alternatively, a non-human homologue of the human NOVX gene, such as a mouse NOVX gene, can be isolated based on hybridization to the human NOVX cDNA (described further supra) and used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably-linked to the NOVX transgene to direct expression of NOVX protein to particular cells. Methods for generating transgenic animals via embryo manipulation and micromjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866;
4,870,009; and 4,873,191; and Hogan, 1986. In: MANIPULATING THE MOUSE EMBRYO, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NN. Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of the ΝOVX transgene in its genome and/or expression of ΝOVX mRΝA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals caπying the transgene. Moreover, transgenic animals carrying a transgene- encoding ΝOVX protein can further be bred to other transgenic animals carrying other transgenes.
To create a homologous recombinant animal, a vector is prepared which contains at least a portion of an ΝOVX gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the ΝOVX gene. The ΝOVX gene can be a human gene (e.g., the cDΝA of SEQ ID ΝOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31 , and 33), but more preferably, is a non-human homologue of a human NOVX gene. For example, a mouse homologue of human NOVX gene of SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33 can be used to construct a homologous recombination vector suitable for altering an endogenous NOVX gene in the mouse genome. In one embodiment, the vector is designed such that, upon homologous recombination, the endogenous NOVX gene is functionally disrupted (i.e., no longer encodes a functional protein; also refeπed to as a "knock out" vector).
Alternatively, the vector can be designed such that, upon homologous recombination, the endogenous NOVX gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous NOVX protein), h the homologous recombination vector, the altered portion of the NOVX gene is flanked at its 5'- and 3'-termini by additional nucleic acid of the NOVX gene to allow for homologous recombination to occur between the exogenous NOVX gene carried by the vector and an endogenous NOVX gene in an embryonic stem cell. The additional flanking NOVX nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5'- and 3'-termini) are included in the vector. See, e.g., Thomas, et al, 1987. Cell 51 : 503 for a description of homologous recombination vectors. The vector is ten introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced NOVX gene has homologously-recombined with the endogenous NOVX gene are selected. See, e.g., Li, et al, 1992. Cell 69: 915.
The selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras. See, e.g., Bradley, 1987. In: TERATOCARCINOMAS AND EMBRYONIC STEM CELLS: A PRACTICAL APPROACH, Robertson, ed. IRL, Oxford, pp. 113-152. A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously-recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously-recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley, 1991. Curr. Opin. Biotechnol 2: 823-829; PCT International Publication Nos.: WO 90/11354; WO 91/01140; WO 92/0968; and WO 93/04169.
In another embodiment, transgenic non-humans animals can be produced that contain selected systems that allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage PI. For a description of the cre/loxP recombinase system, See, e.g., Lakso, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae. See, O'Gorman, et al, 1991. Science 251:1351-1355. Ifa cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, et al, 1997. Nature 385: 810-813. In brief, a cell (e.g., a somatic cell) from the transgenic animal can be isolated and induced to exit the growth cycle and enter G0 phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The reconstructed oocyte is then cultured such that it develops to morula or blastocyte and then transfeπed to pseudopregnant female foster animal. The offspring borne of this female foster animal will be a clone of the animal from which the cell (e.g., the somatic cell) is isolated.
Pharmaceutical Compositions
The NOVX nucleic acid molecules, NOVX proteins, and anti-NOVX antibodies (also refeπed to herein as "active compounds") of the invention, and derivatives, fragments, analogs and homologs thereof, can be incoφorated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incoφorated herein by reference. Prefeπed examples of such carriers or diluents include, but are not limited to, water, saline, finger's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), fransmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL (BASF, Parsippany, N. J.) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin.
Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., an NOVX protein or anti-NOVX antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring. For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penefrants appropriate to the barrier to be permeated are used in the formulation. Such penefrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery. hi one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycohc acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Coφoration and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals. The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see, e.g., U.S. Patent No. 5,328,470) or by stereotactic injection (see, e.g., Chen, et al, 1994. Proc. Natl. Acad. Sci. USA 91: 3054-3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells that produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
Screening and Detection Methods
The isolated nucleic acid molecules of the invention can be used to express NOVX protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect NOVX mRNA (e.g., in a biological sample) or a genetic lesion in an NOVX gene, and to modulate NOVX activity, as described further, below. In addition, the NOVX proteins can be used to screen drugs or compounds that modulate the NOVX protein activity or expression as well as to treat disorders characterized by insufficient or excessive production of NOVX protein or production of NOVX protein forms that have decreased or abeπant activity compared to NOVX wild-type protein (e.g.; diabetes (regulates insulin release); obesity (binds and transport lipids); metabolic disturbances associated with obesity, the metabolic syndrome X as well as anorexia and wasting disorders associated with chronic diseases and various cancers, and infectious disease(possesses anti-microbial activity) and the various dyslipidemias. In addition, the anti-NOVX antibodies of the invention can be used to detect and isolate NOVX proteins and modulate NOVX activity. In yet a further aspect, the invention can be used in methods to influence appetite, absoφtion of nutrients and the disposition of metabolic substrates in both a positive and negative fashion.
The invention further pertains to novel agents identified by the screening assays described herein and uses thereof for treatments as described, supra.
Screening Assays
The invention provides a method (also refeπed to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) that bind to NOVX proteins or have a stimulatory or inhibitory effect on, e.g., NOVX protein expression or NOVX protein activity. The invention also includes compounds identified in the screening assays described herein.
In one embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of the membrane-bound form of an NOVX protein or polypeptide or biologically-active portion thereof. The test compounds of the invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the "one-bead one-compound" library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds. See, e.g., Lam, 1997 '. Anticancer Drug Design 12: 145.
A "small molecule" as used herein, is meant to refer to a composition that has a molecular weight of less than about 5 kD and most preferably less than about 4 kD. Small molecules can be, e.g., nucleic acids, peptides, polypeptides, peptidomimetics, carbohydrates, lipids or other organic or inorganic molecules. Libraries of chemical and/or biological mixtures, such as fungal, bacterial, or algal extracts, are known in the art and can be screened with any of the assays of the invention.
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt, et al, 1993. Proc. Natl. Acad. Sci. U.S.A. 90: 6909; Erb, et al, 1994. Proc. Natl. Acad. Sci. U.S.A. 91: 11422; Zuckermann, et al, 1994. J. Med. Chem. 37: 2678; Cho, et al, 1993. Science 261: 1303; Caπell, et al, 1994. Angew. Chem. Int. Ed. Engl. 33: 2059; Carell, et al, 1994. Angew. Chem. Int. Ed. Engl. 33: 2061; and Gallop, et al, 1994. J. Med. Chem. 37: 1233.
Libraries of compounds maybe presented in solution (e.g., Houghten, 1992. Biotechniques 13: 412-421), or on beads (Lam, 1991. Nature 354: 82-84), on chips (Fodor, 1993. Nature 364: 555-556), bacteria (Ladner, U.S. Patent No. 5,223,409), spores (Ladner, U.S. Patent 5,233,409), plasmids (Cull, et al, 1992. Proc. Natl. Acad. Sci. USA 89: 1865-1869) or on phage (Scott and Smith, 1990. Science 249: 386-390; Devlin, 1990. Science 249: 404-406; Cwirla, et al, 1990. Proc. Natl. Acad. Sci. U.S.A. 87: 6378-6382; Felici, 1991. J. Mol. Biol 222: 301-310; Ladner, U.S. Patent No. 5,233,409.). hi one embodiment, an assay is a cell-based assay in which a cell which expresses a membrane-bound form of NOVX protein, or a biologically-active portion thereof, on the cell surface is contacted with a test compound and the ability of the test compound to bind to an NONX protein determined. The cell, for example, can of mammalian origin or a yeast cell. Determining the ability of the test compound to bind to the ΝONX protein can be accomplished, for example, by coupling the test compound with a radioisotope or enzymatic label such that binding of the test compound to the ΝONX protein or biologically-active portion thereof can be determined by detecting the labeled compound in a complex. For example, test compounds can be labeled with 1251, 35S, 14C, or 3H, either directly or indirectly, and the radioisotope detected by direct counting of radioemission or by scintillation counting. Alternatively, test compounds can be enzymatically-labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product, fri one embodiment, the assay comprises contacting a cell which expresses a membrane-bound form of ΝONX protein, or a biologically-active portion thereof, on the cell surface with a known compound which binds ΝONX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an ΝONX protein, wherein determining the ability of the test compound to interact with an ΝOVX protein comprises determining the ability of the test compound to preferentially bind to ΝOVX protein or a biologically-active portion thereof as compared to the known compound.
In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing a membrane-bound form of ΝOVX protein, or a biologically-active portion thereof, on the cell surface with a test compound and determining the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the ΝOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of ΝOVX or a biologically-active portion thereof can be accomplished, for example, by determining the ability of the ΝOVX protein to bind to or interact with an ΝOVX target molecule. As used herein, a "target molecule" is a molecule with which an ΝOVX protein binds or interacts in nature, for example, a molecule on the surface of a cell which expresses an ΝOVX interacting protein, a molecule on the surface of a second cell, a molecule in the extracellular milieu, a molecule associated with the internal surface of a cell membrane or a cytoplasmic molecule. An ΝOVX target molecule can be a non-ΝOVX molecule or an ΝOVX protein or polypeptide of the invention, hi one embodiment, an ΝOVX target molecule is a component of a signal transduction pathway that facilitates transduction of an extracellular signal (e.g. a signal generated by binding of a compound to a membrane-bound ΝOVX molecule) through the cell membrane and into the cell. The target, for example, can be a second intercellular protein that has catalytic activity or a protein that facilitates the association of downstream signaling molecules with NOVX.
Determining the ability of the NOVX protein to bind to or interact with an NOVX target molecule can be accomplished by one of the methods described above for determining direct binding. In one embodiment, determining the ability of the NOVX protein to bind to or interact with an NOVX target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e. intracellular Ca2+, diacylglycerol, IP3, etc.), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising an NOVX-responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a cellular response, for example, cell survival, cellular differentiation, or cell proliferation.
In yet another embodiment, an assay of the invention is a cell-free assay comprising contacting an NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to bind to the NOVX protein or biologically- active portion thereof. Binding of the test compound to the NOVX protein can be determined either directly or indirectly as described above. In one such embodiment, the assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an NOVX protein, wherein determining the ability of the test compound to interact with an NOVX protein comprises determining the ability of the test compound to preferentially bind to NOVX or biologically-active portion thereof as compared to the known compound. hi still another embodiment, an assay is a cell-free assay comprising contacting NOVX protein or biologically-active portion thereof with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the NOVX protein or biologically-active portion thereof. Determining the ability of the test compound to modulate the activity of NOVX can be accomplished, for example, by determimng the ability of the NOVX protein to bind to an NOVX target molecule by one of the methods described above for determining direct binding. In an alternative embodiment, determimng the ability of the test compound to modulate the activity of NOVX protein can be accomplished by determining the ability of the NOVX protein further modulate an NOVX target molecule. For example, the catalytic/enzymatic activity of the target molecule on an appropriate substrate can be determined as described, supra.
In yet another embodiment, the cell-free assay comprises contacting the NOVX protein or biologically-active portion thereof with a known compound which binds NOVX protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an NOVX protein, wherein determining the ability of the test compound to interact with an NOVX protein comprises determining the ability of the NOVX protein to preferentially bind to or modulate the activity of an NOVX target molecule.
The cell-free assays of the invention are amenable to use of both the soluble form or the membrane-bound form of NOVX protein. In the case of cell-free assays comprising the membrane-bound form of NOVX protein, it may be desirable to utilize a solubilizing agent such that the membrane-bound form of NOVX protein is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl-N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-100, Triton® X-114, Thesit®, Isotridecypoly(ethylene glycol ether)n, N-dodecyl~N,N-dimethyl-3-ammonio-l -propane sulfonate, 3-(3-cholamidopropyl) dimethylamminiol-1 -propane sulfonate (CHAPS), or 3-(3-cholamidopropyl)dimethylamminiol-2-hydroxy- 1 -propane sulfonate (CHAPSO). hi more than one embodiment of the above assay methods of the invention, it maybe desirable to immobilize either NOVX protein or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to NOVX protein, or interaction of NOVX protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtiter plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided that adds a domain that allows one or both of the proteins to be bound to a matrix. For example, GST-NOVX fusion proteins or GST-target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtiter plates, that are then combined with the test compound or the test compound and either the non-adsorbed target protein or NOVX protein, and the mixture is incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtiter plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described, supra. Alternatively, the complexes can be dissociated from the matrix, and the level of NOVX protein binding or activity determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either the NOVX protein or its target molecule can be immobilized utilizing conjugation of biotin and sfreptavidin. Biotinylated NOVX protein or target molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well-known within the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, 111.), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with NOVX protein or target molecules, but which do not interfere with binding of the NOVX protein to its target molecule, can be derivatized to the wells of the plate, and unbound target or NOVX protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the NOVX protein or target molecule, as well as enzyme-linked assays that rely on detecting an enzymatic activity associated with the NOVX protein or target molecule. hi another embodiment, modulators of NOVX protein expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of NOVX mRNA or protein in the cell is determined. The level of expression of NOVX mRNA or protein in the presence of the candidate compound is compared to the level of expression of NOVX mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of NOVX mRNA or protein expression based upon this comparison. For example, when expression of NOVX mRNA or protein is greater (i.e., statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of NOVX mRNA or protein expression. Alternatively, when expression of NOVX mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of NOVX mRNA or protein expression. The level of NOVX mRNA or protein expression in the cells can be determined by methods described herein for detecting NOVX mRNA or protein.
In yet another aspect of the invention, the NOVX proteins can be used as "bait proteins" in a two-hybrid assay or three hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos, et al, 1993. Ce// 72: 223-232; Madura, et al, 1993. J. Biol. Chem. 268: 12046-12054; Bartel, et al, 1993. Biotechniques 14: 920-924; Iwabuchi, et al, 1993. Oncogene 8: 1693-1696; and Brent WO 94/10300), to identify other proteins that bind to or interact with NOVX ("NOVX-binding proteins" or "NOVX-bp") and modulate NOVX activity. Such NOVX-binding proteins are also likely to be involved in the propagation of signals by the NOVX proteins as, for example, upstream or downstream elements of the NOVX pathway.
The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constructs. In one construct, the gene that codes for NOVX is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other construct, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming an NOVX-dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) that is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene that encodes the protein which interacts with NOVX.
The invention further pertains to novel agents identified by the aforementioned screening assays and uses thereof for treatments as described herein.
Detection Assays
Portions or fragments of the cDNA sequences identified herein (and the coπesponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. By way of example, and not of limitation, these sequences can be used to: (ϊ) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Some of these applications are described in the subsections, below.
Chromosome Mapping
Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the NOVX sequences, SEQ ED NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or fragments or derivatives thereof, can be used to map the location of the NOVX genes, respectively, on a chromosome. The mapping of the NOVX sequences to chromosomes is an important first step in coπelating these sequences with genes associated with disease.
Briefly, NOVX genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the NOVX sequences. Computer analysis of the NOVX, sequences can be used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene coπesponding to the NOVX sequences will yield an amplified fragment.
Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but in which human cells can, the one human chromosome that contains the gene encoding the needed enzyme will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. See, e.g., D'Eustachio, et al, 1983. Science 220: 919-924. Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the NOVX sequences to design oligonucleotide primers, sub- localization can be achieved with panels of fragments from specific chromosomes.
Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical like colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1,000 bases, and more preferably 2,000 bases, will suffice to get good results at a reasonable amount of time. For a review of this technique, see, Verma, et al, HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES (Pergamon Press, New York 1988).
Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents coπesponding to noncoding regions of the genes actually are prefeπed for mapping puφoses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be coπelated with genetic map data. Such data are found, e.g., in McKusick, MENDELIAN INHERITANCE IN MAN, available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, e.g., Egeland, et al, 1987. Nature, 325: 783-787.
Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the NONX gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DΝA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymoφhisms.
Tissue Typing
The ΝOVX sequences of the invention can also be used to identify individuals from minute biological samples. In this technique, an individual's genomic DΝA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. The sequences of the invention are useful as additional DΝA markers for RFLP ("restriction fragment length polymoφhisms," described in U.S. Patent No. 5,272,057). Furthermore, the sequences of the invention can be used to provide an alternative technique that determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the NOVX sequences described herein can be used to prepare two PCR primers from the 5'- and 3'-termini of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it.
Panels of coπesponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the invention can be used to obtain such identification sequences from individuals and from tissue. The NOVX sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Much of the allelic variation is due to single nucleotide polymoφhisms (SNPs), which include restriction fragment length polymoφhisms (RFLPs).
Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification puφoses. Because greater numbers of polymoφhisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers that each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as those in SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, are used, a more appropriate number of primers for positive individual identification would be 500-2,000.
Predictive Medicine
The invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, pharmacogenomics, and monitoring clinical trials are used for prognostic (predictive) puφoses to thereby treat an individual prophylactically. Accordingly, one aspect of the invention relates to diagnostic assays for determining NOVX protein and/or nucleic acid expression as well as NOVX activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with abeπant NOVX expression or activity. The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. For example, mutations in an NOVX gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive puφose to thereby prophylactically treat an individual prior to the onset of a disorder characterized by or associated with NOVX protein, nucleic acid expression, or biological activity.
Another aspect of the invention provides methods for determining NOVX protein, nucleic acid expression or activity in an individual to thereby select appropriate therapeutic or prophylactic agents for that individual (refeπed to herein as "pharmacogenomics"). Pharmacogenomics allows for the selection of agents (e.g., drugs) for therapeutic or prophylactic treatment of an individual based on the genotype of the individual (e.g., the genotype of the individual examined to determine the ability of the individual to respond to a particular agent.)
Yet another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NOVX in clinical trials.
These and other agents are described in further detail in the following sections.
Diagnostic Assays
An exemplary method for detecting the presence or absence of NOVX in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes NOVX protein such that the presence of NOVX is detected in the biological sample. An agent for detecting NOVX mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to NOVX mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length NOVX nucleic acid, such as the nucleic acid of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, and 33, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to NOVX mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein. An agent for detecting NOVX protein is an antibody capable of binding to NOVX protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently- labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect NOVX mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of NOVX mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of NOVX protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, and immunofluorescence. In vitro techniques for detection of NOVX genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of NOVX protein include introducing into a subject a labeled anti-NOVX antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A prefeπed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject.
In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting NOVX protein, mRNA, or genomic DNA, such that the presence of NOVX protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of NOVX protein, mRNA or genomic DNA in the control sample with the presence of NOVX protein, mRNA or genomic DNA in the test sample.
The invention also encompasses kits for detecting the presence of NOVX in a biological sample. For example, the kit can comprise: a labeled compound or agent capable of detecting NOVX protein or mRNA in a biological sample; means for determining the amount of NOVX in the sample; and means for comparing the amount of NOVX in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect NOVX protein or nucleic acid.
Prognostic Assays
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with abeπant NOVX expression or activity. For example, the assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with NOVX protein, nucleic acid expression or activity. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disease or disorder. Thus, the invention provides a method for identifying a disease or disorder associated with abeπant NOVX expression or activity in which a test sample is obtained from a subject and NOVX protein or nucleic acid (e.g., mRNA, genomic DNA) is detected, wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with abeπant NOVX expression or activity. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be admimstered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drug candidate) to treat a disease or disorder associated with abeπant NOVX expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a disorder. Thus, the invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with abeπant NOVX expression or activity in which a test sample is obtained and NOVX protein or nucleic acid is detected (e.g., wherein the presence of NOVX protein or nucleic acid is diagnostic for a subject that can be administered the agent to treat a disorder associated with abeπant NOVX expression or activity).
The methods of the invention can also be used to detect genetic lesions in an NOVX gene, thereby determining if a subject with the lesioned gene is at risk for a disorder characterized by abeπant cell proliferation and/or differentiation, hi various embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic lesion characterized by at least one of an alteration affecting the integrity of a gene encoding an NOVX-protein, or the misexpression of the NOVX gene. For example, such genetic lesions can be detected by ascertaining the existence of at least one of: (i) a deletion of one or more nucleotides from an NOVX gene; (ii) an addition of one or more nucleotides to an NOVX gene; (iii) a substitution of one or more nucleotides of an NOVX gene, (iv) a chromosomal reaπangement of an NOVX gene; (v) an alteration in the level of a messenger RNA transcript of an NOVX gene, (vi) abeπant modification of an NOVX gene, such as of the methylation pattern of the genomic DNA, (vii) the presence of a non- wild-type splicing pattern of a messenger RNA transcript of an NOVX gene, (viii) a non-wild-type level of an NOVX protein, (ix) allelic loss of an NOVX gene, and (x) inappropriate post-translational modification of an NOVX protein. As described herein, there are a large number of assay techniques known in the art which can be used for detecting lesions in an NOVX gene. A prefeπed biological sample is a peripheral blood leukocyte sample isolated by conventional means from a subject. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
In certain embodiments, detection of the lesion involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran, et αl, 1988. Science 241 : 1077-1080; and Nakazawa, et αl., 1994. Proc. Nαtl Acαd. Sci. USA 91 : 360-364), the latter of which can be particularly useful for detecting point mutations in the NONX-gene (see, Abravaya, et αl., 1995. Nucl. Acids Res. 23: 675-682). This method can include the steps of collecting a sample of cells from a patient, isolating nucleic acid (e.g., genomic, mRΝA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers that specifically hybridize to an ΝONX gene under conditions such that hybridization and amplification of the ΝONX gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
Alternative amplification methods include: self sustained sequence replication (see, Guatelli, et αl., 1990. Proc. Nαtl. Acαd. Sci. USA 87: 1874-1878), transcriptional amplification system (see, Kwoh, et αl, 1989. Proc. Nαtl. Acαd. Sci. USA 86: 1173-1177); Qβ Replicase (see, Lizardi, et αl, 1988. BioTechnology 6: 1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
In an alternative embodiment, mutations in an NONX gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DΝA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DΝA indicates mutations in the sample DΝA. Moreover, the use of sequence specific ribozymes (see, e.g., U.S. Patent No. 5,493,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In other embodiments, genetic mutations in NONX can be identified by hybridizing a sample and control nucleic acids, e.g., DΝA or RΝA, to high-density aπays containing hundreds or thousands of oligonucleotides probes. See, e.g., Cronin, et al, 1996. Human Mutation 1: 244-255; Kozal, et al, 1996. Nat. Med. 2: 753-759. For example, genetic mutations in ΝONX can be identified in two dimensional aπays containing light-generated DΝA probes as described in Cronin, et al, supra. Briefly, a first hybridization aπay of probes can be used to scan through long stretches of DΝA in a sample and control to identify base changes between the sequences by making linear aπays of sequential overlapping probes. This step allows the identification of point mutations. This is followed by a second hybridization aπay that allows the characterization of specific mutations by using smaller, specialized probe aπays complementary to all variants or mutations detected. Each mutation aπay is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene. hi yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the ΝOVX gene and detect mutations by comparing the sequence of the sample ΝOVX with the coπesponding wild-type (confrol) sequence. Examples of sequencing reactions include those based on techniques developed by Maxim and Gilbert, 1977. Proc. Natl. Acad. Sci. USA 74: 560 or Sanger, 1977. Proc. Natl. Acad. Sci. USA 74: 5463. It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays (see, e.g., Νaeve, et al, 1995. Biotechniques 19: 448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen, et al, 1996. Adv. Chromatography 36: 127-162; and Griffin, et al, 1993. Appl. Biochem. Biotechnol. 38: 147-159). Other methods for detecting mutations in the NOVX gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes. See, e.g., Myers, et al, 1985. Science 230: 1242. In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type NOVX sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent that cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA DNA hybrids treated with Si nuclease to enzymatically digesting the mismatched regions, hi other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, e.g., Cotton, et al, 1988. Proc. Natl Acad. Sci. USA 85: 4397; Saleeba, et al, 1992. Methods Enzymol 217: 286-295. In an embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in NOVX cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches. See, e.g., Hsu, et al, 1994. Carcinogenesis 15: 1657-1662. According to an exemplary embodiment, a probe based on an NOVX sequence, e.g., a wild-type NOVX sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, e.g., U.S. Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in NOVX genes. For example, single strand conformation polymoφhism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids. See, e.g., Orita, et al, 1989. Proc. Natl. Acad. Sci. USA: 86: 2766; Cotton, 1993. Mutat. Res. 285: 125-144; Hayashi, 1992. Genet. Anal. Tech. Appl 9: 73-79. Single-stranded DNA fragments of sample and control NOVX nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In one embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility. See, e.g., Keen, et al, 1991. Trends Genet. 1: 5.
In yet another embodiment, the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE). See, e.g., Myers, et al, 1985. Nature 313: 495. When DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA. See, e.g., Rosenbaum and Reissner, 1987 '. Biophys. Chem. 265: 12753.
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions that permit hybridization only if a perfect match is found. See, e.g., Saiki, et al, 1986. Nature 324: 163; Saiki, et al, 1989. Proc. Natl. Acad. Sci. USA 86: 6230. Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
Alternatively, allele specific amplification technology that depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may cany the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization; see, e.g., Gibbs, et al, 1989. Nucl. Acids Res. 17: 2437-2448) or at the extreme 3'-terminus of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (see, e.g., Prossner, 1993. Tibtech. 11 : 238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection. See, e.g., Gasparini, et al, 1992. Mol. Cell Probes 6: 1. It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification. See, e.g., Barany, 1991. Proc. Natl. Acad. Sci. USA 88: 189. In such cases, ligation will occur only if there is a perfect match at the 3'-terminus of the 5' sequence, making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
The methods described herein may be performed, for example, by utilizing pre-packaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving an NONX gene.
Furthermore, any cell type or tissue, preferably peripheral blood leukocytes, in which ΝONX is expressed may be utilized in the prognostic assays described herein. However, any biological sample containing nucleated cells may be used, including, for example, buccal mucosal cells.
Pharmacogenomics
Agents, or modulators that have a stimulatory or inhibitory effect on ΝONX activity (e.g., ΝONX gene expression), as identified by a screening assay described herein can be admimstered to individuals to treat (prophylactically or therapeutically) disorders (The disorders include metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer- associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, and hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.) hi conjunction with such treatment, the pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) of the individual may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, the pharmacogenomics of the individual permits the selection of effective agents (e.g., drugs) for prophylactic or therapeutic treatments based on a consideration of the individual's genotype. Such pharmacogenomics can further be used to determine appropriate dosages and therapeutic regimens. Accordingly, the activity of ΝONX protein, expression of ΝONX nucleic acid, or mutation content of ΝONX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual.
Pharmacogenomics deals with clinically significant hereditary variations in the response to drugs due to altered drug disposition and abnormal action in affected persons. See e.g., Eichelbaum, 1996. Clin. Exp. Pharmacol. Physiol, 23: 983-985; Linder, 1997. Clin. Chem., 43: 254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drugs act on the body (altered drug action) or genetic conditions transmitted as single factors altering the way the body acts on drugs (altered drug metabolism). These pharmacogenetic conditions can occur either as rare defects or as polymoφhisms. For example, glucose-6-phosphate dehydrogenase (G6PD) deficiency is a common inherited enzymopathy in which the main clinical complication is hemolysis after ingestion of oxidant drugs (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.
As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymoφhisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drug response and serious toxicity after taking the standard and safe dose of a drug. These polymoφhisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymoφhic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drug response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite moφhine. At the other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
Thus, the activity of NONX protein, expression of ΝONX nucleic acid, or mutation content of ΝONX genes in an individual can be determined to thereby select appropriate agent(s) for therapeutic or prophylactic treatment of the individual. In addition, pharmacogenetic studies can be used to apply genotyping of polymoφhic alleles encoding drug-metabolizing enzymes to the identification of an individual's drug responsiveness phenotype. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an ΝONX modulator, such as a modulator identified by one of the exemplary screening assays described herein. Monitoring of Effects During Clinical Trials
Monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of NONX (e.g., the ability to modulate abeπant cell proliferation and/or differentiation) can be applied not only in basic drug screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to increase ΝOVX gene expression, protein levels, or upregulate ΝOVX activity, can be monitored in clinical trails of subjects exhibiting decreased ΝOVX gene expression, protein levels, or downregulated ΝOVX activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease ΝOVX gene expression, protein levels, or downregulate ΝOVX activity, can be monitored in clinical trails of subjects exhibiting increased ΝOVX gene expression, protein levels, or upregulated ΝOVX activity. In such clinical trials, the expression or activity of ΝOVX and, preferably, other genes that have been implicated in, for example, a cellular proliferation or immune disorder can be used as a "read out" or markers of the immune responsiveness of a particular cell.
Byway of example, and not of limitation, genes, including ΝOVX, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) that modulates ΝOVX activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on cellular proliferation disorders, for example, in a climcal trial, cells can be isolated and RΝA prepared and analyzed for the levels of expression of ΝOVX and other genes implicated in the disorder. The levels of gene expression (i.e., a gene expression pattern) can be quantified by Northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of NOVX or other genes. In this manner, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during, treatment of the individual with the agent. one embodiment, the invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, protein, peptide, peptidomimetic, nucleic acid, small molecule, or other drug candidate identified by the screening assays described herein) comprising the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of expression of an NOVX protein, mRNA, or genomic DNA in the preadminisfration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the NOVX protein, mRNA, or genomic DNA in the pre-administration sample with the NOVX protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of NOVX to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased admimsfration of the agent may be desirable to decrease expression or activity of NOVX to lower levels than detected, i.e., to decrease the effectiveness of the agent.
Methods of Treatment
The invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with abenant NOVX expression or activity. The disorders include cardiomyopathy, atherosclerosis, hypertension, congenital heart defects, aortic stenosis, atrial septal defect (ASD), atrioventricular (A-V) canal defect, ductus arteriosus, pulmonary stenosis, subaortic stenosis, ventricular septal defect (VSD), valve diseases, tuberous sclerosis, scleroderma, obesity, transplantation, adrenoleukodysfrophy, congenital adrenal hypeφlasia, prostate cancer, neoplasm; adenocarcinoma, lymphoma, uterus cancer, fertility, hemophilia, hypercoagulation, idiopathic thrombocytopenic pmpura, immunodeficiencies, graft versus host disease, AIDS, bronchial asthma, Crohn's disease; multiple sclerosis, treatment of Albright Hereditary Ostoeodystrophy, and other diseases, disorders and conditions of the like.
These methods of treatment will be discussed more fully, below.
Disease and Disorders
Diseases and disorders that are characterized by increased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that antagonize (i.e., reduce or inhibit) activity. Therapeutics that antagonize activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to: (i) an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; (ii) antibodies to an aforementioned peptide; (iii) nucleic acids encoding an aforementioned peptide; (iv) adminisfration of antisense nucleic acid and nucleic acids that are "dysfunctional" (i.e., due to a heterologous insertion within the coding sequences of coding sequences to an aforementioned peptide) that are utilized to "knockout" endogenous function of an aforementioned peptide by homologous recombination (see, e.g., Capecchi, 1989. Science 244: 1288-1292); or (v) modulators ( i.e., inhibitors, agonists and antagonists, including additional peptide mimetic of the invention or antibodies specific to a peptide of the invention) that alter the interaction between an aforementioned peptide and its binding partner.
Diseases and disorders that are characterized by decreased (relative to a subject not suffering from the disease or disorder) levels or biological activity may be treated with Therapeutics that increase (i.e., axe agonists to) activity. Therapeutics that upregulate activity may be administered in a therapeutic or prophylactic manner. Therapeutics that may be utilized include, but are not limited to, an aforementioned peptide, or analogs, derivatives, fragments or homologs thereof; or an agonist that increases bioavailability.
Increased or decreased levels can be readily detected by quantifying peptide and/or RNA, by obtaining a patient tissue sample (e.g., from biopsy tissue) and assaying it in vitro for RNA or peptide levels, structure and/or activity of the expressed peptides (or mRNAs of an aforementioned peptide). Methods that are well-known within the art include, but are not limited to, immunoassays (e.g., by Western blot analysis, immunoprecipitation followed by sodium dodecyl sulfate (SDS) polyacrylamide gel electrophoresis, immunocytochemistry, etc.) and/or hybridization assays to detect expression of mRNAs (e.g., Northern assays, dot blots, in situ hybridization, and the like).
Prophylactic Methods h one aspect, the invention provides a method for preventing, in a subject, a disease or condition associated with an abeπant NOVX expression or activity, by administering to the subject an agent that modulates NOVX expression or at least one NOVX activity. Subjects at risk for a disease that is caused or contributed to by abeπant NOVX expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the NOVX abeπancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending upon the type of NOVX abeπancy, for example, an NOVX agonist or NOVX antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein. The prophylactic methods of the invention are further discussed in the following subsections. Therapeutic Methods
Another aspect of the invention pertains to methods of modulating NOVX expression or activity for therapeutic puφoses. The modulatory method of the invention involves contacting a cell with an agent that modulates one or more of the activities of NOVX protein activity associated with the cell. An agent that modulates NOVX protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring cognate ligand of an NOVX protein, a peptide, an NOVX peptidomimetic, or other small molecule. In one embodiment, the agent stimulates one or more NOVX protein activity. Examples of such stimulatory agents include active NOVX protein and a nucleic acid molecule encoding NOVX that has been introduced into the cell, hi another embodiment, the agent inhibits one or more NOVX protein activity. Examples of such inhibitory agents include antisense NOVX nucleic acid molecules and anti-NOVX antibodies. These modulatory methods can be performed in- vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the invention provides methods of treating an individual afflicted with a disease or disorder characterized by abeπant expression or activity of an NOVX protein or nucleic acid molecule, hi one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., up-regulates or down-regulates) NOVX expression or activity. In another embodiment, the method involves administering an NOVX protein or nucleic acid molecule as therapy to compensate for reduced or abeπant NOVX expression or activity.
Stimulation of NOVX activity is desirable in situations in which NOVX is abnormally downregulated and/or in which increased NOVX activity is likely to have a beneficial effect. One example of such a situation is where a subject has a disorder characterized by abeπant cell proliferation and/or differentiation (e.g., cancer or immune associated disorders). Another example of such a situation is where the subject has a gestational disease (e.g., preclampsia).
Determination of the Biological Effect of the Therapeutic
In various embodiments of the invention, suitable in vitro or in vivo assays are performed to determine the effect of a specific Therapeutic and whether its administration is indicated for treatment of the affected tissue.
In various specific embodiments, in vitro assays may be performed with representative cells of the type(s) involved in the patient's disorder, to determine if a given Therapeutic exerts the desired effect upon the cell type(s). Compounds for use in therapy may be tested in suitable animal model systems including, but not limited to rats, mice, chicken, cows, monkeys, rabbits, and the like, prior to testing in human subjects. Similarly, for in vivo testing, any of the animal model system known in the art may be used prior to admimsfration to human subjects.
Prophylactic and Therapeutic Uses of the Compositions of the Invention
The NOVX nucleic acids and proteins of the invention are useful in potential prophylactic and therapeutic applications implicated in a variety of disorders including, but not limited to: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer- associated cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias, metabolic disturbances associated with obesity, the metabolic syndrome X and wasting disorders associated with chronic diseases and various cancers.
As an example, a cDNA encoding the NOVX protein of the invention may be useful in gene therapy, and the protein may be useful when admimstered to a subject in need thereof. By way of non-limiting example, the compositions of the invention will have efficacy for treatment of patients suffering from: metabolic disorders, diabetes, obesity, infectious disease, anorexia, cancer-associated cachexia, cancer, neurodegenerative disorders, Alzheimer's Disease, Parkinson's Disorder, immune disorders, hematopoietic disorders, and the various dyslipidemias.
Both the novel nucleic acid encoding the NOVX protein, and the NOVX protein of the invention, or fragments thereof, may also be useful in diagnostic applications, wherein the presence or amount of the nucleic acid or the protein are to be assessed. A further use could be as an anti-bacterial molecule (i.e., some peptides have been found to possess anti-bacterial properties). These materials are further useful in the generation of antibodies, which immunospecifically-bind to the novel substances of the invention for use in therapeutic or diagnostic methods.
The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
Examples Example 1. Identification of NOVX clones
The novel NOVX target sequences identified in the present invention were subjected to the exon linking process to confirm the sequence. PCR primers were designed by starting at the most upstream sequence available, for the forward primer, and at the most downstream sequence available for the reverse primer. Table 12 shows the sequences of the PCR primers used for obtaining different clones, h each case, the sequence was examined, walking inward from the respective termini toward the coding sequence, until a suitable sequence that is either unique or highly selective was encountered, or, in the case of the reverse primer, until the stop codon was reached. Such primers were designed based on in silico predictions for the full length cDNA, part (one or more exons) of the DNA or protein sequence of the target sequence, or by translated homology of the predicted exons to closely related human sequences from other species. These primers were then employed in PCR amplification based on the following pool of human cDNAs: adrenal gland, bone manow, brain - amygdala, brain - cerebellum, brain - hippocampus, brain - substantia nigra, brain - thalamus, brain -whole, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, lymphoma - Raji, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small intestine, spinal cord, spleen, stomach, testis, thyroid, trachea, uterus. Usually the resulting amplicons were gel purified, cloned and sequenced to high redundancy. The PCR product derived from exon linking was cloned into the pCR2.1 vector from hivifrogen. The resulting bacterial clone has an insert covering the entire open reading frame cloned into the pCR2.1 vector. The resulting sequences from all clones were assembled with themselves, with other fragments in CuraGen Coφoration' s database and with public ESTs. Fragments and ESTs were included as components for an assembly when the extent of their identity with another component of the assembly was at least 95% over 50 bp. h addition, sequence traces were evaluated manually and edited for coπections if appropriate. These procedures provide the sequence reported herein.
Table 12A. PCR Primers for Exon Linking
Figure imgf000200_0001
Physical clone: Exons were predicted by homology and the infron/exon boundaries were determined using standard genetic rules. Exons were further selected and refined by means of similarity determination using multiple BLAST (for example, tBlastN, BlastX, and BlastN) searches, and, in some instances, GeneScan and Grail. Expressed sequences from both public and proprietary databases were also added when available to further define and complete the gene sequence. The DNA sequence was then manually conected for apparent inconsistencies thereby obtaining the sequences encoding the full-length protein.
Example 2. Quantitative expression analysis of clones in various cells and tissues
The quantitative expression of various clones was assessed using microtiter plates containing RNA samples from a variety of normal and pathology-derived cells, cell lines and tissues using real time quantitative PCR (RTQ PCR). RTQ PCR was performed on an Applied Biosystems ABI PRISM® 7700 or an ABI PRISM® 7900 HT Sequence Detection System. Various collections of samples are assembled on the plates, and refened to as Panel 1 (containing normal tissues and cancer cell lines), Panel 2 (containing samples derived from tissues from normal and cancer sources), Panel 3 (containing cancer cell lines), Panel 4 (containing cells and cell lines from normal tissues and cells related to inflammatory conditions), Panel 5D/5I (containing human tissues and cell lines with an emphasis on metabolic diseases), AI_comprehensivejpanel (containing normal tissue and samples from autoimmune diseases), Panel CNSD.01 (containing central nervous system samples from normal and diseased brains) and CNS neurodegeneration panel (containing samples from normal and Alzheimer's diseased brains).
RNA integrity from all samples is controlled for quality by visual assessment of agarose gel elecfropherograms using 28S and 18S ribosomal RNA staining intensity ratio as a guide (2:1 to 2.5:1 28s:18s) and the absence of low molecular weight RNAs that would be indicative of degradation products. Samples are controlled against genomic DNA contamination by RTQ PCR reactions run in the absence of reverse transcriptase using probe and primer sets designed to amplify across the span of a single exon.
First, the RNA samples were normalized to reference nucleic acids such as constitutively expressed genes (for example, β-actin and GAPDH). Normalized RNA (5 ul) was converted to cDNA and analyzed by RTQ-PCR using One Step RT-PCR Master Mix
Reagents (Applied Biosystems; Catalog No. 4309169) and gene-specific primers according to the manufacturer's instructions. In other cases, non-normalized RNA samples were converted to single strand cDNA (sscDNA) using Superscript II (hivifrogen Coφoration; Catalog No. 18064-147) and random hexamers according to the manufacturer's instructions. Reactions containing up to 10 μg of total RNA were performed in a volume of 20 μl and incubated for 60 minutes at 42°C. This reaction can be scaled up to 50 μg of total RNA in a final volume of 100 μl. sscDNA samples are then normalized to reference nucleic acids as described previously, using IX TaqMan® Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instructions.
Probes and primers were designed for each assay according to Applied Biosystems Primer Express Software package (version I for Apple Computer's Macintosh Power PC) or a similar algorithm using the target sequence as input. Default settings were used for reaction conditions and the following parameters were set before selecting primers: primer concentration = 250 nM, primer melting temperature (Tm) range = 58°-60°C, primer optimal Tm = 59°C, maximum primer difference = 2°C, probe does not have 5'G, probe Tm must be 10°C greater than primer Tm, amplicon size 75bp to lOObp. The probes and primers selected (see below) were synthesized by Synthegen (Houston, TX, USA). Probes were double purified by HPLC to remove uncoupled dye and evaluated by mass spectroscopy to verify coupling of reporter and quencher dyes to the 5' and 3' ends of the probe, respectively. Their final concentrations were: forward and reverse primers, 900nM each, and probe, 200nM.
PCR conditions: When working with RNA samples, normalized RNA from each tissue and each cell line was spotted in each well of either a 96 well or a 384-well PCR plate (Applied Biosystems). PCR cocktails included either a single gene specific probe and primers set, or two multiplexed probe and primers sets (a set specific for the target clone and another gene-specific set multiplexed with the target probe). PCR reactions were set up using TaqMan® One-Step RT-PCR Master Mix (Applied Biosystems, Catalog No. 4313803) following manufacturer's instructions. Reverse transcription was performed at 48°C for 30 minutes followed by amplification/PCR cycles as follows: 95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were recorded as CT values (cycle at which a given sample crosses a threshold level of fluorescence) using a log scale, with the difference in RNA concentration between a given sample and the sample with the lowest CT value being represented as 2 to the power of delta CT. The percent relative expression is then obtained by taking the reciprocal of this RNA difference and multiplying by 100. When working with sscDNA samples, normalized sscDNA was used as described previously for RNA samples. PCR reactions containing one or two sets of probe and primers were set up as described previously, using IX TaqMan® Universal Master mix (Applied Biosystems; catalog No. 4324020), following the manufacturer's instructions. PCR amplification was performed as follows: 95°C 10 min, then 40 cycles of 95°C for 15 seconds, 60°C for 1 minute. Results were analyzed and processed as described previously.
Panels 1, 1.1, 1.2, and 1.3D
The plates for Panels 1, 1.1, 1.2 and 1.3D include 2 control wells (genomic DNA confrol and chemistry control) and 94 wells containing cDNA from various samples. The samples in these panels are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer. Cell lines used in these panels are widely available through the American Type Culture Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on these panels are comprised of samples derived from all major organ systems from single adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone manow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose.
h the results for Panels 1, 1.1, 1.2 and 1.3D, the following abbreviations are used: ca. = carcinoma, * = established from metastasis, met = metastasis, s cell var = small cell variant, non-s = non-sm = non-small, squam = squamous, pi. eff = pi effusion = pleural effusion, glio = glioma, astro = asfrocytoma, and neuro = neuroblastoma.
General screening jpanel vl .4
The plates for Panel 1.4 include 2 control wells (genomic DNA control and chemistry control) and 94 wells containing cDNA from various samples. The samples in Panel 1.4 are broken into 2 classes: samples derived from cultured cell lines and samples derived from primary normal tissues. The cell lines are derived from cancers of the following types: lung cancer, breast cancer, melanoma, colon cancer, prostate cancer, CNS cancer, squamous cell carcinoma, ovarian cancer, liver cancer, renal cancer, gastric cancer and pancreatic cancer. Cell lines used in Panel 1.4 are widely available through the American Type Culture Collection (ATCC), a repository for cultured cell lines, and were cultured using the conditions recommended by the ATCC. The normal tissues found on Panel 1.4 are comprised of pools of samples derived from all major organ systems from 2 to 5 different adult individuals or fetuses. These samples are derived from the following organs: adult skeletal muscle, fetal skeletal muscle, adult heart, fetal heart, adult kidney, fetal kidney, adult liver, fetal liver, adult lung, fetal lung, various regions of the brain, the spleen, bone manow, lymph node, pancreas, salivary gland, pituitary gland, adrenal gland, spinal cord, thymus, stomach, small intestine, colon, bladder, trachea, breast, ovary, uterus, placenta, prostate, testis and adipose. Abbreviations are as described for Panels 1, 1.1, 1.2, and 1.3D.
Panels 2D and 2.2
The plates for Panels 2D and 2.2 generally include 2 confrol wells and 94 test samples composed of RNA or cDNA isolated from human tissue procured by surgeons working in close cooperation with the National Cancer Institute's Cooperative Human Tissue Network (CHTN) or the National Disease Research Initiative (NDRI). The tissues are derived from human malignancies and in cases where indicated many malignant tissues have "matched margins" obtained from noncancerous tissue just adjacent to the tumor. These are termed normal adjacent tissues and are denoted "NAT" in the results below. The tumor tissue and the "matched margins" are evaluated by two independent pathologists (the surgical pathologists and again by a pathologist at NDRI or CHTN). This analysis provides a gross histopathological assessment of tumor differentiation grade. Moreover, most samples include the original surgical pathology report that provides information regarding the clinical stage of the patient. These matched margins are taken from the tissue sunounding (i.e. immediately proximal) to the zone of surgery (designated "NAT", for normal adjacent tissue, in Table RR). In addition, RNA and cDNA samples were obtained from various human tissues derived from autopsies performed on elderly people or sudden death victims (accidents, etc.). These tissues were ascertained to be free of disease and were purchased from various commercial sources such as Clontech (Palo Alto, CA), Research Genetics, and hivifrogen.
Panel 3D
The plates of Panel 3D are comprised of 94 cDNA samples and two control samples. Specifically, 92 of these samples are derived from cultured human cancer cell lines, 2 samples of human primary cerebellar tissue and 2 controls. The human cell lines are generally obtained from ATCC (American Type Culture Collection), NCI or the German tumor cell bank and fall into the following tissue groups: Squamous cell carcinoma of the tongue, breast cancer, prostate cancer, melanoma, epidermoid carcinoma, sarcomas, bladder carcinomas, pancreatic cancers, kidney cancers, leukemias/lymphomas, ovarian/uterine/cervical, gastric, colon, lung and CNS cancer cell lines. In addition, there are two independent samples of cerebellum. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. The cell lines in panel 3D and 1.3D are of the most common cell lines used in the scientific literature.
Panels 4D, 4R, and 4.1D
Panel 4 includes samples on a 96 well plate (2 control wells, 94 test samples) composed of RNA (Panel 4R) or cDNA (Panels 4D/4.1D) isolated from various human cell lines or tissues related to inflammatory conditions. Total RNA from control normal tissues such as colon and lung (Stratagene, La Jolla, CA) and thymus and kidney (Clontech) was employed. Total RNA from liver tissue from cinhosis patients and kidney from lupus patients was obtained from BioChain (Biochain Institute, e, Hayward, CA). Intestinal tissue for RNA preparation from patients diagnosed as having Crohn's disease and ulcerative colitis was obtained from the National Disease Research Interchange (NDRI) (Philadelphia, PA).
Astrocytes, lung fibroblasts, dermal fibroblasts, coronary artery smooth muscle cells, small airway epithelium, bronchial epithelium, microvascular dermal endothelial cells, microvascular lung endothelial cells, human pulmonary aortic endothelial cells, human umbilical vein endothelial cells were all purchased from Clonetics (Walkersville, MD) and grown in the media supplied for these cell types by Clonetics. These primary cell types were activated with various cytokines or combinations of cytokines for 6 and/or 12-14 hours, as indicated. The following cytokines were used; IL-1 beta at approximately l-5ng/ml, TNF alpha at approximately 5-10ng/ml, IFN gamma at approximately 20-50ng/ml, IL-4 at approximately 5-lOng/ml, IL-9 at approximately 5-lOng/ml, IL-13 at approximately 5- lOng/ml. Endothelial cells were sometimes starved for various times by culture in the basal media from Clonetics with 0.1% serum.
Mononuclear cells were prepared from blood of employees at CuraGen Coφoration, using Ficoll. LAK cells were prepared from these cells by culture in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco/Life Technologies, Rockville, MD), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco) and Ihterleukin 2 for 4-6 days. Cells were then either activated with 10-20ng/ml PMA and l-2μg/ml ionomycin, IL-12 at 5-10ng/ml, IFN gamma at 20-50ng/ml and E -18 at 5- lOng/ml for 6 hours. In some cases, mononuclear cells were cultured for 4-5 days in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco) with PHA (phytohemagglutinin) or PWM (pokeweed mitogen) at approximately 5 μg/ml. Samples were taken at 24, 48 and 72 hours for RNA preparation. MLR (mixed lymphocyte reaction) samples were obtained by taking blood from two donors, isolating the mononuclear cells using Ficoll and mixing the isolated mononuclear cells 1:1 at a final concentration of approximately 2xl06cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol (5.5xl0"5M) (Gibco), and lOmM Hepes (Gibco). The MLR was cultured and samples taken at various time points ranging from 1- 7 days for RNA preparation.
Monocytes were isolated from mononuclear cells using CD 14 Miltenyi Beads, +ve VS selection columns and a Vario Magnet according to the manufacturer's instructions. Monocytes were differentiated into dendritic cells by culture in DMEM 5% fetal calf serum (FCS) (Hyclone, Logan, UT), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco), 50ng/ml GMCSF and 5ng/ml IL-4 for 5-7 days. Macrophages were prepared by culture of monocytes for 5-7 days in DME1VI 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), lOmM Hepes (Gibco) and 10% AB Human Serum or MCSF at approximately 50ng/ml. Monocytes, macrophages and dendritic cells were stimulated for 6 and 12-14 hours with lipopolysaccharide (LPS) at 100ng/ml. Dendritic cells were also stimulated with anti-CD40 monoclonal antibody (Pharmingen) at lOμg/ml for 6 and 12-14 hours.
CD4 lymphocytes, CD8 lymphocytes and NK cells were also isolated from mononuclear cells using CD4, CD8 and CD56 Miltenyi beads, positive VS selection columns and a Vario Magnet according to the manufacturer's instructions. CD45RA and CD45RO CD4 lymphocytes were isolated by depleting mononuclear cells of CD8, CD56, CD14 and CD19 cells using CD8, CD56, CD14 and CD19 Miltenyi beads and positive selection. CD45RO beads were then used to isolate the CD45RO CD4 lymphocytes with the remaining cells being CD45RA CD4 lymphocytes. CD45RA CD4, CD45RO CD4 and CD8 lymphocytes were placed in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco) and plated at 106cells/ml onto Falcon 6 well tissue culture plates that had been coated overnight with 0.5 μg/ml anti-CD28 (Pharmingen) and 3ug/ml anti-CD3 (OKT3, ATCC) in PBS. After 6 and 24 hours, the cells were harvested for RNA preparation. To prepare chronically activated CD8 lymphocytes, we activated the isolated CD8 lymphocytes for 4 days on anti-CD28 and anti-CD3 coated plates and then harvested the cells and expanded them in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco) and IL-2. The expanded CD8 cells were then activated again with plate bound anti-CD3 and anti-CD28 for 4 days and expanded as before. RNA was isolated 6 and 24 hours after the second activation and after 4 days of the second expansion culture. The isolated NK cells were cultured in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco) and IL-2 for 4-6 days before RNA was prepared.
To obtain B cells, tonsils were procured from NDRI. The tonsil was cut up with sterile dissecting scissors and then passed through a sieve. Tonsil cells were then spun down and resupended at 106cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco). To activate the cells, we used PWM at 5μg/ml or anti-CD40 (Pharmingen) at approximately lOμg/ml and IL-4 at 5-10ng/ml. Cells were harvested for RNA preparation at 24,48 and 72 hours. To prepare the primary and secondary Thl/Th2 and Trl cells, six-well Falcon plates were coated overnight with lOμg/ml anti-CD28 (Pharmingen) and 2μg/ml OKT3 (ATCC), and then washed twice with PBS. Umbilical cord blood CD4 lymphocytes (Poietic Systems, German Town, MD) were cultured at 105-106cells/ml in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5x10" 5M (Gibco), lOmM Hepes (Gibco) and IL-2 (4ng/ml). IL-12 (5ng/ml) and anti-IL4 (1 μg/ml) were used to direct to Thl, while IL-4 (5ng/ml) and anti-IFN gamma (1 μg/ml) were used to direct to Th2 and IL-10 at 5ng/ml was used to direct to Trl. After 4-5 days, the activated Thl, Th2 and Trl lymphocytes were washed once in DMEM and expanded for 4-7 days in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), lOmM Hepes (Gibco) and IL-2 (lng/ml). Following this, the activated Thl, Th2 and Trl lymphocytes were re-stimulated for 5 days with anti-CD28/OKT3 and cytokines as described above, but with the addition of anti-CD95L (1 μg/ml) to prevent apoptosis. After 4-5 days, the Thl, Th2 and Trl lymphocytes were washed and then expanded again with IL-2 for 4-7 days. Activated Thl and Th2 lymphocytes were maintained in this way for a maximum of three cycles. RNA was prepared from primary and secondary Thl, Th2 and Trl after 6 and 24 hours following the second and third activations with plate bound anti-CD3 and anti-CD28 niAbs and 4 days into the second and third expansion cultures in hiterleukin 2.
The following leukocyte cells lines were obtained from the ATCC: Ramos, EOL-1, KU-812. EOL cells were further differentiated by culture in O.lmM dbcAMP at 5xl05cells/ml for 8 days, changing the media every 3 days and adjusting the cell concentration to 5xl05cells/ml. For the culture of these cells, we used DMEM or RPMI (as recommended by the ATCC), with the addition of 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xlO"5M (Gibco), lOmM Hepes (Gibco). RNA was either prepared from resting cells or cells activated with PMA at lOng/ml and ionomycin at 1 μg/ml for 6 and 14 hours. Keratinocyte line CCD106 and an airway epithelial tumor line NCI-H292 were also obtained from the ATCC. Both were cultured in DMEM 5% FCS (Hyclone), lOOμM non essential amino acids (Gibco), ImM sodium pyruvate (Gibco), mercaptoethanol 5.5xl0"5M (Gibco), and lOmM Hepes (Gibco). CCD1106 cells were activated for 6 and 14 hours with approximately 5 ng/ml TNF alpha and lng/ml IL-1 beta, while NCI-H292 cells were activated for 6 and 14 hours with the following cytokines: 5ng/ml IL-4, 5ng/ml IL-9, 5ng/ml IL-13 and 25ng/ml IFN gamma. For these cell lines and blood cells, RNA was prepared by lysing approximately 107cells/ml using Trizol (Gibco BRL). Briefly, 1/10 volume of bromochloropropane (Molecular Research Coφoration) was added to the RNA sample, vortexed and after 10 minutes at room temperature, the tubes were spun at 14,000 φm in a Sorvall SS34 rotor. The aqueous phase was removed and placed in a 15ml Falcon Tube. An equal volume of isopropanol was added and left at -20°C overnight. The precipitated RNA was spun down at 9,000 φm for 15 min in a Sorvall SS34 rotor and washed in 70% ethanol. The pellet was redissolved in 300μl of RNAse-free water and 35μl buffer (Promega) 5μl DTT, 7μl RNAsin and 8μl DNAse were added. The tube was incubated at 37°C for 30 minutes to remove contaminating genomic DNA, extracted once with phenol chloroform and re-precipitated with 1/10 volume of 3M sodium acetate and 2 volumes of 100% ethanol. The RNA was spun down and placed in RNAse free water. RNA was stored at -80°C.
Autoimmunity (AI)_comprehensive panel vl.O
The plates for AI_comprehensive panel_vl.O include two confrol wells and 89 test samples comprised of cDNA isolated from surgical and postmortem human tissues obtained from the Backus Hospital and Clinomics (Frederick, MD). Total RNA was extracted from tissue samples from the Backus Hospital in the Facility at CuraGen. Total RNA from other tissues was obtained from Clinomics.
Joint tissues including synovial fluid, synovium, bone and cartilage were obtained from patients undergoing total knee or hip replacement surgery at the Backus Hospital. Tissue samples were immediately snap frozen in liquid nitrogen to ensure that isolated RNA was of optimal quality and not degraded. Additional samples of osteoarthritis and rheumatoid arthritis joint tissues were obtained from Clinomics. Normal control tissues were supplied by Clinomics and were obtained during autopsy of trauma victims.
Surgical specimens of psoriatic tissues and adjacent matched tissues were provided as total RNA by Clinomics. Two male and two female patients were selected between the ages of 25 and 47. None of the patients were taking prescription drugs at the time samples were isolated.
Surgical specimens of diseased colon from patients with ulcerative colitis and Crohns disease and adjacent matched tissues were obtained from Clinomics. Bowel tissue from three female and three male Crohn's patients between the ages of 41-69 were used. Two patients were not on prescription medication while the others were taking dexamethasone, phenobarbital, or tylenol. Ulcerative colitis tissue was from three male and four female patients. Four of the patients were taking lebvid and two were on phenobarbital.
Total RNA from post mortem lung tissue from trauma victims with no disease or with emphysema, asthma or COPD was purchased from Clinomics. Emphysema patients ranged in age from 40-70 and all were smokers, this age range was chosen to focus on patients with cigarette-linked emphysema and to avoid those patients with alpha- lanti-trypsin deficiencies. Asthma patients ranged in age from 36-75, and excluded smokers to prevent those patients that could also have COPD. COPD patients ranged in age from 35-80 and included both smokers and non-smokers. Most patients were taking corticosteroids, and bronchodilators.
In the labels employed to identify tissues in the AI_comprehensive panel_vl.0 panel, the following abbreviations are used:
Al = Autoimmunity Syn = Synovial Normal = No apparent disease Rep22 /Rep20 = individual patients RA = Rheumatoid arthritis Backus = From Backus Hospital OA = Osteoarthritis (SS) (BA) (MF) = Individual patients Adj = Adjacent tissue Match control = adjacent tissues -M = Male -F = Female COPD = Chronic obstructive pulmonary disease
Panels 5D and 51
The plates for Panel 5D and 51 include two control wells and a variety of cDNAs isolated from human tissues and cell lines with an emphasis on metabolic diseases. Metabolic tissues were obtained from patients enrolled in the Gestational Diabetes study. Cells were obtained during different stages in the differentiation of adipocytes from human mesenchymal stem cells. Human pancreatic islets were also obtained. In the Gestational Diabetes study subjects are young (18 - 40 years), otherwise healthy women with and without gestational diabetes undergoing routine (elective) Caesarean section. After delivery of the infant, when the surgical incisions were being repaired/closed, the obstetrician removed a small sample .
Patient 2: Diabetic Hispanic, overweight, not on insulin Patient 7-9: Nondiabetic Caucasian and obese (BMI>30) Patient 10: Diabetic Hispanic, overweight, on insulin Patient 11 : Nondiabetic African American and overweight Patient 12: Diabetic Hispanic on insulin
Adipocyte differentiation was induced in donor progenitor cells obtained from Osirus (a division of Clonetics/BioWhittaker) in triplicate, except for Donor 3U which had only two replicates. Scientists at Clonetics isolated, grew and differentiated human mesenchymal stem cells (HuMSCs) for CuraGen based on the published protocol found in Mark F. Pittenger, et al., Multilineage Potential of Adult Human Mesenchymal Stem Cells Science Apr 2 1999: 143-147. Clonetics provided Trizol lysates or frozen pellets suitable for mRNA isolation and ds cDNA.production. A general description of each donor is as follows:
Donor 2 and 3 U: Mesenchymal Stem cells, Undifferentiated Adipose Donor 2 and 3 AM: Adipose, AdiposeMidway Differentiated Donor 2 and 3 AD: Adipose, Adipose Differentiated
Human cell lines were generally obtained from ATCC (American Type Culture Collection), NCI or the German tumor cell bank and fall into the following tissue groups: kidney proximal convoluted tubule, uterine smooth muscle cells, small intestine, liver HepG2 cancer cells, heart primary stromal cells, and adrenal cortical adenoma cells. These cells are all cultured under standard recommended conditions and RNA extracted using the standard procedures. All samples were processed at CuraGen to produce single stranded cDNA.
Panel 51 contains all samples previously described with the addition of pancreatic islets from a 58 year old female patient obtained from the Diabetes Research Institute at the University of Miami School of Medicine. Islet tissue was processed to total RNA at an outside source and delivered to CuraGen for addition to panel 51.
In the labels employed to identify tissues in the 5D and 51 panels, the following abbreviations are used: GO Adipose = Greater Omenrum Adipose SK = Skeletal Muscle UT = Uterus PL = Placenta
AD = Adipose Differentiated AM = Adipose Midway Differentiated U = Undifferentiated Stem Cells
Central Nervous System Panel CNSD.01
The plates for Panel CNSD.01 include two control wells and 94 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center. Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains two brains from each of the following diagnoses: Alzheimer's disease, Parkinson's disease, Huntington's disease, Progressive Supernuclear Palsy, Depression, and "Normal controls". Within each of these brains, the following regions are represented: cingulate gyrus, temporal pole, globus palladus, substantia nigra, Brodman Area 4 (primary motor strip), Brodman Area 7 (parietal cortex), Brodman Area 9 (prefrontal cortex), and Brodman area 17 (occipital cortex). Not all brain regions are represented in all cases; e.g., Huntington's disease is characterized in part by neurodegeneration in the globus palladus, thus this region is impossible to obtain from confirmed Huntington's cases. Likewise Parkinson's disease is characterized by degeneration of the substantia nigra making this region more difficult to obtain. Normal control brains were examined for neuropathology and found to be free of any pathology consistent with neurodegeneration.
In the labels employed to identify tissues in the CNS panel, the following abbreviations are used:
PSP = Progressive supranuclear palsy Sub Nigra = Substantia nigra Glob Palladus= Globus palladus Temp Pole = Temporal pole Cing Gyr = Cingulate gyrus BA 4 = Brodman Area 4
Panel CNS Neurodegeneration Vl.O
The plates for Panel CNS_Neurodegeneration_V1.0 include two confrol wells and 47 test samples comprised of cDNA isolated from postmortem human brain tissue obtained from the Harvard Brain Tissue Resource Center (McLean Hospital) and the Human Brain and Spinal Fluid Resource Center (VA Greater Los Angeles Healthcare System). Brains are removed from calvaria of donors between 4 and 24 hours after death, sectioned by neuroanatomists, and frozen at -80°C in liquid nitrogen vapor. All brains are sectioned and examined by neuropathologists to confirm diagnoses with clear associated neuropathology.
Disease diagnoses are taken from patient records. The panel contains six brains from Alzheimer's disease (AD) patients, and eight brains from "Normal controls" who showed no evidence of dementia prior to death. The eight normal control brains are divided into two categories: Controls with no dementia and no Alzheimer's like pathology (Controls) and controls with no dementia but evidence of severe Alzheimer's like pathology, (specifically senile plaque load rated as level 3 on a scale of 0-3; 0 = no evidence of plaques, 3 = severe AD senile plaque load). Within each of these brains, the following regions are represented: hippocampus, temporal cortex (Brodman Area 21), parietal cortex (Brodman area 7), and occipital cortex (Brodman area 17). These regions were chosen to encompass all levels of neurodegeneration in AD. The hippocampus is a region of early and severe neuronal loss in AD; the temporal cortex is known to show neurodegeneration in AD after the hippocampus; the parietal cortex shows moderate neuronal death in the late stages of the disease; the occipital cortex is spared in AD and therefore acts as a "control" region within AD patients. Not all brain regions are represented in all cases.
hi the labels employed to identify tissues in the CNS_Neurodegeneration_V1.0 panel, the following abbreviations are used:
AD = Alzheimer's disease brain; patient was demented and showed AD-like pathology upon autopsy
Control = Control brains; patient not demented, showing no neuropathology Confrol (Path) = Control brains; pateint not demented but showing sever AD-like pathology SupTemporal Ctx = Superior Temporal Cortex frif Temporal Ctx = Inferior Temporal Cortex
A. CG55758-01: EGF-Related Protein (SCUBEl)-like protein
Expression of gene CG55758-01 was assessed using the primer-probe set Ag2442, described in Table 12AA. Results of the RTQ-PCR runs are shown in Tables 12AB, 12 AC, AD, and AE.
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Table 12AD. Panel 3D
Figure imgf000217_0002
Figure imgf000218_0001
Table 12AE . Panel 4 . ID
Figure imgf000219_0001
Figure imgf000220_0001
CNS_neurodegeneration_ l.O Summary: Ag2442 Expression of CG55758-01 is low/undetectable in all samples in this panel (CT>35). (Data not shown.)
Panel 1.3D Summary: Ag2442 Two experiments with the same probe and primer set produce results that are in excellent agreement, with both experiments showing highest expression of the CG55758-01 gene in an astrocytoma cell line (CTs=29-30). It is also expressed at lower levels in two lung cancer cell lines and a renal cancer cell line. There is also low level expression in a number of normal tissues including testis, ovary, mammalian gland, lung, trachea, kidney, spleen and brain. The increased expression seen in the astrocytoma cell line suggests that this gene may play a role in the cancers used in the derivation of this cell line. Thus, therapeutic inhibition of the function of this gene product, through the use of antibodies or small molecule drugs, might be of utility in the treatment of this disease.
Both runs show highest expression of this SCUBEl-like gene among metabolically relevant tissues in the small intestine. Lower levels of expression are also seen in the adrenal gland, liver, and stomach. This expression profile suggests that this gene and its product may be involved in the development of these organs and their interaction with the extracellular environment. Therefore, antibody or protein therapeutics targeted towards this gene product maybe effective therapeutics against diseases and conditions involving these organs.
This gene is a novel SCUBEl-like protein that is expresed in the developing brain. This gene or its protein product may therefore be of use in the treatment of developmental disorders such as autism, schizophrenia, attention deficit disorder, and Tourette syndrome. Panel 2D Summary: Ag2442 The CG55758-01 gene is highly expressed in a normal ovary sample (CT=29.1). The level of expression in some lung, prostate, ovary and kidney normal samples appears to be increased when compared to the matched tumor tissue. The reverse appears to be true for liver, where expression is slightly higher in the tumor tissue than the matched normal tissues. Thus, based upon its profile, the expression of this gene could be of use as a marker for distinguishing some cancers from the normal adjacent tissue or as a marker for different grades/ types of cancer. Therapeutic use of this gene, through the use of peptides, polypeptides or small molecule drugs, might be of utility in the treatment of lung, prostate, ovary and kidney cancer; while inhibition of its activity might be used for treatment of liver cancer.
Panel 3D Summary: Ag2442 The CG55758-01 gene is expressed in select cancer cell lines in this panel. The highest level of expression is in a cell line derived from Wilm's tumor , G401 (CT=30.3). A high level of expression is also seen in rhabdomyosarcoma and lung and brain cancer cell lines. Thus, therapeutic inhibition of the function of this gene, through the use of antibodies or small molecule drags, might be of utility in the treatment of cancers from which these cell lines were derived.
Panel 4.1D Summary: Ag2442 The CG55758-01 transcript is expressed at low level in kidney and colon in this panel (CTs=32-35). The putative EGF-related protein encoded by this transcript may play an important role in the normal development and homeostasis of these tissues. Modulation of the expression or function of the protein encoded by this transcript could be important for maintaining or restoring normal function to these organs during inflammation.
Panel 4D Summary: Ag2442 Data from one experiment with this probe and primer set is not included because the amp plot suggests that there was a problem with one of the sample wells.
B. CG55724-01: Adipocyte Complement Related Protein
Expression of gene CG55724-01 was assessed using the primer-probe set Ag3094, described in Table 12BA. Results of the RTQ-PCR runs are shown in Table BB.
Table 12BA. Probe Name Ag3094
Figure imgf000221_0001
Table 12BB. Panel 1.3D
Figure imgf000222_0001
Figure imgf000223_0001
CNS_neurodegeneration_vl.O Summary: Ag3094 Expression of the CG55724-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
Panel 1.3D Summary: Ag3094 The CG55724-01 gene is a novel adipocyte complement-related protein which is expresed in the developing brain. This gene or its protein product may therefore be of use in the treatment of developmental disorders such as autism, schizophrenia, attention deficit disorder, or Tourette syndrome.
Panel 2.2 Summary: Ag3094 Expression of the CG55724-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
Panel 4D Summary: Ag3094 Expression of the CG55724-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
C. CG50345-01 : Beta- Adrenergic receptor kinase
Expression of gene CG50345-01 was assessed using the primer-probe set Ag2303, described in Tablel2CA. Results of the RTQ-PCR runs are shown in Tables CB, and CC.
Table 12CA. Probe Name Ag2303
Figure imgf000223_0002
Table 12CB. Panel 1.3D
Figure imgf000223_0003
Figure imgf000224_0001
Figure imgf000225_0001
Table 12CC. Panel 4D
Figure imgf000225_0002
Figure imgf000226_0001
Panel 1.3D Summary: The CG50345-01 gene is widely expressed across the panel, with highest expression in a colon cancer cell line SW620 (CT=26.4). Of note is the difference in expression between the related colon cancer cell lines SW620 and SW480. SW480 represents the primary lesion from a patient with colon cancer, while SW620 represents a metastasis from the same patient. The difference in expression of this gene between the SW620 and SW480 cell lines indicates that it could be used to distinguish these cells, or others like them. Moreover, therapeutic modulation of the CG50345-01 gene, through the use of small molecule drugs, antibodies or protein therapeutics, may be of effective in the treatment of metastatic colon cancer.
Among tissues with metabolic function, the CG50345-01 gene is moderately expressed in the pancreas, adrenal, thyroid, pituitary, adipose, adult and fetal heart, adult and fetal liver, and adult and fetal liver. This expression profile suggests that the CG50345-01 gene product may be an important small molecule target for the treatment of metabolic disease in any or all of these tissues, including obesity and diabetes.
The CG50345-01 gene, which encodes a beta-adrenergic receptor kinase, also shows high expression in all regions of the brain examined, especially in the cerebral cortex (CT=26.7) The beta adrenergic receptors have been shown to play a role in memory formation and in clinical depression. Since many cuπent anti-depressants produce undesired side effects as a result of non-specific binding (to other receptors), this gene is therefore an excellent small molecule target for the treatment of clinical depression without side effects. Furthermore, the role of beta adrenergic receptors in memory consolidation suggests that the CG50345-01 gene product would also be useful as a small molecule target for the treatment of Alzheimer's disease, vascular dementia, or any memory loss disorder.
References:
Feighner JP. Mechanism of action of antidepressant medications. J Clin Psychiatry 1999;60 Suppl 4:4-11; discussion 12-3
The psychopharmacology of depression is a field that has evolved rapidly in just under 5 decades. Early antidepressant medications—tricyclic antidepressants (TCAs) and monoamine oxidase inhibitors (MAOIs)--were discovered through astute clinical observations. These first- generation medications were effective because they enhanced serotonergic or noradrenergic mechanisms or both. Unfortunately, the TCAs also blocked histaminic, cholinergic, and alphal -adrenergic receptor sites, and this action brought about unwanted side effects such as weight gain, dry mouth, constipation, drowsiness, and dizziness. MAOIs can interact with tyramine to cause potentially lethal hypertension and present potentially dangerous interactions with a number of medications and over-the-counter drags. The newest generation of antidepressants, including the single-receptor selective serotonin reuptake inhibitors (SSRIs) and multiple-receptor antidepressants venlafaxine, mirtazapine, bupropion, frazodone, and nefazodone, target one or more specific brain receptor sites without, in most cases, activating unwanted sites such as histamine and acetylcholine. This paper discusses the new antidepressants, particularly with regard to mechanism of action, and looks at future developments in the treatment of depression.
Feny B, McGaugh JL. Role of amygdala norepinephrine in mediating stress hormone regulation of memory storage. Acta Pharmacol Sin 2000 Jun;21(6):481-93
There is extensive evidence indicating that the noradrenergic system of the amygdala, particularly the basolateral nucleus of the amygdala (BLA), is involved in memory consolidation. This article reviews the central hypothesis that stress hormones released during emotionally arousing experiences activate noradrenergic mechanisms in the BLA, resulting in enhanced memory for those events. Findings from experiments using rats have shown that the memory-modulatory effects of the adrenocortical stress hormones epinephrine and glucocorticoids involve activation of beta-adrenoceptors in the BLA. In addition, both behavioral and microdialysis studies have shown that the noradrenergic system of the BLA also mediates the influences of other neuromodulatory systems such as opioid peptidergic and GABAergic systems on memory storage. Other findings indicate that this stress hormone- induced activation of noradrenergic mechanisms in the BLA regulates memory storage in other brain regions.
Panel 2.2 Summary: Ag2303 Data from Panel 2.2 has not been included because a strange amp plot suggests that there were problems with this experiment.
Panel 4D Summary: The CG50345-01 gene, a beta-adrenergic receptor kinase homolog, is highly expressed (CTs=26-29) in a wide range of cells of significance in the immune response in health and disease. Highest expression of this gene is found in activated B and T cells. Therefore, inhibition of the function of the protein encoded by the CG50345-01 gene with a small molecule drug may block the functions of B cells or T cells and could be beneficial in the treatment of patients suffering from autoimmune and inflammatory diseases such as asthma, allergies, inflammatory bowel disease, lupus erythematosus, or rheumatoid arthritis.
D. CG50301-01: humanTENM4
Expression of gene CG50301-01 was assessed using the primer-probe sets Ag2581 and Ag2910, described in Tables DA and DB. Results of the RTQ-PCR runs are shown in Tables 12DC, 12DD, 12DE, 12DF, and 12DG.
Table 12DA. Probe Name Ag2581
Figure imgf000228_0001
Table 19DB. Probe Name Ag2910
Figure imgf000228_0002
Table 12DC. CNS neurodegeneration vl.O
Figure imgf000228_0003
Figure imgf000229_0001
Table 12DD. Panel 1.3D
Figure imgf000229_0002
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Table 12DF. Panel 3D
Figure imgf000233_0002
Figure imgf000234_0001
Table 12DG. Panel 4D
Figure imgf000234_0002
Figure imgf000235_0001
Figure imgf000236_0001
CNS_neurodegeneration_vl.O Summary: Ag2910/Ag2581 No difference is detected in the expression of the CG50301-01 gene in the postmortem brains of Alzheimer's patients when compared normal controls. However, this panel demonstrates the expression of this gene in the CNS of an independent group of patients. See panel 1.3d for a discussion of utility of this gene in the central nervous system.
Panel 1.3D Summary: Ag2581/Ag2910 Two experiments with the same probe and primer set produce results with very good agreement. Highest expression of the CG50301-01 gene is seen in the ovary and the cerebral cortex (CTs=28). In contrast to the expression in normal ovary, ovarian cancer cell lines either do not express this gene or express it at very low levels. This expression profile suggests that expression of this gene could potentially be used as a marker for ovarian cancer. Conversely, this gene appears to be more highly expressed in prostate cancer cell lines than in the normal prostate, suggesting this gene may also be a diagnostic marker in prostate cancer as well.
This gene is a homolog of the Drosophila TENM4 gene, and is expressed at moderate levels in all brain regions examined. TENM4 is believed to be important in neural development; therefore, this gene may be of use in the induction of compensatory synaptogenesis in the treatment of any diseases/conditions involving neuronal death
(Alzheimer's, Parkinson's, Huntington's diseases, stroke, head or spinal cord trauma). Among metabolic tissues, expression is highest in fetal skeletal muscle. Furthermore, this gene is more highly expressed in fetal skeletal muscle (CTs=29) than in adult skeletal muscle (CT=35). Thus, expression of this gene could be used to differentiate between adult and fetal skeletal muscle. In addition, the higher levels of expression in fetal skeletal muscle suggest that this gene product may play a role in the development of this organ. Therefore, the protem encoded by this gene may be effective in treating weak or dystrophic muscle in the adult. There is also low but significant expression in pituitary, thyroid and adipose. Thus, this gene may be involved in the development and signal transduction pathways of these tissues. Antibody and peptide therapeutics to this gene product may be used in the treatment of metabolic disorders involving these tissues, including obesity and diabetes.
Panel 2D Summary: Ag2581/Ag2910 Two experiments with the same probe and primer set show reasonable concordance, with both runs showing highest expression of the CG50301-01 gene in ovarian cancer. The level of expression of this gene appears to be increased in some lung and gastric cancer tissue samples when compared to the matched normal tissue. The reverse appears to be true for kidney, where expression is slightly higher in 6 of 9 normal tissues than in the matched cancer tissues. Thus, based upon its profile, the expression of this gene could be of use as a marker for distinguishing these cancers from the normal adjacent tissue or as a marker for different grades/ types of cancer. Furthermore, therapeutic inhibition of the activity of the product of this gene, through the use of antibodies, peptides or polypeptides may be useful in the treatment of gastric and lung cancer.
Panel 3D Summary: Ag2581 The CG50301-01 gene is expressed at a low level by select cell lines used in this panel. The highest level of expression is seen in NCI-H526, a lung cancer cell line (CT=27.3). Other cell lines that express this gene include neuroblastoma, bladder carcinoma and renal cell cancer cell lines. Therefore, therapeutic inhibition of the activity of the product of this gene, through the use of antibodies, peptides or polypeptides may be useful in the therapy of cancers used in the derivation of these cell lines.
Panel 4D Summary: Ag2910 The CG50301-01 transcript is moderately expressed in lung fibroblasts and is slightly overexpressed in these cells after treatment with IFNg or IL-4 (CT 27.8).
This transcript encodes a human homolog of Ten-M4, a protein with EGF-repeats (reference) that may play a role in fibroblast growth. Modulation of the expression or activity of the protein encoded by this transcrpt through the application of antibodies or small molecules may be useful for treatment of symptoms associated with fibroplasia, chronic obstructive pulmonary disease, emphysema, asthma, psoriasis and ulcerative colitis. Please note that a second experiment with probe and primer set Ag2582 is not included. The amp plot indicates that there were experimental difficulties with this run.
Reference:
Mieda M, Kikuchi Y, Hirate Y, Aoki M, Okamoto H. Compartmentalized expression of zebrafish ten-m3 and ten-m4, homologues of the Drosophila ten(m)/odd Oz gene, in the central nervous system. Mech Dev 1999 Sep;87(l-2):223-7
Zebrafish ten-m3 and ten-m.4 encode proteins highly similar to the product of Drosophila pair-rule gene ten(m)/odd Oz (odz). Their products contain eight epidermal growth factor (EGF)-like repeats that resemble mostly those of the extracellular matrix molecule tenascin. During segmentation period, ten-m3 is expressed in the somites, notochord, pharyngeal arches, and the brain, while expression of ten-m4 is mainly restricted to the brain, hi the developing brain, ten-m3 and ten-m4 expression delineates several compartments. Interestingly, ten-m3 and ten-m4 show expression patterns complementary to each other in the developing forebrain and midbrain along both rostrocaudal and dorsoventral axes, depending on developmental stages and locations
Panel CNS_1 Summary: Ag2582/Ag2910 Two experiments with the same probe and primer set further confirm expression of the CG50301-01 gene in the brain. Please see Panel 1.3D for discussion of potential utility in the central nervous system.
E. CG55764-01 and CG55764-02: Out-At-First-like
Expression of gene CG55764-01 and variant CG55764-02 was assessed using the primer-probe set Ag3207, described in Table EA. Results of the RTQ-PCR runs are shown in Tables 12EB, 12EC, 12ED, 12EE and 12EF.
Table 12EA. Probe Name Ag3207
Figure imgf000238_0001
Table 12EB . CNS_neurodegeneration_vl .0
Figure imgf000238_0002
Figure imgf000239_0001
Table 12EC. Panel 1.3D
Figure imgf000239_0002
Figure imgf000240_0001
Figure imgf000241_0001
Figure imgf000242_0001
Table 12EE. Panel CNS 1
Figure imgf000242_0002
Figure imgf000243_0001
Table 12EF. Panel CNS 1.1
Figure imgf000243_0002
Figure imgf000244_0001
CNS neurodegeneration vl.O Summary: Ag3207 No difference is detected in the expression of the CG55764-01 gene in the postmortem brains of Alzheimer's patients when compared normal controls. However, this panel demonstrates the expression of this gene in the CNS of an independent group of patients. See panel 1.3d for a discussion of utility of this gene in the central nervous system.
Panel 1.3D Summary: Ag3207 Highest expression of the CG55764-01 gene is seen in the liver (CT=28.5). Other metabolic tissues that express this gene at more moderate levels include fetal skeletal muscle, fetal kidney, fetal liver and adipose. Low but significant levels of expression are also seen in the heart, kidney, fetal heart, pancreas, adrenal, salivary gland, small intestine, skeletal muscle, pituitary and stomach. The widespread expression of this gene among tissues with metabolic function suggests that antibody or peptide therapeutics to this gene product may be useful in metabolic disorders involving these tissues, including obesity and diabetes. In addition, this gene may be used to differentiate between the fetal (CT=29.7) and adult(CT=32.7) sources of skeletal muscle. Furthermore, the higher levels of expression in fetal skeletal muscle, when compared to expression in the adult suggest that the protein encoded by this gene may be involved in the development of this organ. Thus, therapeutic modulation of the activity or function of this gene product may restore muscle mass or function to weak or dystrophic muscle.
This gene is a homolog of the Drosophila Out-At-First protein and is expressed at moderate levels in all brain regions examined, except for the cerebellum where it is not expressed. This protein is believed to be involved in neural development, and may therefore be of use in the treatment of developmental disorders such as autism, schizophrenia, attention deficit disorder, or Tourette syndrome.
Overall, this gene is expressed at moderate levels in almost all cell types on this panel. The ubiquitous expression of this gene suggests that is required for growth and proliferation of cells.
Panel 4D Summary: Ag 3207 The CG55764-01 transcript is found at moderate levels in dermal fibroblasts, small aiway epithelium and lung fibroblasts. The expression of this transcript appears to be up-regulated in these cell types by the inflammatory cytokines TNF-a, IL-lb and IFN-g. This gene is also expressed in KU-812, a basophil cell line. Basophils play an important role in atopic and inflammatory diseases such as asthma, Crohn's disease, and ulcerative colitis. Therefore, the modulation of the expression or activity of the protein encoded by this transcript through the application of antibody or peptide therapeutics may be useful for the treatment of lung inflammatory diseases such as asthma, and chronic obstructive pulmonary diseases, for inflammatory skin diseases such as psoriasis, atopic dermatitis and ulcerative dermatitis, inflammatory bowel diseases and osteoarthritis.
Panel CNS_1 Summary: Ag3207 This experiment further confirms expression of the CG55764-01 gene in the brain. Please see Panel 1.3D for discussion of potential utility of this gene in the central nervous system.
Panel CNS_1.1 Summary: Ag3207 This experiment further confirms expression of the CG55764-01 gene in the brain. Please see Panel 1.3D for discussion of potential utility of this gene in the central nervous system. F. CG55704-01: EPHRIN TYPE-A RECEPTOR 6 PRECURSOR
Expression of gene CG55704-01 was assessed using the primer-probe sets Ag4155, Ag568, Agl486, Ag2879 and Agl302, described in Tables 12FA, 12FB, 12FC, 12FD and 12FE. Results of the RTQ-PCR runs are shown in Tables 12FF, 12FG, 12FH, 12FI, 12FJ, and 12FK.
Table 12FA. Probe Name Ag 155
Figure imgf000246_0001
Table 12FB. Probe Name Ag568
Figure imgf000246_0002
Table 12FC. Probe Name Agl486
Figure imgf000246_0003
Table 12FD. Probe Name Ag2879
Figure imgf000246_0004
Table 12FE. Probe Name Agl302
Figure imgf000246_0005
Table 12FF. CNS_neurodegeneration_vl .0
Figure imgf000246_0006
Figure imgf000247_0001
Table 12FG. General_screening _j?anel_vl .4
Figure imgf000247_0002
Figure imgf000248_0001
Table 12FH. Panel 1.1
Figure imgf000248_0002
Figure imgf000249_0001
Figure imgf000250_0001
Table 12FI. Panel 2.2
Figure imgf000250_0002
Figure imgf000251_0001
Table 12FJ. Panel 4. ID
Figure imgf000251_0002
Figure imgf000252_0001
Figure imgf000253_0001
Table 12FK. Panel 4D
Figure imgf000253_0002
Figure imgf000254_0001
Figure imgf000255_0001
CNS_neurodegeneration_yl.O Summary: Ag4155 The CG55704-01 gene encodes a putative ephrin receptor, and shows a significant downregulation in the AD temporal cortex compared to nondemented controls when CT values are analyzed by ANCONA. The temporal cortex (Brodmann area 21) shows severe neurodegeneration in Alzheimer's disease, though not as early as the hippocampus or entorhinal cortex. It is therefore likely that this gene is downregulated during the process of neurodegeneration, rather than the downregulation being a result of neuron loss. The ephrin receptors have been implicated in axonal and synapse guidance. Furthermore, individuals with Alzheimer's disease (especially late-onset AD with apoE4 genotype) show impaired compensatory synaptogenesis and dendritic arborization. Therefore, this gene is an excellent small molecule target for the treatment of Alzheimer's disease. Please note that a second experiment with the probe and primer set Ag2879 is not included because the amp plot suggests that there were experimental difficulties with this run.
References:
Lai KO, Ip FC, Cheung J, Fu AK, Ip ΝY. Expression of Eph receptors in skeletal muscle and their localization at the neuromuscular junction. Mol Cell Neurosci 2001 Jun;17(6):1034-47
The participation of ephrins and Eph receptors in guiding motor axons during muscle innervation has been well documented, but little is known about their expression and functional significance in muscle at later developmental stages. Our present study investigates the expression and localization of Eph receptors and ephrins in skeletal muscle. Prominent expression of EphA4, EphA7, and ephrin- A ligands was detected in muscle during embryonic development. More importantly, both EphA4 and EphA7, as well as ephrin- A2, were localized at the neuromuscular junction (NMJ) of adult muscle. Despite their relative abundance, they were not localized at the synapses during embryonic stages. The concentration of EphA4, EphA7, and ephrin- A2 at the NMJ was observed at postnatal stages and the synaptic localization became prominent at later developmental stages. In addition, expression of Eph receptors was increased by neuregulin and after nerve injury. Furthermore, we demonstrated that overexpression of EphA4 led to tyrosine phosphorylation of the actin-binding protein cortactin and that EphA4 was coimmunoprecipitated with cortactin in muscle. Taken together, our findings indicate that EphA4 is associated with the actin cytoskeleton. Since actin cytoskeleton is critical to the formation and stability of NMJ, the present findings raise the intriguing possibility that Eph receptors may have a novel role in NMJ formation and/or maintenance.
Arendt T, Schindler C, Bruckner MK, Eschrich K, Bigl V, Zedlick D, Marcova L. Plastic neuronal remodeling is impaired in patients with Alzheimer's disease carrying apolipoprotein epsilon 4 allele. J Neurosci 1997 Jan 15;17(2):516-29
A relationship between the apolipoprotein E (apoE) genotype and the risk to develop Alzheimer's disease has been established recently. Apolipoprotein synthesis is implicated in developmental processes and in neuronal repair of the adult nervous system. In the present study, we investigated the influence of the apolipoprotein polymorphism on the severity of neuronal degeneration and the extent of plastic dendritic remodeling in Alzheimer's disease. Changes in length and arborization of dendrites of Golgi-impregnated neurons in the basal nucleus of Meynert, locus coeruleus, raphe magnus nucleus, medial amygdaloid nucleus, pedunculopontine tegmental nucleus, and substantia nigra were analyzed after three- dimensional reconstruction. Patients with either one or two apoE epsilon 4 alleles not only showed a more severe degeneration in all areas investigated than in patients lacking the apoE 4 allele but also revealed significantly less plastic dendritic changes. ApoE epsilon 4 allele copy number, furthermore, had a significant effect on the pattern of dendritic arborization. Moreover, the relationship between the intensity of dendritic growth and both the extent of neuronal degeneration and the stage of the disease seen in patients lacking the apoE epsilon 4 allele was very weak in the presence of one epsilon 4 allele and completely lost in patients homozygous for the epsilon 4 allele. The results provide direct evidence that neuronal reorganization is affected severely in patients with Alzheimer's disease carrying the apoE epsilon 4 allele. This impairment of neuronal repair might lead to a more rapid functional decompensation, thereby contributing to an earlier onset and more rapid progression of the disease.
Feldheim DA, Vanderhaeghen P, Hansen MJ, Frisen J, Lu Q, Barbacid M, Flanagan JG. Topographic guidance labels in a sensory projection to the forebrain. Neuron 1998 Dec;21(6):1303-13
Visual connections to the mammalian forebrain are known to be patterned by neural activity, but it remains unknown whether the map topography of such higher sensory projections depends on axon guidance labels. Here, we show complementary expression and binding for the receptor EphA5 in mouse retina and its ligands ephrin- A2 and ephrin- A5 in multiple retinal targets, including the major forebrain target, the dorsal lateral geniculate nucleus (dLGN). These ligands can act in vitro as topographically specific repellents for mammalian retinal axons and are necessary for normal dLGN mapping in vivo. The results suggest a general and economic modular mechanism for brain mapping whereby a projecting field is mapped onto multiple targets by repeated use of the same labels. They also indicate the nature of a coordinate system for the mapping of sensory connections to the forebrain.
General_screening_panel_vl.4 Summary: Ag4155 The CG55704-01 gene shows a tissue expression profile that is highly brain-preferential, with highest expression in the fetal brain (CT=27.3). Please see panel CNS_Neurodegeneration for a discussion of utility of this gene in the central nervous system.
Among metabolically relevant tissues, expression of this gene is highest in stomach, small intestine and pancreas, with lower levels in thyroid and very low levels in pituitary, fetal heart and adipose. Therefore, small molecule, peptide or antibody therapeutics designed using this gene product may be effective in modulating the development or activity of cellular processes in tissues that express this gene. Alternatively, these therapeutics may be used to alter the activity of these organs by modifying their innervation.
In addition, this gene is expressed at higher levels in the adult lung (CT=30.9) when compared to expression in the fetal lung (CT=34.8). Thus, expression of this gene could be used to differentiate between adult and fetal sources of lung tissue.
This gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Interestingly, pancreatic and brain cancer cell lines do not express this gene. Hence, the absence of expression of this gene could potentially be used as a diagnostic marker for pancreatic and brain cancer.
Panel 1.1 Summary: Ag568 Highest expression of the CG55704-01 gene is seen in the testis (CT=23.1). In addition, this gene is expressed at much higher levels in the testis than in any other samples on this panel. Thus, expression of this gene could be used as a marker of testis tissue. In addition, therapeutic modulation of the expression or function of this gene product may be beneficial in the treatment of male infertility.
Expression of this gene among metabolically relevant tissues is highest in the small intestine, stomach and pancreas, with correlates well with expression in panel 1.4. Lower levels of expression are seen in heart, pituitary and adrenal. Therefore, small molecule, peptide or antibody therapeutics designed using this gene product may be effective in modulating the development or activity of cellular processes in tissues that express this gene. Alternatively, these therapeutics may be used to alter the activity of these organs by modifying their innervation. This panel also confirms a tissue expression profile that is highly brain-preferential for this gene. Please see panel CNS_Neurodegeneration for a discusion of utility of this gene in the central nervous system.
Overall, this gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Interestingly, pancreatic and brain cancer cell lines do not express this gene. Hence the lack of expression of this gene can be used as a diagnostic marker for pancreatic and brain cancer.
Panel 1.3DSummary: Ag2879 Expression of the CG55704-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) A second experiment with probe and primer set Agl486 is not included because the amp plot suggests that there were experimental difficulties with this run.
Panel 2.2 Summary: Agl486 This gene is expressed at low but significant levels in this panel with highest expression seen in a normal colon tissue sample (CT=32.85). Single representatives of normal prostate, stomach, uterus and ovary samples also show higher expression compared to the adjacent cancer tissue. Hence, expression of this gene might be used as a marker to identify normal tissue from cancerous tissue in these organs.
Panel 2D Summary: Ag2879 Expression of the CG55704-01 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
Panel 4.1D Summary: Ag4155 hi two experiments with the same probe and primer set, the CG55704-01 transcript is expressed at low but significant levels in lung microvasculature treated with TNF-a and IL-4 and in colon. This transcript encodes an ephrin type receptor homolog, that belongs to a family of proteins which may play a role in integrin activity. Some members of this family have been described in vascular development. The regulation of the expression or activity of this protein product through the application of antibodies or small molecules may be important in controlling vascular morphogenesis, angiogenesis, leukocyte extravasation, and chemotaxis . Therefore, this gene product may be beneficial in the treatment of cancer. In addition, the protein encoded by this gene may also be useful in preventing the migration and accumulation to the lung to treat inflammatory lung diseases such asthma, emphysema or bronchitis.
The presence of this transcript in the colon suggests that the protein encoded by this gene may also play a role in the development of the colon.Therapeutics that aim to regulate the function of this protein may function to regulate cellular processes within these tissues.
Please note that a third run, Run 173333201, with the same probe and primer is not included, because the amp plot suggests that there were experimental difficulties with this run. References:
Gu C, Park S. The EphA8 receptor regulates integrin activity through pi lOgamma phosphatidylinositol-3 kinase in a tyrosine kinase activity-independent manner. Mol Cell Biol 2001 Jul;21(14):4579-97
Recent genetic studies suggest that ephrins may function in a kinase-independent Eph receptor pathway. Here we report that expression of EphA8 in either NIH 3T3 or HEK293 cells enhanced cell adhesion to fibronectin via alpha(5)beta(l)- or beta(3) integrins. Interestingly, a kinase-inactive EphA8 mutant also markedly promoted cell attachment to fibronectin in these cell lines. Using a panel of EphA8 point mutants, we have demonstrated that EphA8 kinase activity does not correlate with its ability to promote cell attachment to fibronectin. Analysis using EphA8 extracellular and intracellular domain mutants has revealed that enhanced cell adhesion is dependent on ephrin A binding to the extracellular domain and the juxtamembrane segment of the cytoplasmic domain of the receptor. EphA8-promoted adhesion was efficiently inhibited by wortmannin, a phosphatidylinositol 3-kinase (PI 3- kinase) inhibitor. Additionally, we found that EphA8 had associated PI 3-kinase activity and that the pi lOgamma isoform of PI 3-kinase is associated with EphA8. In vitro binding experiments revealed that the EphA8 juxtamembrane segment was sufficient for the formation of a stable complex with pi lOgamma. Similar results were obtained in assay using cells stripped of endogenous ephrin A ligands by treatment with preclustered ephrin A5-Fc proteins, hi addition, a membrane-targeted lipid kinase-inactive pi lOgamma mutant was demonstrated to stably associate with EphA8 and suppress EphA8 -promoted cell adhesion to fibronectin. Taken together, these results suggest the presence of a novel mechanism by which the EphA8 receptor localizes pi lOgamma PI 3-kinase to the plasma membrane in a tyrosine kinase- independent fashion, thereby allowing access to lipid substrates to enable the signals required for integrin-mediated cell adhesion
Adams RH, Klein R. Eph receptors and ephrin ligands. essential mediators of vascular development. Trends Cardiovasc Med 2000 Jul;10(5):183-8
The molecular and cellular mechanisms governing vascular development are still poorly understood. Prominent among the intercellular signals that control the initial establishment of the vascular network (termed vasculo genesis) and the subsequent remodeling process (called angiogenesis) are soluble ligands that signal through receptor tyrosine kinases (RTKs). Recent reports have added cell-bound ephrin ligands and their cognate Eph RTKs to the list of key players in vascular development.: J Biol Chem 2001 Apr 27;276(17):13771-7 Related Articles, Books, LinkOut Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, Deutsch U, Risau W, Klein R. Roles of ephrinB ligands and EpliB receptors in cardiovascular development: demarcation of arterial/venous domains, Genes Dev 1999 Feb l;13(3):295-306
Eph receptor tyrosine kinases and their cell-surface-bound ligands, the ephrins, regulate axon guidance and bundling in the developing brain, control cell migration and adhesion, and help patterning the embryo. Here we report that two ephrinB ligands and three EphB receptors are expressed in and regulate the formation of the vascular network. Mice lacking ephrinB2 and a proportion of double mutants deficient in EphB2 and EphB3 receptor signaling die in utero before embryonic day 11.5 (El 1.5) because of defects in the remodeling of the embryonic vascular system. Our phenotypic analysis suggests complex interactions and multiple functions of Eph receptors and ephrins in the embryonic vasculature. Interaction between ephrinB2 on arteries and its EphB receptors on veins suggests a role in defining boundaries between arterial and venous domains. Expression of ephrinB 1 by arterial and venous endothelial cells and EphB3 by veins and some arteries indicates that endothelial cell- to-cell interactions between ephrins and Eph receptors are not restricted to the border between arteries and veins. Furthermore, expression of ephrinB2 and EphB2 in mesenchyme adjacent to vessels and vascular defects in ephB2/ephB3 double mutants indicate a requirement for ephrin-Eph signaling between endothelial cells and surrounding mesenchymal cells. Finally, ephrinB ligands induce capillary sprouting in vitro with a similar efficiency as angiopoietin-1 (Angl) and vascular endothelial growth factor (VEGF), demonstrating a stimulatory role of ephrins in the remodeling of the developing vascular system
Panel 4D Summary: Ag 1302/Agl486 Two experiments with two different probe and primer sets show low but significant expression of the CG55704-01 gene in the colon and thymus. This expression is in agreement with the results from Panel 4. ID. The presence of this transcript in the thymus, and the colon suggests that the protein encoded by this gene may play a role in the development of these tissues. Thus, therapeutics that aim to regulate the function of the protein product may act to regulate the cellular processes within these tissues.
Please note that a third experiment with the probe and primer set Ag2879 showed low/undetectable expression in all the samples on this panel (CTs>35).
G. CG55704-03: EPHRIN TYPE-A RECEPTOR 6 PRECURSOR
Expression of gene CG55704-03 was assessed using the primer-probe sets Ag4155, Ag781, Ag568, Agl486, Ag2879 and Agl302, described in Tables 12GA, 12GB, 12GC, 12GD, 12GE and 12GF. Results of the RTQ-PCR runs are shown in Tables 12GG, 12GH, 12GI, 12GJ, 12GK, 12GL, 12GM, and 12GN. Table 12GA. Probe Name Ag4155
Figure imgf000261_0001
Table 12GB. Probe Name Ag781
Figure imgf000261_0002
Table 12GC. Probe Name Ag568
Figure imgf000261_0003
Table 12GD. Probe Name Agl486
Figure imgf000261_0004
Table 12GE. Probe Name Ag2879
Figure imgf000261_0005
Table 12GF. Probe Name Agl302
Figure imgf000261_0006
Figure imgf000261_0007
Figure imgf000262_0001
Figure imgf000263_0001
Figure imgf000263_0002
Figure imgf000264_0001
Figure imgf000265_0001
Figure imgf000266_0001
Table 12GK. Panel 2.2
Figure imgf000267_0001
Figure imgf000268_0001
Table 12GL. Panel 4. ID
Figure imgf000268_0002
Figure imgf000269_0001
Figure imgf000270_0001
Table 12GM. Panel 4D
Figure imgf000270_0002
Figure imgf000271_0001
Table 12GN. Panel CNS_1
Figure imgf000271_0002
Figure imgf000272_0001
Figure imgf000273_0001
CNS_neurodegeneration_vl.O Summary: Ag781/Ag4155 The CG55704-03 gene encodes a putative ephrin receptor, and shows a significant downregulation in the AD temporal cortex compared to nondemented controls when CT values are analyzed by ANCONA. The temporal cortex (Brodmann area 21) shows severe neurodegeneration in Alzheimer's disease, though not as early as the hippocampus or entorhinal cortex. It is therefore likely that this gene is downregulated during the process of neurodegeneration, rather than the downregulation being a result of neuron loss. The ephrin receptors have been implicated in axonal and synapse guidance. Furthermore, individuals with Alzheimer's disease (especially late-onset AD with apoE4 genotype) show impaired compensatory synaptogenesis and dendritic arborization. Therefore, this gene is an excellent small molecule target for the treatment of Alzheimer's disease. Please note that one experiment with the probe and primer set Ag2879 is not included because the amp plot indicates that there were experimental difficulties with this run.
References:
Lai KO, Ip FC, Cheung J, Fu AK, Ip ΝY. Expression of Eph receptors in skeletal muscle and their localization at the neuromuscular junction. Mol Cell Neurosci 2001 Jun;17(6):1034-47
The participation of ephrins and Eph receptors in guiding motor axons during muscle innervation has been well documented, but little is known about their expression and functional significance in muscle at later developmental stages. Our present study investigates the expression and localization of Eph receptors and ephrins in skeletal muscle. Prominent expression of EphA4, EphA7, and ephrin- A ligands was detected in muscle during embryonic development. More importantly, both EphA4 and EphA7, as well as ephrin- A2, were localized at the neuromuscular junction (NMJ) of adult muscle. Despite their relative abundance, they were not localized at the synapses during embryonic stages. The concentration of EphA4, EphA7, and ephrin- A2 at the NMJ was observed at postnatal stages and the synaptic localization became prominent at later developmental stages, h addition, expression of Eph receptors was increased by neuregulin and after nerve injury. Furthermore, we demonstrated that overexpression of EphA4 led to tyrosine phosphorylation of the actin-binding protein cortactin and that Epl A4 was coimmunoprecipitated with cortactin in muscle. Taken together, our findings indicate that EphA4 is associated with the actin cytoskeleton. Since actin cytoskeleton is critical to the formation and stability of NMJ, the present findings raise the intriguing possibility that Eph receptors may have a novel role in NMJ formation and/or maintenance.
Arendt T, Schindler C, Bruckner MK, Eschrich K, Bigl V, Zedlick D, Marcova L. Plastic neuronal remodeling is impaired in patients with Alzheimer's disease carrying apolipoprotein epsilon 4 allele. J Neurosci 1997 Jan 15;17(2):516-29
A relationship between the apolipoprotein E (apoE) genotype and the risk to develop Alzheimer's disease has been established recently. Apolipoprotein synthesis is implicated in developmental processes and in neuronal repair of the adult nervous system. In the present study, we investigated the influence of the apolipoprotein polymorphism on the severity of neuronal degeneration and the extent of plastic dendritic remodeling in Alzheimer's disease. Changes in length and arborization of dendrites of Golgi-impregnated neurons in the basal nucleus of Meynert, locus coeruleus, raphe magnus nucleus, medial amygdaloid nucleus, pedunculopontine tegmental nucleus, and substantia nigra were analyzed after three- dimensional reconstruction. Patients with either one or two apoE epsilon 4 alleles not only showed a more severe degeneration in all areas investigated than in patients lacking the apoE 4 allele but also revealed significantly less plastic dendritic changes. ApoE epsilon 4 allele copy number, furthermore, had a significant effect on the pattern of dendritic arborization. Moreover, the relationship between the intensity of dendritic growth and both the extent of neuronal degeneration and the stage of the disease seen in patients lacking the apoE epsilon 4 allele was very weak in the presence of one epsilon 4 allele and completely lost in patients homozygous for the epsilon 4 allele. The results provide direct evidence that neuronal reorganization is affected severely in patients with Alzheimer's disease carrying the apoE epsilon 4 allele. This impairment of neuronal repair might lead to a more rapid functional decompensation, thereby contributing to an earlier onset and more rapid progression of the disease.
Feldheim DA, Nanderhaeghen P, Hansen MJ, Frisen J, Lu Q, Barbacid M, Flanagan JG. Topographic guidance labels in a sensory projection to the forebrain. Neuron 1998 Dec;21(6):1303-13 Visual connections to the mammalian forebrain are known to be patterned by neural activity, but it remains unknown whether the map topography of such higher sensory projections depends on axon guidance labels. Here, we show complementary expression and binding for the receptor EphA5 in mouse retina and its ligands ephrin- A2 and ephrin- A5 in multiple retinal targets, including the major forebrain target, the dorsal lateral geniculate nucleus (dLGN). These ligands can act in vitro as topographically specific repellents for mammalian retinal axons and are necessary for normal dLGN mapping in vivo. The results suggest a general and economic modular mechanism for brain mapping whereby a projecting field is mapped onto multiple targets by repeated use of the same labels. They also indicate the nature of a coordinate system for the mapping of sensory connections to the forebrain.
General_screeningjpanel_vl.4 Summary: Ag4155 The CG55704-03 gene shows a tissue expression profile that is highly brain-preferential, with highest expression in the fetal brain (CT=27.3). Please see panel CNS_Neurodegeneration for a discussion of utility of this gene in the central nervous system.
Among metabolically relevant tissues, expression of this gene is highest in stomach, small intestine and pancreas, with lower levels in thyroid and very low levels in pituitary, fetal heart and adipose. Therefore, small molecule, peptide or antibody therapeutics designed using this gene product may be effective in modulating the development or activity of cellular processes in tissues that express this gene. Alternatively, these therapeutics may be used to alter the activity of these organs by modifying their innervation.
In addition, this gene is expressed at higher levels in the adult lung (CT=30.9) when compared to expression in the fetal lung (CT=34.8). Thus, expression of this gene could be used to differentiate between adult and fetal sources of lung tissue.
This gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Interestingly, pancreatic and brain cancer cell lines do not express this gene. Hence, the absence of expression of this gene could potentially be used as a diagnostic marker for pancreatic and brain cancer.
Panel 1.1 Summary: Ag568 Highest expression of the CG55704-03 gene is seen in the testis (CT=23.1). In addition, this gene is expressed at much higher levels in the testis than in any other samples on this panel. Thus, expression of this gene could be used as a marker of testis tissue, hi addition, therapeutic modulation of the expression or function of this gene product may be beneficial in the treatment of male infertility.
Expression of this gene among metabolically relevant tissues is highest in the small intestine, stomach and pancreas, with correlates well with expression in panel 1.4. Lower levels of expression are seen in heart, pituitary and adrenal. Therefore, small molecule, peptide or antibody therapeutics designed using this gene product may be effective in modulating the development or activity of cellular processes in tissues that express this gene. Alternatively, these therapeutics may be used to alter the activity of these organs by modifying their innervation.
This panel also confirms a tissue expression profile that is highly brain-preferential for this gene. Please see panel CNS_Neurodegeneration for a discusion of utility of this gene in the central nervous system.
Overall, this gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Interestingly, pancreatic and brain cancer cell lines do not express this gene. Hence the lack of expression of this gene can be used as a diagnostic marker for pancreatic and brain cancer.
Panel 1.2 Summary: Ag781 Highest expression of the CG55704-03 gene in this panel is seen in the cerebral cortex (CT=28.8). This panel confirms a tissue expression profile that is highly brain-preferential for this gene. Please see panel CNS_Neurodegeneration for a discusion of utility of this gene in the central nervous system.
Among metabolically relevant tissues, expression of this gene is seen in pancreas, small intestine and stomach at moderate levels. This is consistent with expression in panel 1.4 and panel 1.1. Therefore, small molecule, peptide or antibody therapeutics designed using this gene product may be effective in modulating the development or activity of cellular processes in tissues that express this gene. Alternatively, these therapeutics may be used to alter the activity of these organs by modifying their innervation.
Overall, this gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Interestingly, pancreatic and brain cancer cell lines do not express this gene. Hence the lack of expression of this gene can be used as a diagnostic marker for pancreatic and brain cancer.
Panel 1.3D Summary: Ag2879 Expression of the CG55704-03 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.) A second experiment with probe and primer set Agl486 is not included because the amp plot suggests that there were experimental difficulties with this run.
Panel 2.2 Summary: Agl486 This gene is expressed at low but significant levels in this panel with highest expression seen in a normal colon tissue sample (CT=32.85). Single representatives of normal prostate, stomach, uterus and ovary samples also show higher expression compared to the adjacent cancer tissue. Hence, expression of this gene might be used as a marker to identify normal tissue from cancerous tissue in these organs.
Panel 2D Summary: Ag2879 Expression of the CG55704-03 gene is low/undetectable in all samples on this panel (CTs>35). (Data not shown.)
Panel 4.1D Summary: Ag4155 In two experiments with the same probe and primer set, the CG55704-03 transcript is expressed at low but significant levels in lung microvasculature treated with TNF-a and IL-4 and in colon. This transcript encodes an ephrin type receptor homolog, that belongs to a family of proteins that may play a role in integrin activity. Some members of this family have been described in vascular development. The regulation of the expression or activity of this protein product through the application of antibodies or small molecules may be important in controlling vascular morphogenesis, angiogenesis, leukocyte extravasation, and chemotaxis. Therefore, this gene product may be beneficial in the treatment of cancer. In addition, the protein encoded by this gene may also be useful in preventing the migration and accumulation of leukocytes to the lung to treat inflammatory lung diseases such asthma, emphysema or bronchitis.
The presence of this transcript in the colon suggests that the protein encoded by this gene may also play a role in the development of the colon.Therapeutics that aim to regulate the function of this protein may function to regulate cellular processes within these tissues.
Please note that a third run, Run 173333201, with the same probe and primer is not included, because the amp plot suggests that there were experimental difficulties with this run.
References:
Gu C, Park S. The EphA8 receptor regulates integrin activity through pi lOgamma phosphatidylinositol-3 kinase in a tyrosine kinase activity-independent manner. Mol Cell Biol 2001 Jul;21(14):4579-97
Recent genetic studies suggest that ephrins may function in a kinase-independent Eph receptor pathway. Here we report that expression of EphA8 in either NIH 3T3 or HEK293 cells enhanced cell adhesion to fibronectin via alpha(5)beta(l)- or beta(3) integrins. Interestingly, a kinase-inactive EphA8 mutant also markedly promoted cell attachment to fibronectin in these cell lines. Using a panel of EphA8 point mutants, we have demonstrated that EphA8 kinase activity does not correlate with its ability to promote cell attachment to fibronectin. Analysis using EphA8 extracellular and intracellular domain mutants has revealed that enhanced cell adhesion is dependent on ephrin A binding to the extracellular domain and the juxtamembrane segment of the cytoplasmic domain of the receptor. EphA8-promoted adhesion was efficiently inhibited by wortmannin, a phosphatidylinositol 3-kinase (PI 3- kinase) inhibitor. Additionally, we found that EphA8 had associated PI 3-kinase activity and that the pi lOgamma isoform of PI 3-kinase is associated with EphA8. In vitro binding experiments revealed that the EphA8 juxtamembrane segment was sufficient for the formation of a stable complex with pi lOgamma. Similar results were obtained in assay using cells stripped of endogenous ephrin A ligands by treatment with preclustered ephrin A5-Fc proteins. In addition, a membrane-targeted lipid kinase-inactive pi lOgamma mutant was demonstrated to stably associate with EphA8 and suppress EphA8-promoted cell adhesion to fibronectin. Taken together, these results suggest the presence of a novel mechanism by which the EphA8 receptor localizes pi lOgamma PI 3-kinase to the plasma membrane in a tyrosine kinase- independent fashion, thereby allowing access to lipid substrates to enable the signals required for integrin-mediated cell adhesion
Adams RH, Klein R. Eph receptors and ephrin ligands. essential mediators of vascular development. Trends Cardiovasc Med 2000 Jul;10(5):183-8
The molecular and cellular mechanisms governing vascular development are still poorly understood. Prominent among the intercellular signals that control the initial establishment of the vascular network (termed vasculogenesis) and the subsequent remodeling process (called angiogenesis) are soluble ligands that signal through receptor tyrosine kinases (RTKs). Recent reports have added cell-bound ephrin ligands and their cognate Eph RTKs to the list of key players in vascular development.: J Biol Chem 2001 Apr 27;276(17): 13771-7 Related Articles, Books, LinkOut
Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, Deutsch U, Risau W, Klein R. Roles of ephrinB ligands and EphB receptors in cardiovascular development: demarcation of arterial/venous domains, Genes Dev 1999 Feb l;13(3):295-306
Eph receptor tyrosine kinases and their cell-surface-bound ligands, the ephrins, regulate axon guidance and bundling in the developing brain, control cell migration and adhesion, and help patterning the embryo. Here we report that two ephrinB ligands and three EphB receptors are expressed in and regulate the formation of the vascular network. Mice lacking ephrinB2 and a proportion of double mutants deficient in EphB2 and EphB3 receptor signaling die in utero before embryonic day 11.5 (El 1.5) because of defects in the remodeling of the embryonic vascular system. Our phenotypic analysis suggests complex interactions and multiple functions of Eph receptors and ephrins in the embryonic vasculature. Interaction between ephrinB2 on arteries and its EphB receptors on veins suggests a role in defining boundaries between arterial and venous domains. Expression of ephrinB 1 by arterial and venous endothelial cells and EphB3 by veins and some arteries indicates that endothelial cell- to-cell interactions between ephrins and Eph receptors are not restricted to the border between arteries and veins. Furthermore, expression of ephrinB2 and EphB2 in mesenchyme adjacent to vessels and vascular defects in eplιB2/ephB3 double mutants indicate a requirement for ephrin-Eph signaling between endothelial cells and surrounding mesenchymal cells. Finally, ephrinB ligands induce capillary sprouting in vitro with a similar efficiency as angiopoietin-1 (Angl) and vascular endothelial growth factor (NEGF), demonstrating a stimulatory role of ephrins in the remodeling of the developing vascular system
Panel 4D Summary: Ag 1302/Agl486 Two experiments with two different probe and primer sets show low but significant expression of the CG55704-03 gene in the colon and thymus. This expression is in agreement with the results from Panel 4. ID. The presence of this transcript in the thymus, and the colon suggests that the protein encoded by this gene may play a role in the development of these tissues. Thus, therapeutics that aim to regulate the function of the protein product may act to regulate the cellular processes within these tissues.
Please note that a third experiment with the probe and primer set Ag2879 showed low/undetectable expression in all the samples on this panel (CTs>35). (Data not shown.)
Panel CΝS_1 Summary: Ag781 This panel confirms a tissue expression profile that is highly brain-preferential; see panel CNS_Neurodegeneration for a discussion of utility the CG55704-03 gene in the central nervous system.
H. CG95545-01/CG95545-01 and CG95545-02: Type IA membrane sushi- containing domain Protein
Expression of gene CG95545-01 and variant CG95545-02 was assessed using the primer-probe sets Ag4000, Agl923 and Ag729, described in Tables 12HA, 12HB and 12HC. Results of the RTQ-PCR runs are shown in Tables 12HD, 12HE, 12HF, 12HG, 12HH and 12HI.
Table 12HA. Probe Name Ag4000
Figure imgf000279_0001
Table 12HB. Probe Name Agl923
Figure imgf000279_0002
ID NO : 166)
Table 12HC. Probe Name Ag729
Figure imgf000280_0001
Table 12HD . CNS_neurodegeneration_vl .0
Figure imgf000280_0002
Table 12HE . General_screening_panel_vl .4
Figure imgf000280_0003
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Table 12HG. Panel 2.2
Figure imgf000283_0002
Figure imgf000284_0001
Table 12HH. Panel 4. ID
Figure imgf000284_0002
Figure imgf000285_0001
Figure imgf000286_0001
Table 12HI . Panel 4D
Figure imgf000286_0002
Figure imgf000287_0001
CNS_neurodegeneration_vl.O Summary: Ag4000 The CG95545-01 gene encodes a novel membrane receptor, and shows a significant downregulation in the AD temporal cortex compared to nondemented controls when CT values are analyzed by ANCOVA. The temporal cortex (Brodman area 21) shows severe neurodegeneration in Alzheimer's disease, though not as early as the hippocampus or entorhinal cortex.Thus, it is likely that this gene is downregulated during the process of neurodegeneration rather than the downregulation being a result of neuron loss. Levels in the brain are also moderate to high as determined by panels 1.2 and General_Screening_1.4. Thus this gene is an excellent small molecule target for the treatment of Alzheimer's disease.
General_screeningjpanel_vl.4 Summary: Agl923 The CG95545-01 gene is ubiquitously expressed in the cancer cell lines used on this panel as well as the normal tissues. The highest level of expression is in the colon cancer CaCo-2 cell line (CT=27.3). This widespread expression suggests that the protem encoded by this gene is potentially useful for cell growth and survival.
This panel further confirms the expression of this gene in the CNS. See panel CNS_Neurodegeneration for a discussion of utility of this gene in the central nervous system. Among metabolic tissues, highest expression of this gene is in the placenta and pancreas. Lower levels of expression are seen in adrenal, adipose, pituitary, thyroid, small intestine, stomach, fetal skeletal muscle, fetal liver, fetal kidney, fetal heart, heart, skeletal muscle, liver and kidney. Thus, peptide and antibody therapeutics using this gene product may also be used to modulate the development and/or physiological activities in these tissues.
Furthermore, higher levels of expression in the fetal liver and lung (CTs=28-29) when compared to expression in the adult liver and lung (CTs=32-33) suggest that expression of this gene could be used to differentiate between adult and fetal sources of these tissues. In addition, the higher levels of expression in the fetal tissues suggests that the protein encoded by this gene may be involved in the development of the liver and lung and thus may be useful in treatment of diseases of these organs in the adult.
Panel 1.2 Summary: Ag729 The CG95545-01 gene is ubiquitously expressed in the cancer cell lines used on this panel as well as the normal tissues. The highest level of expression is in the CaCo-2 cell line (CT=24). Both of these observations are in excellent agreeement with the results from General_screening_panel_vl.4. This expression profile suggests that expression of this gene is potentially useful for cell growth and survival.
Among metabolically relevant tissues, highest expression is seen in the placenta, followed by the kidney, fetal kidney, pituitary, pancreas, small intestine, stomach and thyroid. Relatively high levels of expression according to the CT value are also seen in heart, skeletal muscle, liver and fetal liver. Thus, peptide and antibody therapeutics using this gene product may also be used to modulate the development and/or physiological activities in these tissues.
This panel also confirms the expression of this gene in the CNS. See panel CNS_Neurodegeneration for a discussion of utility of this gene in the central nervous system.
Panel 2.2 Summary: Agl923 The CG95545-01 gene is expressed at a low level in all normal and tumor samples on this panel. The highest level of expression is seen in a sample of normal adjacent kidney CT=29.6). A distinct difference is seen in gastric cancer where normal tissues express it at a slightly higher level than gastric tumors. Thus, expression of this gene could potentially be used as a marker for gastric tumors.
Panel 4.1D Summary: Ag 4000 The highest expression of the CG95545-01 transcript is found in Ku-812 after treatment with PMA and ionomycin(CT=27.4), a condition that stimulates the release of mediators such as histamine and proteases that are responsible for the symptomatology of diverse atopic diseases. This transcript is also expressed in a wide range of cells that participate in the immune response (monocytes, T, B and NK cells)and inflammatory processes (dermal and lung fibroblasts). Therefore, modulation of the expression or activity of the protein encoded by this transcript through the application of antibodies or peptides therapeutics may be beneficial for the treatment of lung inflammatory diseases such as asthma, and chronic obstructive pulmonary diseases, inflammatory skin diseases such as psoriasis, atopic dermatitis, ulcerative dermatitis, ulcerative colitis and autoimmune diseases such as Crohn's disease, lupus erythematosus, rheumatoid arthritis and osteoarthritis.
Panel 4D Summary: Ag4000 Expression of the CG95545-01 transcript is ubiquitous among the samples on this panel. Please see Panel 4. ID for discussion of utility of this gene in the immune/inflammatory response.
I. CG55746-01 and CG55746_05: Butyrophilin-like Protein Expression of gene CG55746-01 and variant CG55746_05 was assessed using the primer-probe set Ag2361, described in Table 12IA. Results of the RTQ-PCR runs are shown in Tables 12IB, 12IC and 12ID.
Table 12IA. Probe Name Ag2361
Figure imgf000289_0001
Table 12IB. Panel 1.3D
Figure imgf000289_0002
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Table 12 ID. Panel 4D
Figure imgf000292_0002
Figure imgf000293_0001
Panel 1.3D Summary: Ag2361 The CG55746-01 gene is expressed at a moderately high level in brain, breast and renal cancer cell lines compared to the normal tissue, with highest expression in a brain cancer cell line (CT=28.5). Hence, the expression of this gene could be of use as a marker for different grades/ types of brain cancer, renal cancer and breast cancer that were used in the derivation of these cell lines, h addition, therapeutic inhibition of the activity of the product of this gene, through the use of small molecule drags, may be useful in the therapy of brain, renal and breast cancer.
Among metabolic tissues, expression of this butyrophilin-like gene is highest in the placenta, with lower levels in fetal skeletal muscle, adipose, stomach, small intestine, adrenal, thyroid, heart, liver and fetal liver. This molecule may be involved in the interaction of the immune system with these organs and modulation of this gene product by peptide and antibody therapeutics may alleviate disorders originating in these tissues.
This gene, a butyrophilin homolog, shows moderate to low expression in the CNS. Butyrophilin has been shown to modulate the immune response in multiple sclerosis, suggesting that this protein may be useful in the treatment of this disease or other diseases associated with immune system-induced myelin damage.
References:
Stefferl A, Schubart A, Storch2 M, Amini A, Mather I, Lassmann H, Linington C. Butyrophilin, a milk protein, modulates the encephalitogenic T cell response to myelin oligodendrocyte glycoprotein in experimental autoimmune encephalomyelitis. J Immunol 2000 Sep l;165(5):2859-65
Experimental autoimmune encephalomyelitis (EAE) induced by sensitization with myelin oligodendrocyte glycoprotein (MOG) is a T cell-dependent autoimmune disease that reproduces the inflammatory demyelinating pathology of multiple sclerosis. We report that an encephalitogenic T cell response to MOG can be either induced or alternatively suppressed as a consequence of immunological cross-reactivity, or "molecular mimicry" with the extracellular IgV-like domain of the milk protein butyrophilin (BTN). In the Dark Agouti rat, active immunization with native BTN triggers an inflammatory response in the CNS characterized by the formation of scattered meningeal and perivascular infiltrates of T cells and macrophages. We demonstrate that this pathology is mediated by a MHC class II- restricted T cell response that cross-reacts with the MOG peptide sequence 76-87, 1 GEG KNA LRIQ Ν (identities underlined). Conversely, molecular mimicry with BTΝ can be exploited to suppress disease activity in MOG-induced EAE. We demonstrate that not only is EAE mediated by the adoptive transfer of MOG74-90 T cell lines markedly ameliorated by i.v. treatment with the homologous BTΝ peptide, BTΝ74-90, but that this protective effect is also seen in actively induced disease following transmucosal (intranasal) administration of the peptide. These results identify a mechanism by which the consumption of milk products may modulate the pathogenic autoimmune response to MOG.
Panel 2D Summary: Ag2361 The CG55746-01 gene is ubiquitously expressed in all tissues in this panel, with highest expression in normal lung tissue adjacent to a tumor (CT=28.4). There is significantly higher expression in normal lung tissue compared to melanomas that have metastasized to lung. Thus, the expression can be used to differentiate between normal lung tissue and metastatic melanomas. Panel 4D Summary: Ag 2361 The CG55746-01 transcript is ubiquitously expressed at moderate levels in all cell types of this panel, with highest expression of this transcript is found in lung fibroblasts upon IFN g treatment(CT=26.8). High levels of expression are also seen in dermal fibroblasts treated with TNF-a , HUNEC treated with TΝF-a and IFΝg and small airway epithelium treated with TΝF-a and IL-lb. In all these cell types, the expression of this transcript, although constitutive, is dramatically up-regulated upon treatment with the potent inflammatory cytokines TΝF-a and IFΝg, suggesting a role for the protein encoded by this transcript in these cell types during inflammation. Therefore, modulation of this gene product by antibodies or small molecules therapeutics may be beneficial for the treatment of the symptoms associated with the inflammatory processes observed in asthma, chronic obstructive pulmonary diseases and psoriasis.
J. CG50329-01: BUTYROPHILIΝ-like protein
Expression of gene CG50329-01 was assessed using the primer-probe sets Ag2563 and Ag2563b, described in Tables 12JA and 12JB. Results of the RTQ-PCR runs are shown in Tables 12JC, and 12D.
Table 12JA. Probe Name Ag2563
Figure imgf000295_0001
Table 1 JB. Probe Name Ag2563b
Figure imgf000295_0002
Table 12 C. General_screening_panel_vl .4
Figure imgf000295_0003
Figure imgf000296_0001
Table 12 JD. Panel 4. ID
Figure imgf000297_0001
Figure imgf000298_0001
CNS_neurodegeneration_vl.O Summary: Ag2563 Expression of the CG50329-01 gene is insignificant/undetectable in all samples on this panel. (Data not shown.)
General_screening_panel_vl.4 Summary: Ag2563b Highest expression of the CG50329-01 gene is seen in fetal liver (CT=28.1). Thus, this gene maybe involved in development of the liver and may be used to differentiate fetal and adult liver, hi addition, peptide or antibody therapeutics may be used to modulate the activity of its gene product to influence development or function of the liver.
This gene is expressed at a low level in most of the cancer cell lines and normal tissues on this panel. Lung and ovarian cancer cell lines express this gene at a higher level than the normal lung and ovary tissues. Hence, expression of this gene can be used as a diagnostic marker for the lung and ovarian cancers used for the derivation of these cell lines.
This gene encodes a novel butyrophilin-like protein with low expression in the spinal cord. Butyrophilin has been shown to modulate the immune response in multiple sclerosis, suggesting that this protein may be useful in the treatment of this or other diseases associated with immune system-induced myelin damage.
References:
Stefferl A, Schubart A, Storch2 M, Amini A, Mather I, Lassmann H, Linington C. Butyrophilin, a milk protein, modulates the encephalitogenic T cell response to myelin oligodendrocyte glycoprotein in experimental autoimmune encephalomyelitis. J Immunol 2000 Sep l;165(5);2859-65 Experimental autoimmune encephalomyelitis (EAE) induced by sensitization with myelin oligodendrocyte glycoprotein (MOG) is a T cell-dependent autoimmune disease that reproduces the inflammatory demyelinating pathology of multiple sclerosis. We report that an encephalitogenic T cell response to MOG can be either induced or alternatively suppressed as a consequence of immunological cross-reactivity, or "molecular mimicry" with the extracellular IgN-like domain of the milk protein butyrophilin (BTΝ). In the Dark Agouti rat, active immunization with native BTΝ triggers an inflammatory response in the CΝS characterized by the formation of scattered meningeal and perivascular infiltrates of T cells and macrophages. We demonstrate that this pathology is mediated by a MHC class II- restricted T cell response that cross-reacts with the MOG peptide sequence 76-87, 1 GEG KNA LRIQ Ν (identities underlined). Conversely, molecular mimicry with BTΝ can be exploited to suppress disease activity in MOG-induced EAE. We demonstrate that not only is EAE mediated by the adoptive transfer of MOG74-90 T cell lines markedly ameliorated by i.v. treatment with the homologous BTΝ peptide, BTΝ74-90, but that this protective effect is also seen in actively induced disease following transmucosal (intranasal) administration of the peptide. These results identify a mechanism by which the consumption of milk products may modulate the pathogenic autoimmune response to MOG.
Panel 1.3D Summary: Ag2563 One experiment with this probe and primer set failed along with the genomic DNA control. (Data not shown.)
Panel 2D Summary: Ag2563 Ag2563 Expression of the CG50329-01 gene is low/undetectable in all samples on this panel. (CTs>35)(Data not shown.)
Panel 4.1D Summary: Ag 2563b: The highest expression of the CG50329-01 transcript is found in kidney, thymus and lung. Thus, the protein encoded by this transcript may play an important role in the normal homeostasis of these tissues. This gene is also expressed in KU-812, a basophil cell line. This cell type is involved in atopic diseases such as asthma, contact dermatitis and other inflammatory diseases such as inflammatory bowel disease. Therefore, antibodies or small molecule therapeutics designed with the protein encoded by this transcript may be important for maintaining or restoring normal function to thymus and lung during inflammation and in particular for the treatment of asthma, inflammatory bowel disease and allergies.
Panel 4D Summary: Ag2563 Two experiments with this probe and primer set failed along with the genomic DNA control. (Data not shown.) Example 3. SNP analysis of NOVX clones
SeqCallingTM Technology: cDNA was derived from various human samples representing multiple tissue types, normal and diseased states, physiological states, and developmental states from different donors. Samples were obtained as whole tissue, cell lines, primary cells or tissue cultured primary cells and cell lines. Cells and cell lines may have been treated with biological or chemical agents that regulate gene expression for example, growth factors, chemokines, steroids. The cDNA thus derived was then sequenced using CuraGen's proprietary SeqCalling technology. Sequence traces were evaluated manually and edited for corrections if appropriate. cDNA sequences from all samples were assembled with themselves and with public ESTs using bioinformatics programs to generate CuraGen's human SeqCalling database of SeqCalling assemblies. Each assembly contains one or more overlapping cDNA sequences derived from one or more human samples. Fragments and ESTs were included as components for an assembly when the extent of identity with another component of the assembly was at least 95% over 50 bp. Each assembly can represent a gene and/or its variants such as splice forms and/or single nucleotide polymoφhisms (SNPs) and their combinations.
Variant sequences are included in this application. A variant sequence can include a single nucleotide polymorphism (SNP). A SNP can, in some instances, be referred to as a "cSNP" to denote that the nucleotide sequence containing the SNP originates as a cDNA. A SNP can arise in several ways. For example, a SNP may be due to a substitution of one nucleotide for another at the polymorphic site. Such a substitution can be either a transition or a transversion. A SNP can also arise from a deletion of a nucleotide or an insertion of a nucleotide, relative to a reference allele. In this case, the polymoφhic site is a site at which one allele bears a gap with respect to a particular nucleotide in another allele. SNPs occurring within genes may result in an alteration of the amino acid encoded by the gene at the position of the SNP. hitragenic SNPs may also be silent, however, in the case that a codon including a SNP encodes the same amino acid as a result of the redundancy of the genetic code. SNPs occurring outside the region of a gene, or in an intron within a gene, do not result in changes in any amino acid sequence of a protein but may result in altered regulation of the expression pattern for example, alteration in temporal expression, physiological response regulation, cell type expression regulation, intensity of expression, stability of transcribed message.
Method of novel SNP Identification: SNPs are identified by analyzing sequence assemblies using CuraGen's proprietary SNPTool algorithm. SNPTool identifies variation in assemblies with the following criteria: SNPs are not analyzed within 10 base pairs on both ends of an alignment; Window size (number of bases in a view) is 10; The allowed number of mismatches in a window is 2; Minimum SNP base quality (PHRED score) is 23; Minimum number of changes to score an SNP is 2/assembly position. SNPTool analyzes the assembly and displays SNP positions, associated individual variant sequences in the assembly, the depth of the assembly at that given position, the putative assembly allele frequency, and the SNP sequence variation. Sequence traces are then selected and brought into view for manual validation. The consensus assembly sequence is imported into CuraTools along with variant sequence changes to identify potential amino acid changes resulting from the SNP sequence variation. Comprehensive SNP data analysis is then exported into the SNPCalling database.
Method of novel SNP Confirmation: SNPs are confirmed employing a validated method know as Pyrosequencing (Pyrosequencing, Westborough, MA). Detailed protocols for Pyrosequencing can be found in: Alderbom et al. Determination of Single Nucleotide Polymoφhisms by Real-time Pyrophosphate DNA Sequencing. (2000). Genome Research. 10, Issue 8, August. 1249-1265. In brief, Pyrosequencing is a real time primer extension process of genotyping. This protocol takes double-stranded, biotinylated PCR products from genomic DNA samples and binds them to streptavidin beads. These beads are then denatured producing single stranded bound DNA. SNPs are characterized utilizing a technique based on an indirect bioluminometric assay of pyrophosphate (PPi) that is released from each dNTP upon DNA chain elongation. Following Klenow polymerase-mediated base incoφoration, PPi is released and used as a substrate, together with adenosine 5'-phosphosulfate (APS), for ATP sulfurylase, which results in the foπnation of ATP. Subsequently, the ATP accomplishes the conversion of luciferin to its oxi-derivative by the action of luciferase. The ensuing light output becomes proportional to the number of added bases, up to about four bases. To allow processivity of the method dNTP excess is degraded by apyrase, which is also present in the starting reaction mixture, so that only dNTPs are added to the template during the sequencing. The process has been fully automated and adapted to a 96-well format, which allows rapid screening of large SNP panels. The DNA and protem sequences for the novel single nucleotide polymoφhic variants are reported. Variants are reported individually but any combination of all or a select subset of variants are also included. In addition, the positions of the variant bases and the variant amino acid residues are underlined.
Results
Variants are reported individually but any combination of all or a select subset of variants are also included as contemplated NOVX embodiments of the invention. NOV3
The DNA and protein sequences for the novel single nucleotide polymoφhic variants of the Beta Adrenergic Receptor Kinase-like gene of CuraGen Ace. No. CG50345-01 are reported in Table 13. Variants are reported individually but any combination of all or a select subset of variants are also included. The positions of the variant bases and the variant amino acid residues are underlined. In summary, there are 5 variants reported, whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID NOs:l 1 and 12, respectively. Variant 13375845 is an A to C SNP at 203 bp of the nucleotide sequence that results in no change in the protein sequence (silent), variant 13375846 is an A to G SNP at 292 bp of the nucleotide sequence that results in a Lys to Arg change at amino acid 62 of protein sequence, variant 13376064 is a G to A SNP at 1814 bp of the nucleotide sequence that results in a Tφ to End change at amino acid 569 of protein sequence, variant 13376063 is a T to C SNP at 1885 bp of the nucleotide sequence that results in an He to Thr change at amino acid 593 of protein sequence, and variant 13376062 is a G to A SNP at 2001 bp of the nucleotide sequence that results in a Glu to Lys change at amino acid 632 of protein sequence.
Figure imgf000302_0001
NOV4
There are 3 variants reported in Table 14, whose variant positions for its nucleotide and amino acid sequences is numbered according to SEQ ID Nos: 13 and 14, respectively. Variant 13374261 is an A to G SNP at 117 bp of the nucleotide sequence that results in an Asp to Gly change at amino acid 28 of protein sequence, variant 13374262 is a T to C SNP at 225 bp of the nucleotide sequence that results in a Val to Ala change at amino acid 64 of protein sequence, and variant 13374263 is a G to A SNP at 260 bp of the nucleotide sequence that results in an Ala to Thr change at amino acid 76 of protein sequence.
Figure imgf000303_0001
NOV5A
The DNA and protein sequences for the novel single nucleotide polymoφhic variants of the Out-At-First-like gene of CuraGen Ace. No. CG55764-01 are reported in Table 15. Variants are reported individually but any combination of all or a select subset of variants are also included. There are 4 variants reported whose variant positions for its nucleotide and amino acid sequences which are numbered according to SEQ ID NOs:15 and 16, respectively. Variant 13374591 is an A to G SNP at 281 bp of the nucleotide sequence that results in a Gin to Arg change at amino acid 94 of protein sequence, variant 13374592 is an A to G SNP at 344 bp of the nucleotide sequence that results in a Glu to Gly change at amino acid 115 of protein sequence, variant 13374593 is a G to A SNP at 629 bp of the nucleotide sequence that results in an Arg to His change at amino acid 210 of protein sequence, and variant 13374594 is an A to G SNP at 650 bp of the nucleotide sequence that results in a His to Arg change at amino acid 217 of protein sequence.
Figure imgf000303_0002
Figure imgf000304_0001
NOV6A
The DNA and protein sequences for the novel single nucleotide polymoφhic variants of the EphA6 ehk 2-like gene of CuraGen Ace. No. CG55704-01 are reported in Table 16. Variants are reported individually but any combination of all or a select subset of variants are also included. There are 2 variants reported whose variant positions for its nucleotide and amino acid sequences are numbered according to SEQ ID NOs:19 and 20, respectively. Variant 13376314 is a C to T SNP at 1674 bp of the nucleotide sequence that results in no change in the protein sequence (silent), and variant 13376315 is a G to A SNP at 2889 bp of the nucleotide sequence that results in no change in the protein sequence (silent).
Figure imgf000304_0002
NOV8 AND NOV9
The DNA and protein sequences for the novel single nucleotide polymoφhic variants of the Type la Membrane Sushi-Containing Domain-like gene of CuraGen Ace. No. CG95545-01 are reported in Table 17. Variants are reported individually but any combination of all or a select subset of variants are also included. There is one variant reported whose variant position for its nucleotide and amino acid sequences are numbered according to SEQ ID NOs:25, 26, 27 and 28, respectively. Variant 13376324 is a T to G SNP at 2693 bp of the nucleotide sequence that results in no change in the protein sequence since the SNP is not in the amino acid coding region.
Table 17. cSNP and Coding Variants for NOV8 and NOV9
NT Position Wild Type Variant Amino Acid Amino Acid
Figure imgf000305_0001
NOV10A
The DNA and protein sequences for the novel single nucleotide polymoφhic variants of the BUTYROPHILIN-like gene of CuraGen Ace. No. CG55746-01 are reported in Table 18. Variants are reported individually but any combination of all or a select subset of variants are also included. There are 6 variants reported whose variant positions for its nucleotide and amino acid sequences are numbered according to SEQ ID NOs:29 and 30, respectively. Variant 13376321 is an A to G SNP at 426 bp of the nucleotide sequence that results in no change in the protein sequence (silent), variant 13376320 is a C to T SNP at 506 bp of the nucleotide sequence that results in an Ala to Val change at amino acid 154 of protein sequence, variant 13376319 is a G to A SNP at 515 bp of the nucleotide sequence that results in a Ser to Asn change at amino acid 157 of protein sequence, variant 13376318 is an A to T SNP at 583 bp of the nucleotide sequence that results in an Arg to End change at amino acid 180 of protein sequence, variant 13376317 is a T to C SNP at 641 bp of the nucleotide sequence that results in an He to Thr change at amino acid 199 of protein sequence, and variant 13376316 is a T to C SNP at 743 bp of the nucleotide sequence that results in an He to Thr change at amino acid 233 of protein sequence.
Figure imgf000305_0002
EXAMPLE 4. PCR CLONING
NOV4: CG50301-01: human TENM4 The cDNA coding for a domain of the full length of CG50301-01 between residues 371 to 830 was targeted for "in-frame" cloning by PCR. The PCR template is based on human cDNA(s).
The following oligonucleotide primers identified as SEQ ID NOs:92 and 93 were used to clone the target cDNA sequence:
FI 5'-GGATCC CACCTGCAGCCGATGGAGGGGCAGATGTATGAG-3*
Rl 5'-CTCGAGACAGCCAGCTCCTCTCCAGCCCAGCTGGCAGACG-3'
For downstream cloning puφoses, the forward primer (FI: SEQ ID NO:92) includes an in-frame BamHI restriction site and the reverse primer (Rl : SEQ ID NO:93) contains an in- frame Xhol restriction site.
Two parallel PCR reactions were set up using a total of 0.5-1.0 ng human pooled cDNAs as template for each reaction. The pool is composed of 5 micrograms of each of the following human tissue cDNAs: adrenal gland, whole brain, amygdala, cerebellum, thalamus, bone marrow, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, liver, lymphoma, Burkitt's Raji cell line, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small Intestine, spleen, stomach, thyroid, trachea, uterus.
When the tissue of expression is known and available, the second PCR was performed using the above primers and 0.5ng-1.0 ng of one of the following human tissue cDNAs: skeleton muscle, testis, mammary gland, adrenal gland, ovary, colon, normal cerebellum, normal adipose, normal skin, bone marrow, brain amygdala, brain hippocampus, brain substantia nigra, brain thalamus, thyroid, fetal lung, fetal liver, fetal brain, kidney, heart, spleen, uterus, pituitary gland, lymph node, salivary gland, small intestine, prostate, placenta, spinal cord, peripheral blood, trachea, stomach, pancreas, hypothalamus.
The reaction mixtures contained 2 microliters of each of the primers (original concentration: 5 pmol/ul), 1 microliter of lOmM dNTP (Clontech Laboratories, Palo Alto CA) and 1 microliter of 50xAdvantage-HF 2 polymerase (Clontech Laboratories) in 50 microliter- reaction volume. The following reaction conditions were used:
PCR condition 1: a) 96°C 3 minutes b) 96°C 30 seconds denaturation c) 60°C 30 seconds, primer annealing d) 72°C 6 minutes extension Repeat steps b-d 15 times e) 96°C 15 seconds denaturation f) 60°C 30 seconds, primer annealing g) 72°C 6 minutes extension Repeat steps e-g 29 times e) 72°C 10 minutes final extension PCR condition 2: a) 96°C 3 minutes b) 96°C 15 seconds denaturation c) 76°C 30 seconds, primer annealing, reducing the temperature by 1 °C per cycle d) 72°C 4 minutes extension Repeat steps b-d 34 times e) 72°C 10 minutes final extension
An amplified product was detected by agarose gel electrophoresis. The fragment was gel-purified and ligated into the pCR2.1 vector (Invitrogen, Carlsbad, CA) following the manufacturer's recommendation. Twelve clones per PCR reaction were picked and sequenced. The inserts were sequenced using vector-specific Ml 3 Forward and Ml 3 Reverse primers and the following gene-specific primers:
SEQIDNO:94: SF1: TGGAGATCTCAAGTGTTCATAGACCATC
SEQIDNO:95: SF2: ACAGGCTTCATCCAGTATTTGGATTC
SEQIDNO:96: SF3: AAATGGCCAATACATGAAAGGCA
SEQDDNO:97: SF4: ATTGCTTTGTGGGATGGGGAG
SEQIDNO:98: SF5: AATGGCGAACACTGCACCATC
SEQIDNO:99: SRI: AAGTGCCAGGAGGAATCTTCTGGGAGG
SEQIDNO:100: SR2: GAAGCCTGTCTCATGGCTGGAG
SEQIDNO:101: SR3: ATTTCCGCTACAGAGCACGGG
SEQ ID NO:102: SR4: ATTCGCCTCTCACGCAGACAC
SEQIDNO:103: SR5: ACCACAGTCGGCAGCACAGAT
The insert 172885447 was found to encode an open reading frame similar to that between residues 371 and 830 of the target sequence of CG50301-01. The cloned insert is 99% identical to the original sequence. It differs from the original sequence at 3 nucleotide positions and one amino acid position.
NOV 11: CG50329-01
The cDNA coding for a domian of CG50329-01 from residue 32 to 236 was targeted for "in-frame" cloning by PCR. The PCR template is based human cDNA(s).
The following oligonucleotide primers were used to clone the target cDNA sequence:
FI 5'-GGATCC AAAGCTGACTTTGATGTCACTGGGCCTCATGC-3'
R3 5'-CTCGAG CCTTTCAGGGAGGAGGGGGCTGGAGATGG-3'
For downstream cloning puφoses, the forward primer (FI: SEQ ID NO.T04) includes an in-frame BamHI restriction site and the reverse primer (R3: SEQ ID NO: 105) contains an in-frame Xhol restriction site.
Two parallel PCR reactions were set up using a total of 0.5-1.0 ng human pooled cDNAs as template for each reaction. The pool is composed of 5 micro grams of each of the following human tissue cDNAs: adrenal gland, whole brain, amygdala, cerebellum, thalamus, bone marrow, fetal brain, fetal kidney, fetal liver, fetal lung, heart, kidney, liver, lymphoma, Burkitt's Raji cell line, mammary gland, pancreas, pituitary gland, placenta, prostate, salivary gland, skeletal muscle, small Intestine, spleen, stomach, thyroid, trachea, uterus.
When the tissue of expression is known and available, the second PCR was performed using the above primers and 0.5ng-1.0 ng of one of the following human tissue cDNAs: skeleton muscle, testis, mammary gland, adrenal gland, ovary, colon, normal cerebellum, normal adipose, normal skin, bone marrow, brain amygdala, brain hippocampus, brain substantia nigra, brain thalamus, thyroid, fetal lung, fetal liver, fetal brain, kidney, heart, spleen, uterus, pituitary gland, lymph node, salivary gland, small intestine, prostate, placenta, spinal cord, peripheral blood, trachea, stomach, pancreas, hypothalamus.
The reaction mixtures contained 2 microliters of each of the primers (original concentration: 5 pmol/ul), 1 microliter of lOmM dNTP (Clontech Laboratories, Palo Alto CA) and 1 microliter of 50xAdvantage-HF 2 polymerase (Clontech Laboratories) in 50 microliter- reaction volume. The following reaction conditions were used:
PCR condition 1: a) 96°C 3 minutes b) 96°C 30 seconds denaturation c) 60°C 30 seconds, primer annealing d) 72°C 6 minutes extension Repeat steps b-d 15 times e) 96°C 15 seconds denaturation f) 60°C 30 seconds, primer annealing g) 72°C 6 minutes extension Repeat steps e-g 29 times e) 72°C 10 minutes final extension
PCR condition 2: a) 96°C 3 minutes b) 96°C 15 seconds denaturation c) 76°C 30 seconds, primer annealing, reducing the temperature by 1 °C per cycle d) 72°C 4 minutes extension Repeat steps b-d 34 times e) 72°C 10 minutes final extension
An amplified product was detected by agarose gel electrophoresis. The fragment was gel-purified and ligated into the pCR2.1 vector (Invitrogen, Carlsbad, CA) following the manufacturer's recommendation. Twelve clones per PCR reaction were picked and sequenced. The inserts were sequenced using vector-specific M13 Forward and M13 Reverse primers and the following gene-specific primers:
SEQ ID NO: 106: SF1: CCACCTTCATGAGTGACCACG SEQ ID NO:107: SF2: ACTGTGCAGGTGCAGGTGGCAGGTAAG SEQ ID NO: 108: SRI: GAAGGTGGTCCTTCCTCTGTACT SEQ ID NO: 109: SR2: CGCCGAACTTTACACCATCCT The insert assemblies 174124888, 174124900, and 174124912 were all found to encode an open reading frame between residues 32 to 236 of the target sequence of CG50329- 01. All of the assemblies have an 3 amino acid deletion as compared to the original sequence. 174124888 and 174124912 also differ from the original sequence at 3 nucleotide positions and 2 amino acid positions. 174124900 also differs from the original sequence at 2 nucleotide positions and 1 amino acid position. OTHER EMBODIMENTS
Although particular embodiments have been disclosed herein in detail, this has been done by way of example for puφoses of illustration only, and is not intended to be limiting with respect to the scope of the appended claims, which follow. In particular, it is contemplated by the inventors that various substitutions, alterations, and modifications may be made to the invention without departing from the spirit and scope of the invention as defined by the claims. The choice of nucleic acid starting material, clone of interest, or library type is believed to be a matter of routine for a person of ordinary skill in the art with knowledge of the embodiments described herein. Other aspects, advantages, and modifications considered to be within the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. An isolated polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form;
(c) an amino acid sequence selected from the group consisting SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34; and
(d) a variant of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence.
2 The polypeptide of claim 1, wherein said polypeptide comprises the amino acid sequence of a naturally-occurring allelic variant of an amino acid sequence selected from the group consisting SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34.
3. The polypeptide of claim 2, wherein said allelic variant comprises an amino acid sequence that is the translation of a nucleic acid sequence differing by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 35.
4. The polypeptide of claim 1 , wherein the amino acid sequence of said variant comprises a conservative amino acid substitution.
5. An isolated nucleic acid molecule comprising a nucleic acid sequence encoding a polypeptide comprising an amino acid sequence selected from the group consisting of:
(a) a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(b) a variant of a mature form of an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of the amino acid residues from the amino acid sequence of said mature form;
(c) an amino acid sequence selected from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34;
(d) a variant of an amino acid sequence selected from the group consisting SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, and 34, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence;
(e) a nucleic acid fragment encoding at least a portion of a polypeptide comprising an amino acid sequence chosen from the group consisting of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, or a variant of said polypeptide, wherein one or more amino acid residues in said variant differs from the amino acid sequence of said mature form, provided that said variant differs in no more than 15% of amino acid residues from said amino acid sequence; and
(f) a nucleic acid molecule comprising the complement of (a), (b), (c), (d) or (e).
6. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises the nucleotide sequence of a naturally-occurring allelic nucleic acid variant.
7. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule encodes a polypeptide comprising the amino acid sequence of a naturally-occurring polypeptide variant.
8. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule differs by a single nucleotide from a nucleic acid sequence selected from the group consisting of SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 35.
9. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of:
(a) a nucleotide sequence selected from the group consisting of SEQ ID NOS: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 35;
(b) a nucleotide sequence differing by one or more nucleotides from a nucleotide sequence selected from the group consisting of SEQ HD NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 35, provided that no more than 20%) of the nucleotides differ from said nucleotide sequence;
(c) a nucleic acid fragment of (a); and
(d) a nucleic acid fragment of (b).
10. The nucleic acid molecule of claim 5, wherein said nucleic acid molecule hybridizes under stringent conditions to a nucleotide sequence chosen from the group consisting SEQ ID NOS:l, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, and 35, or a complement of said nucleotide sequence.
11. The nucleic acid molecule of claim 5, wherein the nucleic acid molecule comprises a nucleotide sequence selected from the group consisting of:
(a) a first nucleotide sequence comprising a coding sequence differing by one or more nucleotide sequences from a coding sequence encoding said amino acid sequence, provided that no more than 20% of the nucleotides in the coding sequence in said first nucleotide sequence differ from said coding sequence;
(b) an isolated second polynucleotide that is a complement of the first polynucleotide; and
(c) a nucleic acid fragment of (a) or (b).
12. A vector comprising the nucleic acid molecule of claim 11.
13. The vector of claim 12, further comprising a promoter operably-linked to said nucleic acid molecule.
14. A cell comprising the vector of claim 12.
15. An antibody that binds immunospecifically to the polypeptide of claim 1.
16. The antibody of claim 15, wherein said antibody is a monoclonal antibody.
17. The antibody of claim 15, wherein the antibody is a humanized antibody.
18. A method for determining the presence or amount of the polypeptide of claim 1 in a sample, the method comprising:
(a) providing the sample;
(b) contacting the sample with an antibody that binds immunospecifically to the polypeptide; and
(c) determining the presence or amount of antibody bound to said polypeptide, thereby determining the presence or amount of polypeptide in said sample.
19. A method for determining the presence or amount of the nucleic acid molecule of claim 5 in a sample, the method comprising:
(a) providing the sample;
(b) contacting the sample with a probe that binds to said nucleic acid molecule; and
(c) determining the presence or amount of the probe bound to said nucleic acid molecule, thereby determining the presence or amount of the nucleic acid molecule in said sample.
20. The method of claim 19 wherein presence or amount of the nucleic acid molecule is used as a marker for cell or tissue type.
21. The method of claim 20 wherein the cell or tissue type is cancerous.
22. A method of identifying an agent that binds to a polypeptide of claim 1, the method comprising:
(a) contacting said polypeptide with said agent; and
(b) determining whether said agent binds to said polypeptide.
23. The method of claim 22 wherein the agent is a cellular receptor or a downstream effector.
24. A method for identifying an agent that modulates the expression or activity of the polypeptide of claim 1, the method comprising:
(a) providing a cell expressing said polypeptide;
(b) contacting the cell with said agent, and
(c) determining whether the agent modulates expression or activity of said polypeptide, whereby an alteration in expression or activity of said peptide indicates said agent modulates expression or activity of said polypeptide.
25. A method for modulating the activity of the polypeptide of claim 1, the method comprising contacting a cell sample expressing the polypeptide of said claim with a compound that binds to said polypeptide in an amount sufficient to modulate the activity of the polypeptide.
26. A method of treating or preventing a NO VX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the polypeptide of claim 1 in an amount sufficient to treat or prevent said NO VX- associated disorder in said subject.
27. The method of claim 26 wherein the disorder is selected from the group consisting of cardiomyopathy and atherosclerosis.
28. The method of claim 26 wherein the disorder is related to cell signal processing and metabolic pathway modulation.
29. The method of claim 26, wherein said subject is a human.
30. A method of treating or preventing a NO VX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the nucleic acid of claim 5 in an amount sufficient to treat or prevent said NO VX- associated disorder in said subject.
31. The method of claim 30 wherein the disorder is selected from the group consisting of cardiomyopathy and atherosclerosis.
32. The method of claim 30 wherein the disorder is related to cell signal processing and metabolic pathway modulation.
33. The method of claim 30, wherein said subject is a human.
34. A method of treating or preventing a NO VX-associated disorder, said method comprising administering to a subject in which such treatment or prevention is desired the antibody of claim 15 in an amount sufficient to treat or prevent said NO VX- associated disorder in said subject.
35. The method of claim 34 wherein the disorder is diabetes.
36. The method of claim 34 wherein the disorder is related to cell signal processing and metabolic pathway modulation.
37. The method of claim 34, wherein the subject is a human.
38. A phaπnaceutical composition comprising the polypeptide of claim 1 and a pharmaceutically-acceptable carrier.
39. A phaπnaceutical composition comprising the nucleic acid molecule of claim 5 and a pharmaceutically-acceptable carrier.
40. A pharmaceutical composition comprising the antibody of claim 15 and a pharmaceutically-acceptable carrier.
41. A kit comprising in one or more containers, the pharmaceutical composition of claim 38.
42. A kit comprising in one or more containers, the pharmaceutical composition of claim 39.
43. A kit comprising in one or more containers, the pharmaceutical composition of claim 40.
44. A method for determining the presence of or predisposition to a disease associated with altered levels of the polypeptide of claim 1 in a first mammalian subject, the method comprising:
(a) measuring the level of expression of the polypeptide in a sample from the first mammalian subject; and
(b) comparing the amount of said polypeptide in the sample of step (a) to the amount of the polypeptide present in a control sample from a second mammalian subject known not to have, or not to be predisposed to, said disease; wherein an alteration in the expression level of the polypeptide in the first subject as compared to the control sample indicates the presence of or predisposition to said disease.
45. The method of claim 44 wherein the predisposition is to a cancer.
46. A method for determining the presence of or predisposition to a disease associated with altered levels of the nucleic acid molecule of claim 5 in a first mammalian subject, the method comprising:
(a) measuring the amount of the nucleic acid in a sample from the first mammalian subject; and
(b) comparing the amount of said nucleic acid in the sample of step (a) to the amount of the nucleic acid present in a control sample from a second mammalian subject known not to have or not be predisposed to, the disease; wherein an alteration in the level of the nucleic acid in the first subject as compared to the control sample indicates the presence of or predisposition to the disease.
47. The method of claim 46 wherein the predisposition is to a cancer.
48. A method of treating a pathological state in a mammal, the method comprising administering to the mammal a polypeptide in an amount that is sufficient to alleviate the pathological state, wherein the polypeptide is a polypeptide having an amino acid sequence at least 95% identical to a polypeptide comprising an amino acid sequence of at least one of SEQ ID NOS:2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, and 34, or a biologically active fragment thereof.
49. A method of treating a pathological state in a mammal, the method comprising administering to the mammal the antibody of claim 15 in an amount sufficient to alleviate the pathological state.
PCT/US2001/050331 2000-12-19 2001-12-19 Polypetides and nucleic acids encoding same WO2002057453A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2002558505A JP2005508601A (en) 2000-12-19 2001-12-19 Polypeptide and nucleic acid encoding it
EP01993342A EP1352065A2 (en) 2000-12-19 2001-12-19 Polypetides and nucleic acids encoding same
CA002433313A CA2433313A1 (en) 2000-12-19 2001-12-19 Polypetides and nucleic acids encoding same

Applications Claiming Priority (18)

Application Number Priority Date Filing Date Title
US26570400P 2000-12-19 2000-12-19
US60/256,704 2000-12-19
US25731400P 2000-12-20 2000-12-20
US60/257,314 2000-12-20
US28815301P 2001-05-02 2001-05-02
US60/288,153 2001-05-02
US29407501P 2001-05-29 2001-05-29
US60/294,075 2001-05-29
US30750601P 2001-07-24 2001-07-24
US60/307,506 2001-07-24
US31159001P 2001-08-10 2001-08-10
US31161301P 2001-08-10 2001-08-10
US60/311,613 2001-08-10
US60/311,590 2001-08-10
US31561701P 2001-08-29 2001-08-29
US60/315,617 2001-08-29
US32235801P 2001-09-14 2001-09-14
US60/322,358 2001-09-14

Publications (2)

Publication Number Publication Date
WO2002057453A2 true WO2002057453A2 (en) 2002-07-25
WO2002057453A3 WO2002057453A3 (en) 2003-08-14

Family

ID=27578753

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/050331 WO2002057453A2 (en) 2000-12-19 2001-12-19 Polypetides and nucleic acids encoding same

Country Status (3)

Country Link
JP (1) JP2005508601A (en)
CA (1) CA2433313A1 (en)
WO (1) WO2002057453A2 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003038094A1 (en) * 2001-10-30 2003-05-08 Bayer Healthcare Ag Regulation of human glucose transporter
WO2003054190A1 (en) * 2001-12-21 2003-07-03 Takeda Chemical Industries, Ltd. Novel proteins and dnas thereof
WO2002055704A3 (en) * 2001-01-09 2003-10-30 Curagen Corporation Proteins, polynucleotides encoding them and methods of using the same
WO2004106935A2 (en) * 2003-05-27 2004-12-09 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g protein-coupled receptor 103 (gpr103)
WO2005079840A2 (en) * 2004-02-20 2005-09-01 Develogen Aktiengesellschaft Use of secreted protein products for preventing and treating pancreatic diseases and/or obesity and/or metabolic syndrome
WO2006055704A2 (en) * 2004-11-17 2006-05-26 Curagen Corporation Antibodies directed to ten-m proteins and uses thereof
WO2006062135A1 (en) * 2004-12-07 2006-06-15 Riken Diagnostic for mental disease
US7560540B2 (en) 2000-04-28 2009-07-14 The Johns Hopkins University Nucleic acid encoding dendritic cell co-stimulatory molecules
WO2010052288A1 (en) * 2008-11-07 2010-05-14 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Teneurin and cancer
US8008332B2 (en) 2006-05-31 2011-08-30 Takeda San Diego, Inc. Substituted indazoles as glucokinase activators
US8034822B2 (en) 2006-03-08 2011-10-11 Takeda San Diego, Inc. Glucokinase activators
US8124617B2 (en) 2005-09-01 2012-02-28 Takeda San Diego, Inc. Imidazopyridine compounds
US8163779B2 (en) 2006-12-20 2012-04-24 Takeda San Diego, Inc. Glucokinase activators
US8173645B2 (en) 2007-03-21 2012-05-08 Takeda San Diego, Inc. Glucokinase activators
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001066690A2 (en) * 2000-03-06 2001-09-13 Smithkline Beecham Corporation Novel compounds
WO2001072961A2 (en) * 2000-03-24 2001-10-04 Smithkline Beecham Corporation Novel compounds
WO2002046409A2 (en) * 2000-12-06 2002-06-13 Curagen Corporation Proteins and nucleic acids encoding same

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001066690A2 (en) * 2000-03-06 2001-09-13 Smithkline Beecham Corporation Novel compounds
WO2001072961A2 (en) * 2000-03-24 2001-10-04 Smithkline Beecham Corporation Novel compounds
WO2002046409A2 (en) * 2000-12-06 2002-06-13 Curagen Corporation Proteins and nucleic acids encoding same

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL [Online] EBI; CEGP1 protein, 1 October 2000 (2000-10-01) retrieved from EMBL Database accession no. Q9NQ36 XP002223479 *
GRIMMOND SEAN ET AL: "Cloning, mapping, and expression analysis of a gene encoding a novel mammalian EGF-related protein (SCUBE1)." GENOMICS, vol. 70, no. 1, 15 November 2000 (2000-11-15), pages 74-81, XP002223477 ISSN: 0888-7543 *
GRIMMOND SEAN ET AL: "Expression of a novel mammalian epidermal growth factor-related gene during mouse neural development." MECHANISMS OF DEVELOPMENT, vol. 102, no. 1-2, April 2001 (2001-04), pages 209-211, XP002223478 ISSN: 0925-4773 *
See also references of EP1352065A2 *

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
US7560540B2 (en) 2000-04-28 2009-07-14 The Johns Hopkins University Nucleic acid encoding dendritic cell co-stimulatory molecules
US8053558B2 (en) 2000-04-28 2011-11-08 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US8053414B2 (en) 2000-04-28 2011-11-08 The Johns Hopkins University Methods of using B7-DC molecules to induce or enhance an immune response
US7122345B2 (en) 2001-01-09 2006-10-17 Curagen Corporation Nucleic acid encoding a NOVX13 polypeptide
WO2002055704A3 (en) * 2001-01-09 2003-10-30 Curagen Corporation Proteins, polynucleotides encoding them and methods of using the same
WO2003038094A1 (en) * 2001-10-30 2003-05-08 Bayer Healthcare Ag Regulation of human glucose transporter
WO2003054190A1 (en) * 2001-12-21 2003-07-03 Takeda Chemical Industries, Ltd. Novel proteins and dnas thereof
WO2004106935A2 (en) * 2003-05-27 2004-12-09 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g protein-coupled receptor 103 (gpr103)
WO2004106935A3 (en) * 2003-05-27 2008-01-17 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with g protein-coupled receptor 103 (gpr103)
WO2005079840A2 (en) * 2004-02-20 2005-09-01 Develogen Aktiengesellschaft Use of secreted protein products for preventing and treating pancreatic diseases and/or obesity and/or metabolic syndrome
WO2005079840A3 (en) * 2004-02-20 2006-10-26 Develogen Ag Use of secreted protein products for preventing and treating pancreatic diseases and/or obesity and/or metabolic syndrome
US9803015B2 (en) 2004-10-06 2017-10-31 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US11242387B2 (en) 2004-10-06 2022-02-08 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US11939378B2 (en) 2004-10-06 2024-03-26 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
WO2006055704A2 (en) * 2004-11-17 2006-05-26 Curagen Corporation Antibodies directed to ten-m proteins and uses thereof
WO2006055704A3 (en) * 2004-11-17 2006-10-12 Curagen Corp Antibodies directed to ten-m proteins and uses thereof
WO2006062135A1 (en) * 2004-12-07 2006-06-15 Riken Diagnostic for mental disease
US8124617B2 (en) 2005-09-01 2012-02-28 Takeda San Diego, Inc. Imidazopyridine compounds
US8034822B2 (en) 2006-03-08 2011-10-11 Takeda San Diego, Inc. Glucokinase activators
US8394843B2 (en) 2006-05-31 2013-03-12 Takeda California, Inc. Substituted isoindoles as glucokinase activators
US8008332B2 (en) 2006-05-31 2011-08-30 Takeda San Diego, Inc. Substituted indazoles as glucokinase activators
US8163779B2 (en) 2006-12-20 2012-04-24 Takeda San Diego, Inc. Glucokinase activators
US8173645B2 (en) 2007-03-21 2012-05-08 Takeda San Diego, Inc. Glucokinase activators
WO2010052288A1 (en) * 2008-11-07 2010-05-14 Novartis Forschungsstiftung, Zweigniederlassung, Friedrich Miescher Institute For Biomedical Research Teneurin and cancer
US8642280B2 (en) 2008-11-07 2014-02-04 Novartis Forschungsstiftung Zweigniederlassung Friedrich Miescher Institute For Biomedical Research Teneurin and cancer
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1

Also Published As

Publication number Publication date
WO2002057453A3 (en) 2003-08-14
JP2005508601A (en) 2005-04-07
CA2433313A1 (en) 2002-07-25

Similar Documents

Publication Publication Date Title
US20060063200A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20050287564A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
WO2002057453A2 (en) Polypetides and nucleic acids encoding same
US20040029222A1 (en) Proteins and nucleic acids encoding same
US7109000B2 (en) Proteins and nucleic acids encoding same
US20040005558A1 (en) Proteins, polynucleotides ecoding them and methods of using the same
WO2002046229A2 (en) Novel proteins and nucleic acids encoding same
CA2448073A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
US20030236389A1 (en) Proteins, polynucleotides encoding them and methods of using the same
US20030236188A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
CA2448540A1 (en) Therapeutic polypeptides, nucleic acids encoding same, and methods of use
CA2446437A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
US20030203843A1 (en) Proteins and nucleic acids encoding same
US20040018594A1 (en) Novel antibodies that bind to antigenic polypeptides, nucleic acids encoding the antigens, and methods of use
US20040018970A1 (en) Novel nucleic acids and polypeptides and methods of use thereof
WO2002040539A2 (en) Gpcr-like protein and nucleic acids encoding same
US20060084086A1 (en) Proteins, polynucleotides encoding them and methods of using the same
US20030203363A1 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
WO2002064793A2 (en) G protein-coupled receptor-like proteins and nucleics acids encoding same
WO2002081629A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same
US20030165858A1 (en) Novel GPCR-like proteins and nucleic acids encoding same
EP1352065A2 (en) Polypetides and nucleic acids encoding same
AU2002245183A1 (en) Polypetides and nucleic acids encoding same
US20040033971A1 (en) Polypeptides and nucleic acids encoding same
WO2002072770A2 (en) Novel human proteins, polynucleotides encoding them and methods of using the same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US US US US US US US US US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2433313

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2002558505

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2001993342

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2002245183

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2001993342

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWW Wipo information: withdrawn in national office

Ref document number: 2001993342

Country of ref document: EP