US20040006003A1 - Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation - Google Patents

Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation Download PDF

Info

Publication number
US20040006003A1
US20040006003A1 US10/360,274 US36027403A US2004006003A1 US 20040006003 A1 US20040006003 A1 US 20040006003A1 US 36027403 A US36027403 A US 36027403A US 2004006003 A1 US2004006003 A1 US 2004006003A1
Authority
US
United States
Prior art keywords
group
tyr
ala
phe
ile
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/360,274
Inventor
Kathleen Rodgers
Gere DiZerega
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Southern California USC
Original Assignee
University of Southern California USC
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 University of Southern California USC filed Critical University of Southern California USC
Priority to US10/360,274 priority Critical patent/US20040006003A1/en
Publication of US20040006003A1 publication Critical patent/US20040006003A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • A61K38/085Angiotensins
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/14Angiotensins: Related peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0645Macrophages, e.g. Kuepfer cells in the liver; Monocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0663Bone marrow mesenchymal stem cells (BM-MSC)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/32Angiotensins [AT], angiotensinogen
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells

Definitions

  • This present invention relates to methods and kits for use in accelerating the proliferation and differentiation of hematopoietic and mesenchymal cells.
  • Bone marrow contains pluripotent stem cells that are capable of reconstituting either the hematopoietic system or a wide range of mesenchymal tissues.
  • the mechanisms by which hematopoietic and mesenchymal stem cells produce a range of lineage-specific cell types are quite dissimilar.
  • the hematopoietic system is composed of a multitude of cell generations ranging from the terminally differentiated to very primitive hematopoietic lineage-specific cells, including a multipotent, self-renewing hematopoietic stem cell with long-term repopulating capability (HPC).
  • HPC are pluripotent lineage-specific cells that possess the ability to terminally differentiate into hematopoietic lineage-specific cells (HLSC).
  • HPC Hematopoiesis is an ongoing process, and therefore HPC must provide a continuous source of HLSC, which in turn can differentiate into red cells, platelets, monocytes, granulocytes and lymphocytes. (Prockop, Science 276:71-74, 1997). HPC proliferate either by “self-renewal”, to produce HPC-type progeny cells, or with accompanying differentiation, to produce HLSC. (Traycoff, et al., supra).
  • HPC transplantation therapy has been successful for a variety of malignant and inherited diseases and also provides myelopoietic support for patients undergoing high-dose chemotherapy or radiotherapy.
  • stem cell transplantation has been limited by several features. First, acquiring a sufficient quantity of stem cells to achieve benefit after transfusion requires either extensive, operative bone marrow harvests or extensive pheresis procedures. (Emerson, supra). Next, even under these circumstances, only a limited number of useful cells is obtained. Finally, mature: blood cell regeneration after transfusion is slow, so that little direct therapeutic benefit is seen for periods of 1 to 3 weeks. (Emerson, supra).
  • ex vivo expansion may render inadequate HPC populations in peripheral blood and umbilical cord blood sufficient for autologous transplantation and adult allogeneic transplantation respectively.
  • ex vivo expansion of HPC will greatly increase their utility as gene therapy vehicles.
  • ex vivo expansion of HLSC promises substantial clinical benefits, such as re-infusion of expanded populations of myeloid precursor cells to reduce the period of obligate neutropenia following autologous transplantation, the generation of natural killer cells for use in adoptive immunotherapy protocols, generation of megakaryocyte precursors to alleviate post-transplant-associated thrombocytopenia and more efficient generation of delivery systems for gene therapy. (Alcorn and Holyoake, supra).
  • G-CSF granulocyte colony-stimulating factor
  • GM-CSF granulocyte/macrophage colony stimulating factor
  • SCF stem cell factor
  • M-CSF macrophage colony-stimulating factor
  • interleukins 1, 3, 6, and 11 Reviewed in Takaku, J. Cancer Res. Clin. Oncol. 121:701-709, 1995; Holyoake, et al., supra.
  • MIP-1 ⁇ macrophage inhibitory protein-1 ⁇
  • TGF- ⁇ tumor necrosis factor ⁇
  • TGF ⁇ transforming growth factor ⁇
  • MSC Mesenchymal stem cells
  • mesenchymal stem cells are pluripotent progenitor cells that possess the ability to differentiate into a variety of mesenchymal tissue, including bone, cartilage, tendon, muscle, marrow stroma, fat and dermis as demonstrated in a number of organisms, including humans (Bruder, et al., J. Cellul. Biochem. 56:283-294 (1994).
  • the formation of mesenchymal tissues is known as the mesengenic process, which continues throughout life, but proceeds much more slowly in the adult than in the embryo (Caplan, Clinics in Plastic Surgery 21:429-435 (1994).
  • the mesengenic process in the adult is a repair process but involves the same cellular events that occur during embryonic development (Reviewed in Caplan, 1994, supra).
  • chemoattraction brings MSC to the site of repair where they proliferate into a mass of cells that spans the break. These cells then undergo commitment and enter into a specific lineage pathway (differentiation), where they remain capable of proliferating.
  • the cells in the different pathways terminally differentiate (and are no longer able to proliferate) and combine to form the appropriate skeletal tissue, in a process controlled by the local concentration of tissue-specific cytokines and growth factors (Caplan, 1994, supra).
  • MSC therapy can serve as a means to deliver high densities of repair-competent cells to a defect site when adequate numbers of MSC and MSC lineage-specific cells are not present in vivo, especially in older and/or diseased patients.
  • methods for rapidly producing large numbers of MSC are necessary.
  • MSC have been exposed to a number of growth factors in vitro, but only platelet-derived growth factor (PDGF) showed mitotic activity (Caplan et al., 1994, supra). Methods that increase the ex vivo proliferation rate of MSC will greatly increase the utility of MSC therapy. Similarly, methods that increase in vivo proliferation rate of MSC will enhance the utility of MSC therapy by rapidly increasing local concentrations of MSC at the repair site.
  • PDGF platelet-derived growth factor
  • the present invention provides methods that promote hematopoietic stem and lineage-specific cell proliferation and differentiation, and mesenchymal stem and lineage-specific cell proliferation by contacting the cells with angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof or AII AT 2 type 2 receptor agonists, either alone or in combination with other growth factors and cytokines.
  • AI angiotensinogen
  • AI AI analogues
  • AI fragments and analogues thereof angiotensin II
  • AII AT 2 type 2 receptor agonists either alone or in combination with other growth factors and cytokines.
  • an improved cell culture medium for the proliferation and differentiation of hematopoietic stem and lineage-specific cells, and for the proliferation of mesenchymal stem and lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments or analogues thereof or AII AT 2 type 2 receptor agonists.
  • kits for the propagation of hematopoietic and mesenchymal stem and lineage-specific cells comprising cell an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments or analogues thereof or AII AT 2 type 2 receptor agonists, and instructions for using the amount effective of active agent as a cell culture medium supplement.
  • the kit further comprises a cell culture media.
  • the kit further comprises a sterile container for cell culturing.
  • HPC refers to any hematopoietic pluripotent progenitor cells capable of giving rise to a wide variety of differentiated hematopoietic cell types.
  • Cell types from this definition include, but are not limited to CD34 + bone marrow mononuclear cells (BMMC) (Berardi, et al., Blood 86:2123-2129, 1995), PBSC (Fritsch, et al., Bone Marrow transplantation 17:169-178, 1996), cobblestone area forming cells (CAFC) (Lemieux, et al., Blood 86:1339-1347, 1995) and 5-FU BM cells (Alcorn and Holyoake, Blood Reviews 10:167-176, 1996).
  • BMMC bone marrow mononuclear cells
  • CAFC cobblestone area forming cells
  • 5-FU BM cells Alcorn and Holyoake, Blood Reviews 10:167-176, 1996.
  • HLSC refers to hematopoietic lineage-specific cells, and includes the progeny of HPC that are committed to a cell-specific differentiation pathway, as well as fully differentiated hematopoietic cells.
  • MSC mesenchymal stem cells
  • proliferation encompasses both cell self renewal and cellular proliferation with accompanying differentiation.
  • differentiation includes both entry into a specific lineage pathway and functional activation of differentiated cells.
  • U.S. Pat. No. 5,015,629 to DiZerega describes a method for increasing the rate of healing of wound tissue, comprising the application to such tissue of angiotensin II (AII) in an amount which is sufficient for said increase.
  • AII angiotensin II
  • the application of AII to wound tissue significantly increases the rate of wound healing, leading to a more rapid re-epithelialization and tissue repair.
  • AII refers to an octapeptide present in humans and other species having the sequence Asp-Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:1].
  • angiotensin The biological formation of angiotensin is initiated by the action of renin on the plasma substrate angiotensinogen (Clouston et al., Genomics 2:240-248 (1988); Kageyama et al, Biochemistry 23:3603-3609; Ohkubo et al., Proc. Natl. Acad. Sci. 80:2196-2200 (1983); each reference hereby incorporated in its entirety).
  • the substance so formed is a decapeptide called angiotensin I (AI) which is converted to AII by the converting enzyme angiotensinase which removes the C-terminal His-Leu residues from AI [SEQ ID NO:38].
  • AII is a known pressor agent and is commercially available.
  • AII was shown to be angiogenic in rabbit corneal eye and chick chorioallantoic membrane models (Fernandez, et al., J. Lab. Clin. Med. 105:141 (1985); LeNoble, et al., Eur. J. Pharmacol. 195:305-6 (1991).
  • AII may accelerate wound repair through increased neovascularization, growth factor release, reepithelialization and/or production of extracellular matrix.
  • angiotensinogen AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues and fragments thereof, or AII receptor agonists would be useful in accelerating the proliferation and differentiation of hematopoietic and mesenchymal stem and lineage-specific cells.
  • a peptide agonist selective for the AT2 receptor (AII has 100 times higher affinity for AT2 than AT1) has been identified.
  • This peptide is p-amninophenylalanine6-AII [“p-NH 2 -Phe)6-AII)”], Asp-Arg-Val-Tyr-Ile-Xaa-Pro-Phe [SEQ ID NO.36] wherein Xaa is p-NH 2 -Phe (Speth and Kim, BBRC 169:997-1006 (1990).
  • This peptide gave binding characteristics comparable to AT2 antagonists in the experimental models tested (Catalioto, et al., Eur. J. Pharmacol. 256:93-97 (1994); Bryson, et al., Eur. J. Pharmacol. 225:119-127 (1992).
  • AII receptor and AII receptor antagonists have been examined in two experimental models of vascular injury and repair which suggest that both AII receptor subtypes (AT1 and AT2) play a role in wound healing (Janiak et al., Hypertension 20:737-45 (1992); Prescott, et al., Am. J. Pathol. 139:1291-1296 (1991); Kauffman, et al., Life Sci. 49:223-228 (1991); Viswanathan, et al., Peptides 13:783-786 (1992); Kimura, et al., BBRC 187:1083-1090 (1992).
  • AII(1-7) AII residues 1-7) or other fragments of AII to evaluate their activity.
  • AII(1-7) elicits some, but not the full range of effects elicited by AII.
  • Pfeilschifter, et al. Eur. J. Pharmacol. 225:57-62 (1992); Jaiswal, et al., Hypertension 19(Supp. II):II-49-II-55 (1992); Edwards and Stack, J. Pharmacol. Exper. Ther. 266:506-510 (1993); Jaiswal, et al., J. Pharmacol. Exper. Ther. 265:664-673 (1991); Jaiswal, et al., Hypertension 17:1115-1120 (1-991); Portsi, et a, Br. J. Pharmacol. 111:652-654 (1994).
  • a preferred class of AT2 agonists for use in accordance with the present invention comprises AII, AII analogues or active fragments thereof having p-NH-Phe in a position corresponding to a position 6 of AII.
  • various nonpeptidic agents e.g., peptidomimetics
  • having the requisite AT2 agonist activity are further contemplated for use in accordance with the present invention.
  • the active AII analogues, fragments of AII and analogues thereof of particular interest in accordance with the present invention comprise a sequence consisting of at least three contiguous amino acids of groups R 1 -R 8 in the sequence of general
  • X is H or a one to three peptide group and a peptide bond between R A and R B is labile to aminopeptidase A cleavage;
  • R 3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
  • R 4 is selected from the group consisting of Tyr, Tyr(PO 3 ) 2 , Thr, Ser, homoSer and azaTyr;
  • R 5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
  • R 6 is His, Arg or 6-NH 2 -Phe
  • R 7 is Pro or Ala
  • R 8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R 4 as a terminal Tyr group.
  • Compounds falling within the category of AT2 agonists useful in the practice of the invention include the AII analogues set forth above subject to the restriction that R 6 is p-NH 2 -Phe.
  • R A is suitably selected from Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me 2 Gly, Pro, Bet, Glu(NH 2 ), Gly, Asp(NH 2 ) and Suc.
  • R B is suitably selected, from Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys. Particularly preferred combinations for R A and R B are Asp-Arg, Asp-Lys, Glu-Arg and Glu-Lys.
  • Particularly preferred embodiments of this class include the following: AII, AIII or AII(2-8), Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:2]; AII(3-8), also known as des1-AIII or AIV, Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:3]; AII(1-7), Asp-Arg-Val-Tyr-Ile-His-Pro ⁇ SEQ ID NO:4]; AII(2-7).
  • Arg-norLeu-Tyr-Ile-His-Pro-Phe [SEQ ID NO:12] and Arg-Val-Tyr-norLeu-His-Pro-Phe [SEQ ID NO:13].
  • Still another preferred embodiment encompassed within the scope of the invention is a peptide having the sequence Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe [SEQ ID NO:31].
  • AII(6-8), His-Pro-Phe [SEQ ID NO:14] and AII(4-8), Tyr-Ile-His-Pro-Phe [SEQ ID NO:15] were also tested and found not to be effective.
  • R 2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
  • R 3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
  • R 4 is selected from the group consisting of Tyr, Tyr(PO 3 ) 2 , Thr, Ser, homoSer and azaTyr;
  • R 5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
  • R 6 is His, Arg or 6-NH 2 -Phe
  • R 7 is Pro or Ala
  • R 8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
  • a particularly preferred subclass of the compounds of general formula II has the formula
  • R 2 , R 3 and R 5 are as previously defined.
  • Particularly preferred is angiotensin III of the formula Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:2].
  • Other preferred compounds include peptides having the structures Arg-Val-Tyr-Gly-His-Pro-Phe [SEQ ID NO:17] and Arg-Val-Tyr-Ala-His-Pro-Phe [SEQ ID NO:18].
  • the fragment AII(4-8) was ineffective in repeated tests; this is believed to be due to the exposed tyrosine on the N-terminus.
  • AII and its analogues adopt either a gamma or a beta turn (Regoli, et al., Pharmacological Reviews 26:69 (1974).
  • neutral side chains in position R 3 , R 5 and R 7 may be involved in maintaining the appropriate distance between active groups in positions R 4 , R 6 and R 8 primarily responsible for binding to receptors and/or intrinsic activity.
  • Hydrophobic side chains in positions R 3 , R 5 and R 8 may also play an important role in the whole conformation of the peptide and/or contribute to the formation of a hypothetical hydrophobic pocket.
  • R 2 Appropriate side chains on the amino acid in position R 2 may contribute to affinity of the compounds for target receptors and/or play an important role in the conformation of the peptide. For this reason, Arg and Lys are particularly preferred as R 2 .
  • R 3 may be involved in the formation of linear or nonlinear hydrogen bonds with R 5 (in the gamma turn model) or R 6 (in the beta turn model). R 3 would also participate in the first turn in a beta antiparallel structure (which has also been proposed as a possible structure). In contrast to other positions in general formula I, it appears that beta and gamma branching are equally effective in this position. Moreover, a single hydrogen bond may be sufficient to maintain a relatively stable conformation. Accordingly, R 3 may suitably be selected from Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr.
  • R 4 is preferably selected from Tyr, Thr, Tyr (PO 3 ) 2 , homoSer, Ser and azaTyr.
  • Tyr is particularly preferred as it may form a hydrogen bond with the receptor site capable of accepting a hydrogen from the phenolic hydroxyl (Regoli, et al. (1974), supra).
  • an amino acid with a ⁇ aliphatic or alicyclic chain is particularly desirable. Therefore, while Gly is suitable in position R 5 , it is preferred that the amino acid in this position be selected from Ile, Ala, Leu, norLeu, Gly and Val.
  • R 6 is His, Arg or 6-NH 2 -Phe.
  • the unique properties of the imidazole ring of histidine e.g., ionization at physiological pH, ability to act as proton donor or acceptor, aromatic character) are believed to contribute to its particular utility as R 6 .
  • conformational models suggest that His may participate in hydrogen bond formation (in the beta model) or in the second turn of the antiparallel structure by influencing the orientation of R 7 .
  • R 7 should be Pro in order to provide the most desirable orientation of R 8 .
  • both a hydrophobic ring and an anionic carboxyl terminal appear to be particularly useful in binding of the analogues of interest to receptors; therefore, Tyr and especially Phe are preferred for purposes of the present invention.
  • Analogues of particular interest include the following: TABLE 2 Angiotensin II Analogues All Analogue Sequence Name Amino Acid Sequence Identifier Analogue 1 Asp-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 19 Analogue 2 Asn-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 20 Analogue 3 Ala-Pro-Gly-Asp-Arg-Ile-Tyr-Val-His-Pro-Phe SEQ ID NO: 21 Analogue 4 Glu-Arg-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 22 Analogue 5 Asp-Lys-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 23 Analogue 6 Asp-Arg-Ala-Tyr-Ile-His-Pro-Phe SEQ ID NO: 24 Analogue 7 Asp-Arg-Val-Thr-Ile-His-Pro-Phe SEQ ID NO:
  • polypeptides of the instant invention may be synthesized by any conventional method, including, but not limited to, those set forth in J. M. Stewart and J. D. Young, Solid Phase Peptide Synthesis, 2nd ed., Pierce Chemical Co., Rockford, m. (1984) and J. Meienhofer, Hormonal Proteins and Peptides, Vol. 2, Academic Press, New York, (1973) for solid phase synthesis and E. Schroder and K. Lubke, The Peptides, Vol. 1, Academic Press, New York, (1965) for solution synthesis.
  • the disclosures of the foregoing treatises are incorporated by reference herein.
  • these methods involve the sequential addition of protected amino acids to a growing peptide chain (U.S. Pat. No. 5,693,616, herein incorporated by reference in its entirety). Normally, either the amino or carboxyl group of the first amino acid and any reactive side chain group are protected. This protected amino acid is then either attached to an inert solid support, or utilized in solution, and the next amino acid in the sequence, also suitably protected, is added under conditions amenable to formation of the amide linkage. After all the desired amino acids have been linked in the proper sequence, protecting groups and any solid support are removed to afford the crude polypeptide. The polypeptide is desalted and purified, preferably chromatographically, to yield the final product.
  • peptides are synthesized according to standard solid-phase methodologies, such as may be performed on an Applied Biosystems Model 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.), according to manufacturer's instructions. Other methods of synthesizing peptides or peptidomimetics, either by solid phase methodologies or in liquid phase, are well known to those skilled in the art.
  • AII has been shown to increase the proliferation of a number of cell types in vitro, it does not necessarily increase the proliferation of all cell types.
  • TGF ⁇ transforming growth factor ⁇
  • Emerson has shown that inclusion of TGF- ⁇ in most expansion cultures resulted in a decreased HPC and HLSC yield, it is unexpected that AII, through the action of TGF- ⁇ , would be of benefit in such situations. No studies have reported that AII has an effect on the differentiation of either HPC or MSC.
  • a method of increasing in vitro and ex vivo HPC and HLSC proliferation by exposure to angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof, or AII AT 2 type 2 receptor agonists is disclosed.
  • HPC are isolated from bone marrow, peripheral blood or umbilical cord blood. HPC is then selected for in these samples. HPC-enriched samples are cultured under appropriate growth conditions, in the presence of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists. HPC proliferation is assessed at various time points during culture.
  • HPC and HLSC are isolated from bone marrow aspirates from the posterior iliac crest (Caplan and Haynesworth, U.S. Pat. No. 5,486,359).
  • CD34 + HPC are isolated from the aspirate by attaching a biotinylated monoclonal antibody specific for CD34 (available from Becton Dickinson, Sunnyvale, Calif., USA) to a streptavidin affinity column (Ceprate SC; CellPro, Bothell, Wash., USA) and passing the aspirate through the column, followed by appropriate column washing and stripping, according to standard techniques in the art.
  • CD34 30 HPC are suspended in culture medium and incubated in the presence of, preferably, between about 0.1 ng/ml and about 1 mg/ml angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists.
  • the cells are expanded for a period of between 8 and 21 days and cellular proliferation with accompanying differentiation is assessed via phase microscopy following standard methylcellulose colony formation assays (Berardi, et al., supra) at various points during this time period.
  • “self-renewal” of HPC is assessed periodically by reactivity to an antibody directed against CD34 + .
  • HPC that have been cultured in the presence of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists are used for autologous transplantation, to reconstitute a depleted hematopoietic system. Prior to transplantation, the cells are rinsed to remove all traces of culture fluid, resuspended in an appropriate medium and then pelleted and rinsed several times.
  • the cells are resuspended at between 0.7 ⁇ 10 6 and 50 ⁇ 10 6 cells per ml in an appropriate medium and reinfused into a subject through intravenous infusions.
  • subject peripheral blood samples are evaluated for increases in the number of HPC, HLSC, and more mature blood cells at various time points by standard flow cytometry and cell sorting techniques. (Talmadge, et al., supra).
  • a method of increasing in vitro and ex vivo MSC and lineage-specific mesenchymal cell proliferation by exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists is disclosed.
  • Experimental conditions for the isolation, purification and in vitro growth of lineage-specific mesenchymal cells, such as bone-marrow derived stromal cells, have been reported (Johnson and Dorshkind, Blood 68(6):1348-1354 (1986); hereby incorporated by reference in its entirety).
  • MSC are isolated from bone marrow aspirates from the posterior iliac crest and/or femoral head cancellous bone, purified, resuspended in appropriate growth medium, counted and diluted to an appropriate concentration to seed in tissue culture plates (Caplan and Haynesworth, U.S. Pat. No. 5,486,359).
  • Purified MSC are cultured in an appropriate growth medium and growth conditions in a humidified atmosphere. The cells are allowed sufficient time to attach to the tissue culture dish, whereupon non-attached cells are discarded.
  • Adherent cells are placed in growth medium at 37° C.
  • angiotensinogen AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists.
  • the cells are expanded for a period of between 2 and 21 days and cellular proliferation is assessed at various time points during this time period. Subsequent medium changes are performed as needed. When the primary cultures are nearly confluent, the cells are harvested for reinfusion into a subject. Cells are examined microscopically to verify the absence of contamination.
  • the cells are rinsed to remove all traces of culture fluid, resuspended in an appropriate medium and then pelleted and rinsed several times. After the final rinse, the cells are resuspended at between 0.7 ⁇ 10 6 and 50 ⁇ 10 6 cells per ml in an appropriate medium and reinfused into a subject through intravenous infusions. Subjects are evaluated for MSC proliferation in vivo by means of a repeat diagnostic bone marrow aspirate and biopsy to be compared with the original aspirate and biopsy.
  • in vivo proliferation is assessed by reactivity to an antibody directed against a protein known to be present in higher concentrations in proliferating cells than in non-proliferating cells, such as proliferating cell nuclear antigen (PCNA, or cyclin).
  • PCNA proliferating cell nuclear antigen
  • Such antibodies are commercially available from a number of sources, including Zymed Laboratories (South San Francisco, Calif., USA).
  • isolated MSC are placed into Dulbecco's medium MEM (DMEM-LG) (Gibco, Grand Island, N.Y., USA).
  • DMEM-LG Dulbecco's medium MEM
  • the cells are purified by a series of steps. Initially, the cells are pelleted and resuspended in Complete Medium. The cells are centrifuged through a 70% Percoll (Sigma Corporation, St. Louis, Mo., USA) gradient at 460 ⁇ g for 15 minutes, the top 25% of the gradients are transferred to a tube containing 30 ml of Complete Medium and centrifuged to pellet the cells, which will then be resuspended in Complete Medium, counted and diluted to seed in 100-mm plates at 50 ⁇ 10 6 nucleated cells per plate.
  • DMEM-LG Dulbecco's medium MEM
  • purified MSC are cultured in Complete Medium at 37° C. in a humidified atmosphere containing 95% air and 5% CO 2 and the cells are allowed to attach for 3 days, whereupon non-adherent cells are removed by changing the culture medium.
  • Cellular proliferation of adherent cells and the presence of normal MSC morpholgy are assessed by phase microscopy at various time points during the subsequent growth period. Subsequent medium changes are performed every four days.
  • the primary cultures are nearly confluent, the cells are detached with 0.25% trypsin containing 0.1 mM EDTA (Gibco) and either diluted and replated as second passage cells, or used for reinfusion into a subject.
  • cells are rinsed free of culture fluid using Tyrode's solution (Gibco). After the final rinse, cells are placed in Tyrode's solution and placed in an incubator at 37° C. for one hour in order to shed serum proteins. The Tyrode's solution is removed and the cells are preferably placed into TC199 medium (Gibco) supplemented with 1% serum albumin. The cells are rinsed a number of times with this medium and after the final rinse MSC are resuspended in TC199 plus 1% serum albumin. Subsequently, MSC are injected slowly intravenously over 15 minutes. Evaluation of subsequent bone marrow aspirates are conducted up to 8 weeks after injection.
  • Tyrode's solution Gibco
  • assessment of the in vivo proliferative effect is done by histochemical evaluations of various tissues.
  • in vivo proliferation of MSC and mesenchymal lineage-specific cells is assessed by reactivity to an antibody directed against a protein known to be present in higher concentrations in proliferating cells than in non-proliferating cells, such as proliferating cell nuclear antigen (PCNA, or cyclin; Zymed Laboratories).
  • PCNA proliferating cell nuclear antigen
  • angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists on HPC, HLSC differentiation are assessed by examination of changes in gene expression, phenotype, morphology, or any other method that distinguishes a cell undergoing differentiation from a lineage-specific cell.
  • macrophage differentiation to an elicited or activated state is assessed after exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists, as described above.
  • the macrophages are assessed for phagocytotic ability by any of the well known art methods, including but not limited to determination of the number of macrophages that have ingested opsonized yeast particles, and the number of yeast per macrophage ingested. (Rodgers and Xiong, Fundamental and Applied Toxicology 33:100-108 (1996)).
  • the respiratory burst activity of leukocytes is assessed after exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists as described above.
  • the leukocytes are assessed for respiratory burst activity by any method; known in the art including, but not limited to, the ability to generate hydrogen peroxide via the respiratory burst system. (Rodgers et al., International Journal of Immunopharmacology 10:111-120 (1988); Rodgers, In: Modern Methods in Immunotoxicology (ed. G. Burleson), Wiley Liss 2:67-77 (1995)).
  • Macrophage activation by the compounds of the present invention can be utilized for treating viral and microbial infections, stimulating macrophages to produce cytokines made by activated macrophages, improving macrophage presentation to T cells, and augmenting anti-tumor immunity in a mammal.
  • angiotensinogen AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or.
  • AII AT 2 type 2 receptor agonists are used to increase in vivo HPC, HLSC, MSC and mesenchymal lineage-specific cell proliferation.
  • angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists may be administered by any suitable route, including orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles.
  • parenteral as used herein includes, subcutaneous, intravenous, imtramuscular, intrasternal, intratendinous, intraspinal, intracranial, intrathoracic, infusion techniques or intraperitoneally.
  • a method of increasing in vivo HPC, HLSC, MSC and lineage-specific mesenchymal cell proliferation by exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AI fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists is disclosed, either in the presence or absence of other growth factors and cytokines.
  • growth factors and cytokines include, but are not limited to lymphokines, interleukins-1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, granulocyte colony-stimulating factor, granulocyte/macrophage colony stimulating factor, macrophage colony-stimulating factor, tumor necrosis factor, epidermal growth factor, fibroblast growth factor, platelet derived growth factor, transforming growth factor beta, and stem cell factor.
  • the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions).
  • the compounds of the invention may be applied in a variety of solutions. Suitable solutions for use in accordance with the invention are sterile, dissolve sufficient amounts of the peptide, and are not harmful for the proposed application. In this regard, the compounds of the present invention are very stable but are hydrolyzed by strong acids and bases.
  • the compounds of the present invention are soluble in organic solvents and in aqueous solutions at pH 5-8.
  • the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc.
  • angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more other agents.
  • the therapeutic agents can be formulated as separate compositions that are given at the same time or different times, or the therapeutic agents can be given as a single composition.
  • angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration.
  • the compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, acacia, gelatin, sodium alginate, polyvinylpyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration.
  • the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, carboxymethyl cellulose colloidal solutions, ethanol, corn oil, peanut oil cottonseed oil, sesame oil, tragacanth gum, and/or various buffers.
  • the carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose.
  • liquid or semi-liquid preparations suitable for penetration through the skin e.g., liniments, lotions, ointments, creams, or pastes
  • drops suitable for administration to the eye, ear, or nose e.g., liniments, lotions, ointments, creams, or pastes
  • the dosage regimen for increasing in vivo proliferation or differentiation of HPC, HLSCP, MSC and lineage-specific mesenchymal cell with angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists is based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined; routinely by a physician using standard methods.
  • Dosage levels of the order of between 0.1 ng/kg and 1 mg/kg angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists per body weight are useful for all methods of use disclosed herein.
  • the treatment regime will vary depending on the disease being treated, based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed.
  • angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists are administered to an oncology patient for up to 30 days prior to a course of radiation therapy.
  • the therapy is administered for 1 to 6 times per day at dosages as described, above.
  • hematopoietic lineage-specific cells for donation by plasmapheresis the therapy is for up to 30 days for 1 to 6 times per day.
  • the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonist is administered topically.
  • a suitable topical dose of active ingredient of angiotensinogen, AI, AI analogues; AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists is preferably between about 0.1 ng/ml and about 1 mg/ml administered twice daily.
  • the active ingredient may comprise from 0.0001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • an improved cell culture medium for the proliferation and differentiation of hematopoietic and mesenchymal stem and lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists, as described above.
  • Any cell culture media that can support the growth of hematopoietic and mesenchymal stem and lineage-specific cells can be used with the present invention.
  • Such cell culture media include, but are not limited to Basal Media Eagle, Dulbecco's Modified Eagle Medium, Iscove's Modified Dulbecco's Medium, McCoy's Medium, Minimum Essential Medium, F-10 Nutrient Mixtures, Opti-MEMO Reduced-Serum Medium, RPMI Medium, and Macrophage-SFM Medium or combinations thereof
  • the improved cell culture medium can be supplied in either a concentrated (ie: 10 ⁇ ) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophilizate.
  • the cell culture may be either chemically defined, or may contain a serum supplement.
  • Culture media is commercially available from many sources, such as GIBCO BRL (Gaithersburg, Md.) and Sigma (St. Louis, Mo.)
  • kits for the propagation of hematopoietic and mesenchymal stem and lineage-specific cells comprising an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT 2 type 2 receptor agonists, and instructions for its use as a cell culture media supplement.
  • kits further comprise cell culture growth medium.
  • Any cell culture media that can support the growth of hematopoietic and mesenchymal stem and lineage-specific cells can be used with the present invention. Examples of such cell culture media are described above.
  • the cell culture medium can be supplied in either a concentrated (ie: 10 ⁇ ) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophalizate.
  • the cell culture may be either chemically defined, or may contain a serum supplement.
  • the kit further comprises a sterile container, which can comprise either a sealed container, such as a cell culture flask, a roller bottle, or a centrifuge tube, or a non-sealed container, such as a cell culture plate or microtiter plate (Nunc; Naperville, Ill.).
  • a sterile container which can comprise either a sealed container, such as a cell culture flask, a roller bottle, or a centrifuge tube, or a non-sealed container, such as a cell culture plate or microtiter plate (Nunc; Naperville, Ill.).
  • the kit further comprises an antibiotic supplement for inclusion in the reconstituted cell growth medium.
  • antibiotic supplements include, but are not limited to actimonycin D, Fungizone®, kanamycin, neomycin, nystatin, penicillin, streptomycin, or combinations thereof (GIBCO).
  • the present invention by providing a method for enhanced proliferation of HPC and HLSC, will greatly increase the clinical benefits of HPC transplantation. This is true both for increased “self-renewal”, which will provide a larger supply of PC capable of reconstituting the entire hematopoietic system, and for proliferation with differentiation, which will provide a larger supply of lineage-specific cells, for more rapid reconstitution of mature, functioning blood cells.
  • methods that increase in vivo proliferation of HPC will enhance the utility of HPC transplantation therapy by rapidly increasing local concentrations of HPC (and HLSC) in the bone marrow, and thereby more rapidly producing functioning blood cells.
  • MSC and mesenchymal lineage-specific cells will greatly increase the utility of MSC therapy in the repair of skeletal tissues such as bone, cartilage, tendon and ligament. More rapid proliferation of large numbers of MSC and mesenchymal lineage-specific cells will permit more efficient delivery of high densities of these cells to a defect site and more rapid in vivo amplification in the local concentration of stem and lineage-specific cells at an appropriate repair site.
  • the method of the present invention also increases the potential utility of HPC and HLSC as vehicles for gene therapy in hematopoietic disorders, as well as MSC and mesenchymal lineage-specific cells as vehicles for gene therapy in skeletal disorders by more efficiently providing a large number of such cells for transfection, and also by providing a more efficient means to rapidly expand transfected HPC, HLSC, MSC and mesenchymal lineage-specific cells.
  • Resident peritoneal macrophages have very little phagocytic activity. Exposure of macrophages to inflammatory or activating agents will increase this macrophage function. Resident peritoneal macrophages were harvested from C57B1/6 mice or Sprague Dawley rats and resuspended at a concentration of 1 ⁇ 10 6 cells/ml. in phosphate buffered saline (PBS). Five separate 0.5 ml cell aliquots were placed on a glass coverslip in a 35 mm petri dish. Prior to incubation, either 0.5 ml of PBS, AII, or AII analogues or fragments at between 1-1000 ug/ml final concentration was added to the individual cover slips.
  • PBS phosphate buffered saline
  • the dishes containing the cover slips were then incubated at 37° C. for 4 hours, after which the cover slips were washed 3 to 6 times with PBS.
  • Opsonized yeast particles (Sigma Chemical Co.) (yeast opsonized with adult serum from the same species as that under study) were added to the cover slips and incubated for 2 hours, after which the cover slips were again washed with PBS and inverted onto a glass slide.
  • the number of macrophages that ingested yeast and the number of yeast per macrophage ingested was then determined microscopically. At least 100 macrophages per coverslip were counted.
  • Table 5 describes the AII analogues and fragments used in these studies.
  • the respiratory burst of leukocytes is one component of the mediator system used to kill bacteria.
  • the level of this respiratory burst activity in resident macrophages is low.
  • differentiation either to an elicited (inflammatory) or activated state, this functional activity is significantly elevated.
  • PBMC peripheral blood mononuclear cells
  • the murine or rat peritoneal cells were harvested by lavage with 5-15 ml of cold PBS with 0.5% bovine serum albumin.
  • the human PBMC were harvested by venipuncture from normal human volunteers and isolated from peripheral blood by Ficoll Hypaque density centrifugation. After isolation, the cells were resuspended at 1 ⁇ 10 6 cells/ml and placed at 100 ⁇ l per well into 96 well plates. The cells were incubated with various concentrations of AII or AII analogues for 4 hours at 37° C. The cells were then preloaded with a fluorescent probe for hydrogen peroxide, 2,7 dichlorofluorescein acetate, which is nonfluorescent in the absence of hydrogen peroxide.
  • Human PBMC were collected from normal volunteers and isolated via Ficoll Hypaque (Sigma Chemical, St. Louis) density centrifugation. After isolation of the buffy coat, the cells were washed 3 ⁇ to remove the Ficoll Hypaque, counted in trypan blue (0.01%) and resuspended at a concentration of 1 ⁇ 10 6 cells/ml in RPMI 1640 containing 10% fetal calf serum and antibiotics. A 100 ⁇ l aliquot of cells was added to each well. Thereafter, various concentrations (0.1 to ⁇ 1000 ⁇ g/ml final concentration) of AII in RPMI 1640 containing 10% FCS and antibiotics were added to various wells in triplicate.
  • Ficoll Hypaque Sigma Chemical, St. Louis
  • Bone marrow cells were harvested from the femur and tibia of female Sprague Dawley rats by flushing the bones with Dulbecco's Minimal Essential Medium-High Glucose (“DMEM-HG”) with a syringe and an 18 gauge needle. These cells were cultured in 24 well plates at 5 ⁇ 10 3 cells/mm 2 in DMEM-HG containing selected lots of fetal calf serum (FCS) and antibiotics (complete medium) at 37?C incubator containing 5% CO 2 in air. Twenty-four hours after the initiation of the cultures the medium and nonadherent cells were aspirated and fresh medium was added.
  • DMEM-HG Dulbecco's Minimal Essential Medium-High Glucose
  • HPC were harvested from C57B1/6 mice by immunomagnetic affinity chromatography and placed in semi-solid cultures with optimal growth medium. At various times after initiation of culture, the formation of colonies and the size of the colonies (number of cells/colony) was assessed microscopically.
  • mice Female C57B1/6 mice were purchased from Simonson and used as a source of bone marrow cells in this study.
  • the bone marrow was harvested, either from untreated mice or from mice injected with 5-fluorouracil,(5-FU) (3 mg/mouse; approximately 150 mg/kg) in the tail vein 48 hours before harvest, from the femur and tibia of mice by flushing with phosphate buffered saline (PBS), pH 7.4, containing 2% fetal bovine serum (FBS) with a 21-gauge needle.
  • PBS phosphate buffered saline
  • FBS fetal bovine serum
  • the reagents for immunomagnetic labeling were purchased from Stem Cell Technologies, Inc. (Vancouver, BC). Biotin-labeled monoclonal antibodies to the following murine lineage-specific cell surface antigens were included in a cocktail for HPC enrichment and used according to the manufacturer's instructions: CD5 (Ly-1), CD45-R (B220), CD11b (Mac-1), Myeloid Differentiation Antigen (Gr-1) and Erythroid Cells (TER 119) Ten ⁇ l of antibody cocktail was added for each of the 2 sets of bone marrow (normal and 5-FU-treated), mixed and allowed to incubate at 4° C. for 15 minutes.
  • the cells were then resuspended at 4 ⁇ 10 7 cells/ml in PBS containing 2% FBS.
  • the antibody cocktail was then washed out and 100 ⁇ l anti-biotin tetramer was added for each ml of cells.
  • the suspension was mixed and incubated at 4° C. for 15 minutes.
  • Sixty ⁇ l of magnetic colloid was then added for each ml of cells, the combination was mixed and incubated at 4° C. for 15 minutes to yield the immunomagnetically-labeled bone marrow cells.
  • a column containing a stainless steel matrix was prepared by washing the matrix with PBS followed by washing with PBS containing 2% protein.
  • the immunomagnetically-labeled bone marrow cells were loaded onto the column and unlabeled cell-containing medium (enriched HPC) was collected in the flow through fraction at a flow rate of 0.2 ml/minute. Medium was added to the top of the column so that it did not run dry until 8 to 10 ml of enriched HPC were harvested. Approximately 2% of the cells loaded onto the column were isolated-in the enriched HPC fractions.
  • the enriched HPC cell fractions were diluted into a semi-solid medium containing 0.9% methylcellulose in alpha minimal essential medium (alpha MEM), 30% fetal calf serum, 1% bovine serum albumin, 10 ⁇ 4 M 2-mercaptoethanol, 2 mM L-glutamine, and 2% conditioned medium containing colony stimulating factors.
  • alpha MEM alpha minimal essential medium
  • bovine serum albumin 1% bovine serum albumin
  • 10 ⁇ 4 M 2-mercaptoethanol 10 ⁇ 4 M 2-mercaptoethanol
  • 2 mM L-glutamine 2% conditioned medium containing colony stimulating factors.
  • the conditioned medium was supernatant from splenocyte cultures (1 ⁇ 10 6 cells/ml) incubated for 48 hours in RPMI 1640 containing 10 ⁇ g/ml pokeweed mitogen (PWM), 10% FCS, and antibiotics.
  • PWM pokeweed mitogen
  • FIGS. 12 - 14 and 16 represent the number of colonies containing more than a certain number of cells/colony as a function of the duration and concentration of AII exposure (FIGS. 12 - 14 for normal cells; FIG. 16 for 5-FU treated cells.)
  • FIGS. 15 and 17 represents the number of cells per colony seen after incubation of bone marrow from normal or 5-FU treated mice with various concentrations of AII as a function of time. The results clearly demonstrate that HPC colony size increases proportionately with exposure to increased concentrations of AII, and thus that AII increases HPC proliferation.
  • Bone marrow cells were harvested from the femur and tibia of female Sprague Dawley rats by flushing the bones with Dulbecco's Minimal Essential Medium-High Glucose (“DMEM-HG”) with a syringe and an 18 gauge needle. These cells were cultured in 24 well plates at 5 ⁇ 10 cells/mm 2 in DMEM-HG containing selected lots of fetal calf serum (FCS) and antibiotics (complete medium) at 37° C. incubator containing 5% CO 2 in air.
  • FCS fetal calf serum
  • Mesenchymal stem cells isolated from bone marrow and grown under appropriate conditions can express characteristics of multiple cell types, including cells involved in the aspiration of bone, cartilage, muscle and tendons.
  • Osteogenic cells cells that can form bone tissue express the enzyme alkaline phosphatase when cultured in medium that drives them toward their osteogenic differentiation.
  • Bone marrow from female Sprague Dawley rats were harvested by flushing the femur with medium.
  • the cells were placed in culture dishes 9 cm 2 in diameter, allowed to adhere overnight, and then placed in DMEM-LG medium containing antibiotics and 10% fetal calf serum together with varying concentrations of AII.
  • the cells were washed with Tyrode's buffer and placed in osteogenic medium (DMEM-LG containing 10% fetal calf serum, 100 nM dexamethasone and 0.05 mM ascorbic acid) for 4 days prior to assessment of the level of alkaline phosphatase activity per well.
  • alkaline phosphatase substrate solution 50 mM glycine, pH 10.5, containing 1 mM magnesium chloride and 2.5 mM p-nitrophenyl phosphate
  • the buffer was removed from the culture and mixed with 1 ml of 1N sodium hydroxide to stop the reaction.
  • the absorbance of the resultant mixture at 405 nm was then determined via spectroscopy.
  • the level of alkaline phosphatase activity is expressed as the level of absorbance per culture dish.
  • HPC were harvested from C57B1/6 mice by immunomagnetic affinity chromatography and placed in semi-solid cultures with optimal growth medium in the absence of colony stimulating factors. At various times after initiation of culture, the formation of colonies and the size of the colonies. (number of cells/colony) was assessed microscopically.
  • mice Female C57B1/6 mice were purchased from Simonson and used as a source of bone marrow cells in this study.
  • the bone marrow was harvested from the femur and tibia of mice by flushing with phosphate buffered saline (PBS), pH 7.4, containing 2% fetal bovine serum (FBS) with a 21-gauge needle.
  • PBS phosphate buffered saline
  • FBS fetal bovine serum
  • the reagents for immunomagnetic labeling were purchased from Stem Cell Technologies, Inc. (Vancouver, BC). Biotin-labeled monoclonal antibodies to the following murine lineage-specific cell surface antigens were included in a cocktail for HPC enrichment and used according to the manufacturer's instructions: CD5 (Ly- 1 ), CD45-R (B220), CD11b (Mac-1), Myeloid Differentiation Antigen (Gr-1) and Erythroid Cells (TER 119) Ten ⁇ l of antibody cocktail was added to the bone marrow, mixed and allowed to incubate at 4° C. for 15 minutes.
  • the cells were then resuspended at 4 ⁇ 10 7 cells/ml in PBS containing 2% FBS.
  • the antibody cocktail was then washed out and 100 ⁇ l anti-biotin tetramer was added for each ml of cells.
  • the suspension was mixed and incubated at 4° C. for 15 minutes.
  • Sixty ill of magnetic colloid was then added for each ml of cells, the combination was mixed and incubated at 4° C. for 15 minutes to yield the immunomagnetically-labeled bone marrow cells.
  • a column containing a stainless steel matrix was prepared by washing the matrix with PBS followed by washing with PBS containing 2% protein.
  • the immunomagnetically-labeled bone marrow cells were loaded onto the column and unlabeled cell-containing medium (enriched HPC) was collected in the flow through fraction at a flow rate of 0.2 ml/minute. Medium was added to the top of the column so that it did not run dry until 8 to 10 ml of enriched HPC were harvested. Approximately 2% of the cells loaded onto the column were isolated in the enriched HPC fractions.
  • the enriched HPC cell fractions were diluted into a semi-solid medium containing 0.9% methylcellulose in alpha minimal essential medium (alpha MEM), 30% fetal calf serum, 1% bovine serum albumin, 10 ⁇ 4 M 2-mercaptoethanol, 2 mM L-glutamine, and 2% conditioned medium.
  • alpha MEM alpha minimal essential medium
  • bovine serum albumin 1% bovine serum albumin
  • 10 ⁇ 4 M 2-mercaptoethanol 10 ⁇ 4 M 2-mercaptoethanol
  • 2 mM L-glutamine 2% conditioned medium.
  • the conditioned medium was supernatant from splenocyte cultures (1 ⁇ 10 6 cells/ml) incubated for 48 hours in RPMI 1640 containing 10 ⁇ g/ml pokeweed mitogen PWM), 10% FCS, and antibiotics.

Abstract

The present invention provides methods, improved cell culture medium and kits for promoting hematopoietic and mesenchymal stem and lineage-specific cell proliferation and differentiation by growth in the presence of angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof or AII AT2 type 2 receptor agonists, either alone or in combination with other growth factors and cytokines.

Description

    CROSS REFERENCE
  • This application is a Continuation-In-Part of U.S. Provisional Application No. 60/036,507, filed Jan. 28, 1997, No. 60/046,859, filed May 8, 1997; No. 60/063,684 filed Oct. 28, 1997; No. 60/063,910 filed Oct. 31, 1997; No. 60/065,612 filed Nov. 18, 1997; and No. 60/066,593 filed Nov. 26, 1997.[0001]
  • FIELD OF THE INVENTION
  • This present invention relates to methods and kits for use in accelerating the proliferation and differentiation of hematopoietic and mesenchymal cells. [0002]
  • BACKGROUND OF THE INVENTION
  • Bone marrow contains pluripotent stem cells that are capable of reconstituting either the hematopoietic system or a wide range of mesenchymal tissues. The mechanisms by which hematopoietic and mesenchymal stem cells produce a range of lineage-specific cell types are quite dissimilar. [0003]
  • a. The Hematopoietic System [0004]
  • The hematopoietic system is composed of a multitude of cell generations ranging from the terminally differentiated to very primitive hematopoietic lineage-specific cells, including a multipotent, self-renewing hematopoietic stem cell with long-term repopulating capability (HPC). (Traycoff, et al., [0005] Experimental Hematology 24:299-306, 1996). HPC are pluripotent lineage-specific cells that possess the ability to terminally differentiate into hematopoietic lineage-specific cells (HLSC). Hematopoiesis is an ongoing process, and therefore HPC must provide a continuous source of HLSC, which in turn can differentiate into red cells, platelets, monocytes, granulocytes and lymphocytes. (Prockop, Science 276:71-74, 1997). HPC proliferate either by “self-renewal”, to produce HPC-type progeny cells, or with accompanying differentiation, to produce HLSC. (Traycoff, et al., supra).
  • HPC transplantation therapy has been successful for a variety of malignant and inherited diseases and also provides myelopoietic support for patients undergoing high-dose chemotherapy or radiotherapy. (Emerson, [0006] Blood 87:3082-3088, 1996). However, stem cell transplantation has been limited by several features. First, acquiring a sufficient quantity of stem cells to achieve benefit after transfusion requires either extensive, operative bone marrow harvests or extensive pheresis procedures. (Emerson, supra). Next, even under these circumstances, only a limited number of useful cells is obtained. Finally, mature: blood cell regeneration after transfusion is slow, so that little direct therapeutic benefit is seen for periods of 1 to 3 weeks. (Emerson, supra).
  • The development of in vitro culture techniques for hematopoietic cells combined with technologies for isolating relatively pure populations of HPC and HLSC has made possible their ex vivo expansion. (Alcorn and Holyoake, [0007] Blood Reviews 10:167-176, 1996, which is incorporated by reference herein). Successful ex vivo expansion of HPC, both by self-renewal and proliferation with differentiation, promises many clinical benefits, such as reduction of the number and duration of leucapheresis procedures required for autologous transplantation, thus reducing the risk of disease contamination in the apheresis products. (Alcorn and Holyoake, supra). Furthermore, ex vivo expansion may render inadequate HPC populations in peripheral blood and umbilical cord blood sufficient for autologous transplantation and adult allogeneic transplantation respectively. Finally, ex vivo expansion of HPC will greatly increase their utility as gene therapy vehicles. (Alcorn and Holyoake, supra). Similarly, ex vivo expansion of HLSC promises substantial clinical benefits, such as re-infusion of expanded populations of myeloid precursor cells to reduce the period of obligate neutropenia following autologous transplantation, the generation of natural killer cells for use in adoptive immunotherapy protocols, generation of megakaryocyte precursors to alleviate post-transplant-associated thrombocytopenia and more efficient generation of delivery systems for gene therapy. (Alcorn and Holyoake, supra).
  • Human bone marrow, umbilical cord blood, and peripheral blood lineage-specific cells mobilized by chemotherapy and/or cytokine treatment have been shown to be effective sources of HPC for transplantation following the administration of high-dose therapy to treat malignancy. (Holyoake, et al., [0008] Blood 87:4589-4595, 1996). Whatever the source of hematopoietic cells, most studies have used cultured cell populations selected on the basis of HPC-specific surface antigens, such as CD34. These cells can be readily obtained by a number of techniques. (Alcorn and Holyoake, supra). The results of several clinical trials using ex vivo expanded hematopoietic cells suggests that a fairly small number of HPC cultured ex vivo under appropriate conditions can initiate hematologic reconstitution. (Emerson, supra).
  • Survival and proliferation of HPC in ex vivo culture requires a combination of synergizing growth factors; the choice of cytokine/growth factor combination and culture system used will largely determine the fate of cells used to initiate the culture. (Alcorn and Holyoake, supra). In vivo, blood cell production is thought to be regulated locally by interactions of hematopoietic stem cells with a variety of cell-bound and secreted factors produced by adjacent bone marrow stromal cells. (Alcorn and Holyoake, supra). The addition of growth factors and cytokines to the culture medium is intended to compensate for the absence of stroma-associated activities. Growth factors and cytokines that have been shown to increase production of HPC (in various combinations) include granulocyte colony-stimulating factor (G-CSF), granulocyte/macrophage colony stimulating factor (GM-CSF), stem cell factor (SCF), macrophage colony-stimulating factor (M-CSF), and [0009] interleukins 1, 3, 6, and 11 (Reviewed in Takaku, J. Cancer Res. Clin. Oncol. 121:701-709, 1995; Holyoake, et al., supra). Conversely, inclusion of macrophage inhibitory protein-1α (MIP-1α), tumor necrosis factor α (TNF-α) or transforming growth factor β (TGFβ) in most expansion cultures reported to date results in decreased HPC and HLSC yields. (Emerson, supra).
  • A great deal of effort has gone into defining the optimal conditions for ex vivo culture of hematopoietic cells. Improved methods that increase of ex vivo proliferation rate of HPC will greatly increase the clinical benefits of HPC transplantation. This is true both for increased “self-renewal”, which will provide a larger supply of HPC capable of reconstituting the entire hematopoietic system, and for proliferation with differentiation, which will provide a larger supply of lineage-specific cells. Similarly, methods that increase in vivo proliferation of HPC will enhance the utility of HPC transplantation therapy by rapidly increasing local concentrations of HPC (and HLSC) in the bone marrow. Furthermore, methods that result in the differentiation of HPC and HLSC are useful in providing populations of specific cell types for use in cell therapy. [0010]
  • Transfection of mammalian HPC has been accomplished. (Larochelle, et al., [0011] Nature Medicine 2:1329-1337, 1996). Thus, methods that increase the proliferation of HBC and HLSC are also useful in rapidly providing a large population of transfected cells for use in gene therapy.
  • b. Mesenchymal Stem Cells [0012]
  • Mesenchymal stem cells (MSC) are pluripotent progenitor cells that possess the ability to differentiate into a variety of mesenchymal tissue, including bone, cartilage, tendon, muscle, marrow stroma, fat and dermis as demonstrated in a number of organisms, including humans (Bruder, et al., [0013] J. Cellul. Biochem. 56:283-294 (1994). The formation of mesenchymal tissues is known as the mesengenic process, which continues throughout life, but proceeds much more slowly in the adult than in the embryo (Caplan, Clinics in Plastic Surgery 21:429-435 (1994). The mesengenic process in the adult is a repair process but involves the same cellular events that occur during embryonic development (Reviewed in Caplan, 1994, supra). During repair processes, chemoattraction brings MSC to the site of repair where they proliferate into a mass of cells that spans the break. These cells then undergo commitment and enter into a specific lineage pathway (differentiation), where they remain capable of proliferating. Eventually, the cells in the different pathways terminally differentiate (and are no longer able to proliferate) and combine to form the appropriate skeletal tissue, in a process controlled by the local concentration of tissue-specific cytokines and growth factors (Caplan, 1994, supra).
  • Recently, it has been hypothesized that the limiting factor for MSC-based repair processes is the lack of adequate numbers of responsive MSC at the repair site (Caplan, 1994, supra). Thus, it has been suggested that by supplying a sufficient number of MSC to a specific tissue site the repair process can be controlled, since the repair site will supply the appropriate exposure to lineage-specific growth factors and differentiation molecules (Caplan, 1994, supra). Towards this end, several animal studies have demonstrated the feasibility of using autologous MSC for repair of various defects associated with mesenchymal tissue. (For review, see Caplan, et al., in [0014] The Anterior Cruciate Ligament: Current and Future Concepts, ed. D. W. Jackson, Raven Press, Ltd. NY pp.405-417 (1993). Recent work has demonstrated the feasibility of collection, ex vivo expansion in culture, and intravenous infusion of MSC in humans (Lazarus, et al., Bone Marrow Transplantation 16:557-564 (1995); Caplan and Haynesworth, U.S. Pat. No. 5,486,359, hereby incorporated by reference in its entirety). Further, MSC of animal origin have been transfected with retroviruses and have achieved high level gene expression both in vitro and in vivo (Allay, et al., Blood 82:477A (1993). Thus, the manipulation of MSC via such techniques seems a promising tool for reconstructive therapies and may be useful for gene therapy.
  • MSC therapy can serve as a means to deliver high densities of repair-competent cells to a defect site when adequate numbers of MSC and MSC lineage-specific cells are not present in vivo, especially in older and/or diseased patients. In order to efficiently deliver high densities of MSC to a defect site, methods for rapidly producing large numbers of MSC are necessary. MSC have been exposed to a number of growth factors in vitro, but only platelet-derived growth factor (PDGF) showed mitotic activity (Caplan et al., 1994, supra). Methods that increase the ex vivo proliferation rate of MSC will greatly increase the utility of MSC therapy. Similarly, methods that increase in vivo proliferation rate of MSC will enhance the utility of MSC therapy by rapidly increasing local concentrations of MSC at the repair site. [0015]
  • Furthermore, methods that enhance the proliferation rate of lineage-specific descendants of MSC, including but not limited to bone marrow stromal cells, osteoclasts, chondrocytes, and adipocytes, will enhance the therapeutic utility of MSC therapy by increasing the concentration of lineage-specific cell types at appropriate repair sites. [0016]
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides methods that promote hematopoietic stem and lineage-specific cell proliferation and differentiation, and mesenchymal stem and lineage-specific cell proliferation by contacting the cells with angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof or AII AT[0017] 2 type 2 receptor agonists, either alone or in combination with other growth factors and cytokines.
  • In another aspect of the present invention, an improved cell culture medium is provided for the proliferation and differentiation of hematopoietic stem and lineage-specific cells, and for the proliferation of mesenchymal stem and lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments or analogues thereof or AII AT[0018] 2 type 2 receptor agonists.
  • In a further aspect, the present invention provides kits for the propagation of hematopoietic and mesenchymal stem and lineage-specific cells, wherein the kits comprise cell an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments or analogues thereof or AII AT[0019] 2 type 2 receptor agonists, and instructions for using the amount effective of active agent as a cell culture medium supplement. In a preferred embodiment, the kit further comprises a cell culture media. In another preferred embodiment, the kit further comprises a sterile container for cell culturing.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • As defined herein, the term “HPC” refers to any hematopoietic pluripotent progenitor cells capable of giving rise to a wide variety of differentiated hematopoietic cell types. Cell types from this definition include, but are not limited to CD34[0020] + bone marrow mononuclear cells (BMMC) (Berardi, et al., Blood 86:2123-2129, 1995), PBSC (Fritsch, et al., Bone Marrow transplantation 17:169-178, 1996), cobblestone area forming cells (CAFC) (Lemieux, et al., Blood 86:1339-1347, 1995) and 5-FU BM cells (Alcorn and Holyoake, Blood Reviews 10:167-176, 1996). As defined herein, the term “HLSC” refers to hematopoietic lineage-specific cells, and includes the progeny of HPC that are committed to a cell-specific differentiation pathway, as well as fully differentiated hematopoietic cells. As defined herein, mesenchymal stem cells (MSC) are pluripotent progenitor cells that possess the ability to differentiate into a variety of mesenchymal tissue, including bone, cartilage, tendon, muscle, marrow stroma, fat and dermis, and include, but are not limited to, cells such as those described in Caplan and Haynesworth, U.S. Pat. No. 5,486,359. As defined herein, “proliferation” encompasses both cell self renewal and cellular proliferation with accompanying differentiation. As defined herein “differentiation” includes both entry into a specific lineage pathway and functional activation of differentiated cells.
  • U.S. Pat. No. 5,015,629 to DiZerega (the entire disclosure of which is hereby incorporated by reference) describes a method for increasing the rate of healing of wound tissue, comprising the application to such tissue of angiotensin II (AII) in an amount which is sufficient for said increase. The application of AII to wound tissue significantly increases the rate of wound healing, leading to a more rapid re-epithelialization and tissue repair. The term AII refers to an octapeptide present in humans and other species having the sequence Asp-Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:1]. The biological formation of angiotensin is initiated by the action of renin on the plasma substrate angiotensinogen (Clouston et al., [0021] Genomics 2:240-248 (1988); Kageyama et al, Biochemistry 23:3603-3609; Ohkubo et al., Proc. Natl. Acad. Sci. 80:2196-2200 (1983); each reference hereby incorporated in its entirety). The substance so formed is a decapeptide called angiotensin I (AI) which is converted to AII by the converting enzyme angiotensinase which removes the C-terminal His-Leu residues from AI [SEQ ID NO:38]. AII is a known pressor agent and is commercially available.
  • Studies have shown that AI increases mitogenesis and chemotaxis in cultured cells that are involved in wound repair, and also increases their release of growth factors and extracellular matrices (diZerega, U.S. Pat. No. 5,015,629; Dzau et. al., [0022] J. Mol. Cell. Cardiol. 21:S7 (Supp III) 1989; Berk et. al., Hypertension 13:305-14 (1989); Kawahara, et al., BBRC 150:52-9 (1988); Naftilan, et al., J. Clin. Invest. 83:1419-23 (1989); Taubman et al., J. Biol. Chem. 264:526-530 (1989); Nakahara, et al., BBRC 184:811-8 (1992); Stouffer and Owens, Circ. Res. 70:820 (1992); Wolf, et al., Am. J. Pathol. 140:95-107 (1992); Bell and Madri, Am. J. Pathol. 137;7-12 (1990). In addition, AII was shown to be angiogenic in rabbit corneal eye and chick chorioallantoic membrane models (Fernandez, et al., J. Lab. Clin. Med. 105:141 (1985); LeNoble, et al., Eur. J. Pharmacol. 195:305-6 (1991). Therefore, AII may accelerate wound repair through increased neovascularization, growth factor release, reepithelialization and/or production of extracellular matrix. However, it is not known in the literature as to whether angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues and fragments thereof, or AII receptor agonists would be useful in accelerating the proliferation and differentiation of hematopoietic and mesenchymal stem and lineage-specific cells.
  • A peptide agonist selective for the AT2 receptor (AII has 100 times higher affinity for AT2 than AT1) has been identified. This peptide is p-amninophenylalanine6-AII [“p-NH[0023] 2-Phe)6-AII)”], Asp-Arg-Val-Tyr-Ile-Xaa-Pro-Phe [SEQ ID NO.36] wherein Xaa is p-NH2-Phe (Speth and Kim, BBRC 169:997-1006 (1990). This peptide gave binding characteristics comparable to AT2 antagonists in the experimental models tested (Catalioto, et al., Eur. J. Pharmacol. 256:93-97 (1994); Bryson, et al., Eur. J. Pharmacol. 225:119-127 (1992).
  • The effects of AII receptor and AII receptor antagonists have been examined in two experimental models of vascular injury and repair which suggest that both AII receptor subtypes (AT1 and AT2) play a role in wound healing (Janiak et al., [0024] Hypertension 20:737-45 (1992); Prescott, et al., Am. J. Pathol. 139:1291-1296 (1991); Kauffman, et al., Life Sci. 49:223-228 (1991); Viswanathan, et al., Peptides 13:783-786 (1992); Kimura, et al., BBRC 187:1083-1090 (1992).
  • Many studies have focused upon AII(1-7) (AII residues 1-7) or other fragments of AII to evaluate their activity. AII(1-7) elicits some, but not the full range of effects elicited by AII. Pfeilschifter, et al., [0025] Eur. J. Pharmacol. 225:57-62 (1992); Jaiswal, et al., Hypertension 19(Supp. II):II-49-II-55 (1992); Edwards and Stack, J. Pharmacol. Exper. Ther. 266:506-510 (1993); Jaiswal, et al., J. Pharmacol. Exper. Ther. 265:664-673 (1991); Jaiswal, et al., Hypertension 17:1115-1120 (1-991); Portsi, et a, Br. J. Pharmacol. 111:652-654 (1994).
  • As hereinafter defined, a preferred class of AT2 agonists for use in accordance with the present invention comprises AII, AII analogues or active fragments thereof having p-NH-Phe in a position corresponding to a [0026] position 6 of AII. In addition to peptide agents, various nonpeptidic agents (e.g., peptidomimetics) having the requisite AT2 agonist activity are further contemplated for use in accordance with the present invention.
  • The active AII analogues, fragments of AII and analogues thereof of particular interest in accordance with the present invention comprise a sequence consisting of at least three contiguous amino acids of groups R[0027] 1-R8 in the sequence of general
  • formula I [0028]
  • R1—R2—R3—R4—R5—R6—R7-R8
  • in which R[0029] 1 and R2 together form a group of formula
  • X—RA—RB—,
  • wherein X is H or a one to three peptide group and a peptide bond between R[0030] A and RB is labile to aminopeptidase A cleavage;
  • R[0031] 3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
  • R[0032] 4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
  • R[0033] 5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
  • R[0034] 6 is His, Arg or 6-NH2-Phe;
  • R[0035] 7 is Pro or Ala and
  • R[0036] 8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
  • Compounds falling within the category of AT2 agonists useful in the practice of the invention include the AII analogues set forth above subject to the restriction that R[0037] 6 is p-NH2-Phe.
  • In one class of preferred embodiments, R[0038] A is suitably selected from Asp, Glu, Asn, Acpc (1-aminocyclopentane carboxylic acid), Ala, Me2Gly, Pro, Bet, Glu(NH2), Gly, Asp(NH2) and Suc. RB is suitably selected, from Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys. Particularly preferred combinations for RA and RB are Asp-Arg, Asp-Lys, Glu-Arg and Glu-Lys.
  • Particularly preferred embodiments of this class include the following: AII, AIII or AII(2-8), Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:2]; AII(3-8), also known as des1-AIII or AIV, Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:3]; AII(1-7), Asp-Arg-Val-Tyr-Ile-His-Pro {SEQ ID NO:4]; AII(2-7). Arg-Val-Tyr-Ile-His-Pro [SEQ ID NO:5]; AII(3-7), Val-Tyr-Ile-His-Pro [SEQ ID NO:6]; AII(5-8), Ile-His-Pro-Phe [SEQ ID NO:7]; AII(1-6), Asp-Arg-Val-Tyr-Ile-His [SEQ ID NO:8]; AII(1-5), Asp-Arg-Val-Tyr-Ile [SEQ ID NO:9]; AII(1-4), Asp-Arg-Val-Tyr [SEQ ID NO:10]; and AII(1-3), Asp-Arg-Val [SEQ ID NO:11]. Other preferred embodiments include: Arg-norLeu-Tyr-Ile-His-Pro-Phe [SEQ ID NO:12] and Arg-Val-Tyr-norLeu-His-Pro-Phe [SEQ ID NO:13]. Still another preferred embodiment encompassed within the scope of the invention is a peptide having the sequence Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe [SEQ ID NO:31]. AII(6-8), His-Pro-Phe [SEQ ID NO:14] and AII(4-8), Tyr-Ile-His-Pro-Phe [SEQ ID NO:15] were also tested and found not to be effective. [0039]
  • Another class of compounds of particular interest in accordance with the present invention are those of the general formula II [0040]
  • R2—R3—R4—R5—R6—R7—R8
  • in which R[0041] 2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
  • R[0042] 3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
  • R[0043] 4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
  • R[0044] 5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
  • R[0045] 6 is His, Arg or 6-NH2-Phe;
  • R[0046] 7 is Pro or Ala; and
  • R[0047] 8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
  • A particularly preferred subclass of the compounds of general formula II has the formula [0048]
  • R2—R3-Tyr-R5-His-Pro-Phe [SEQ ID NO:16]
  • wherein R[0049] 2, R3 and R5 are as previously defined. Particularly preferred is angiotensin III of the formula Arg-Val-Tyr-Ile-His-Pro-Phe [SEQ ID NO:2]. Other preferred compounds include peptides having the structures Arg-Val-Tyr-Gly-His-Pro-Phe [SEQ ID NO:17] and Arg-Val-Tyr-Ala-His-Pro-Phe [SEQ ID NO:18]. The fragment AII(4-8) was ineffective in repeated tests; this is believed to be due to the exposed tyrosine on the N-terminus.
  • In the above formulas, the standard three-letter abbreviations for amino acid residues are employed. In the absence of an indication to the contrary, the L-form of the amino acid is intended. Other residues are abbreviated as follows: [0050]
    TABLE 1
    Abbreviation for Amino Acids
    Me2Gly N,N-dimethylglycyl
    Bet 1-carboxy-N,N,N-trimethylmethanaminium hydroxide inner salt
    (betaine)
    Suc Succinyl
    Phe(Br) p-bromo-L-phenylalanyl
    azaTyr aza-α'-homo-L-tyrosyl
    Acpc 1-aminocyclopentane carboxylic acid
    Aib 2-aminoisobutyric acid
    Sar N-methylglycyl (sarcosine)
  • It has been suggested that AII and its analogues adopt either a gamma or a beta turn (Regoli, et al., [0051] Pharmacological Reviews 26:69 (1974). In general, it is believed that neutral side chains in position R3, R5 and R7 may be involved in maintaining the appropriate distance between active groups in positions R4, R6 and R8 primarily responsible for binding to receptors and/or intrinsic activity. Hydrophobic side chains in positions R3, R5 and R8 may also play an important role in the whole conformation of the peptide and/or contribute to the formation of a hypothetical hydrophobic pocket.
  • Appropriate side chains on the amino acid in position R[0052] 2 may contribute to affinity of the compounds for target receptors and/or play an important role in the conformation of the peptide. For this reason, Arg and Lys are particularly preferred as R2.
  • For purposes of the present invention, it is believed that R[0053] 3 may be involved in the formation of linear or nonlinear hydrogen bonds with R5 (in the gamma turn model) or R6 (in the beta turn model). R3 would also participate in the first turn in a beta antiparallel structure (which has also been proposed as a possible structure). In contrast to other positions in general formula I, it appears that beta and gamma branching are equally effective in this position. Moreover, a single hydrogen bond may be sufficient to maintain a relatively stable conformation. Accordingly, R3 may suitably be selected from Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr.
  • With respect to R[0054] 4, conformational analyses have suggested that the side chain in this position (as well as in R3 and R5) contribute to a hydrophobic cluster believed to be essential for occupation and stimulation of receptors. Thus, R4 is preferably selected from Tyr, Thr, Tyr (PO3)2, homoSer, Ser and azaTyr. In this position, Tyr is particularly preferred as it may form a hydrogen bond with the receptor site capable of accepting a hydrogen from the phenolic hydroxyl (Regoli, et al. (1974), supra).
  • In position R[0055] 5, an amino acid with a β aliphatic or alicyclic chain is particularly desirable. Therefore, while Gly is suitable in position R5, it is preferred that the amino acid in this position be selected from Ile, Ala, Leu, norLeu, Gly and Val.
  • In the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII analogues, fragments and analogues of fragments of particular interest in accordance with the present invention, R[0056] 6 is His, Arg or 6-NH2-Phe. The unique properties of the imidazole ring of histidine (e.g., ionization at physiological pH, ability to act as proton donor or acceptor, aromatic character) are believed to contribute to its particular utility as R6. For example, conformational models suggest that His may participate in hydrogen bond formation (in the beta model) or in the second turn of the antiparallel structure by influencing the orientation of R7. Similarly, it is presently considered that R7 should be Pro in order to provide the most desirable orientation of R8. In position R8, both a hydrophobic ring and an anionic carboxyl terminal appear to be particularly useful in binding of the analogues of interest to receptors; therefore, Tyr and especially Phe are preferred for purposes of the present invention.
  • Analogues of particular interest include the following: [0057]
    TABLE 2
    Angiotensin II Analogues
    All
    Analogue Sequence
    Name Amino Acid Sequence Identifier
    Analogue 1 Asp-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 19
    Analogue 2 Asn-Arg-Val-Tyr-Val-His-Pro-Phe SEQ ID NO: 20
    Analogue 3 Ala-Pro-Gly-Asp-Arg-Ile-Tyr-Val-His-Pro-Phe SEQ ID NO: 21
    Analogue 4 Glu-Arg-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 22
    Analogue 5 Asp-Lys-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 23
    Analogue 6 Asp-Arg-Ala-Tyr-Ile-His-Pro-Phe SEQ ID NO: 24
    Analogue 7 Asp-Arg-Val-Thr-Ile-His-Pro-Phe SEQ ID NO: 25
    Analogue 8 Asp-Arg-Val-Tyr-Leu-His-Pro-Phe SEQ ID NO: 26
    Analogue 9 Asp-Arg-Val-Tyr-Ile-Arg-Pro-Phe SEQ ID NO: 27
    Analogue 10 Asp-Arg-Val-Tyr-Ile-His-Ala-Phe SEQ ID NO: 28
    Analogue 11 Asp-Arg-Val-Tyr-Ile-His-Pro-Tyr SEQ ID NO: 29
    Analogue 12 Pro-Arg-Val-Tyr-Ile-His-Pro-Phe SEQ ID NO: 30
    Analogue 13 Asp-Arg-Pro-Tyr-Ile-His-Pro-Phe SEQ ID NO: 31
    Analogue 14 Asp-Arg-Val-Tyr(Po3)2-Ile-His-Pro-Phe SEQ ID NO: 32
    Analogue 15 Asp-Arg-norLeu-Tyr-Ile-His-Pro-Phe SEQ ID NO: 33
    Analogue 16 Asp-Arg-Val-Tyr-norLeu-His-Pro-Phe SEQ ID NO: 34
    Analogue 17 Asp-Arg-Val-homoSer-Tyr-Ile-His-Pro-Phe SEQ ID NO: 35
  • The polypeptides of the instant invention may be synthesized by any conventional method, including, but not limited to, those set forth in J. M. Stewart and J. D. Young, [0058] Solid Phase Peptide Synthesis, 2nd ed., Pierce Chemical Co., Rockford, m. (1984) and J. Meienhofer, Hormonal Proteins and Peptides, Vol. 2, Academic Press, New York, (1973) for solid phase synthesis and E. Schroder and K. Lubke, The Peptides, Vol. 1, Academic Press, New York, (1965) for solution synthesis. The disclosures of the foregoing treatises are incorporated by reference herein.
  • In general, these methods involve the sequential addition of protected amino acids to a growing peptide chain (U.S. Pat. No. 5,693,616, herein incorporated by reference in its entirety). Normally, either the amino or carboxyl group of the first amino acid and any reactive side chain group are protected. This protected amino acid is then either attached to an inert solid support, or utilized in solution, and the next amino acid in the sequence, also suitably protected, is added under conditions amenable to formation of the amide linkage. After all the desired amino acids have been linked in the proper sequence, protecting groups and any solid support are removed to afford the crude polypeptide. The polypeptide is desalted and purified, preferably chromatographically, to yield the final product. [0059]
  • Preferably, peptides are synthesized according to standard solid-phase methodologies, such as may be performed on an Applied Biosystems Model 430A peptide synthesizer (Applied Biosystems, Foster City, Calif.), according to manufacturer's instructions. Other methods of synthesizing peptides or peptidomimetics, either by solid phase methodologies or in liquid phase, are well known to those skilled in the art. [0060]
  • Although AII has been shown to increase the proliferation of a number of cell types in vitro, it does not necessarily increase the proliferation of all cell types. Studies have shown that AII accelerates cellular proliferation through the production of transforming growth factor β (TGFβ) (Gibbons et al., [0061] J. Clin. Invest. 90:456-461 (1992). Thus, since only PDGF is known to be mitogenic for MSC, an ability of AII to effect MSC proliferation would be unexpected. Furthermore, as Emerson (supra) has shown that inclusion of TGF-β in most expansion cultures resulted in a decreased HPC and HLSC yield, it is unexpected that AII, through the action of TGF-β, would be of benefit in such situations. No studies have reported that AII has an effect on the differentiation of either HPC or MSC.
  • In one aspect of the present invention, a method of increasing in vitro and ex vivo HPC and HLSC proliferation by exposure to angiotensinogen, angiotensin I (AI), AI analogues, AI fragments and analogues thereof, angiotensin II (AII), AII analogues, AII fragments or analogues thereof, or AII AT[0062] 2 type 2 receptor agonists is disclosed. Experimental conditions for the isolation, purification, ex vivo growth and in vivo mobilization of HPC and HLSC have been reported (Berardi, et al., Blood 86(6):2123-2129, 1995; Fritsch, et al., Bone Marrow Transplantation 17:169-178, 1996; LaRochelle, et al., Nature Medicine 12:1329-1337, 1996; Traycoff, et al., Experimental Hematology 24:299-306, 1996; Holyoake, et al., Blood 87:4589-4595, 1996; Lemieux, et al., Blood 86:1339-1347, 1995; Talmadge, et al., Bone Marrow Transplantation 17:101-109, 1996; Bodine, et al., Blood 88:89-97, 1996; all references hereby incorporated by reference herein.)
  • In one embodiment of the invention, HPC are isolated from bone marrow, peripheral blood or umbilical cord blood. HPC is then selected for in these samples. HPC-enriched samples are cultured under appropriate growth conditions, in the presence of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0063] 2 type 2 receptor agonists. HPC proliferation is assessed at various time points during culture.
  • In a preferred embodiment, HPC and HLSC are isolated from bone marrow aspirates from the posterior iliac crest (Caplan and Haynesworth, U.S. Pat. No. 5,486,359). CD34[0064] + HPC are isolated from the aspirate by attaching a biotinylated monoclonal antibody specific for CD34 (available from Becton Dickinson, Sunnyvale, Calif., USA) to a streptavidin affinity column (Ceprate SC; CellPro, Bothell, Wash., USA) and passing the aspirate through the column, followed by appropriate column washing and stripping, according to standard techniques in the art. CD3430 HPC are suspended in culture medium and incubated in the presence of, preferably, between about 0.1 ng/ml and about 1 mg/ml angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists., The cells are expanded for a period of between 8 and 21 days and cellular proliferation with accompanying differentiation is assessed via phase microscopy following standard methylcellulose colony formation assays (Berardi, et al., supra) at various points during this time period. Similarly, “self-renewal” of HPC is assessed periodically by reactivity to an antibody directed against CD34+.
  • In a further preferred embodiment, HPC that have been cultured in the presence of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0065] 2 type 2 receptor agonists are used for autologous transplantation, to reconstitute a depleted hematopoietic system. Prior to transplantation, the cells are rinsed to remove all traces of culture fluid, resuspended in an appropriate medium and then pelleted and rinsed several times. After the final rinse, the cells are resuspended at between 0.7×106 and 50×106 cells per ml in an appropriate medium and reinfused into a subject through intravenous infusions. Following transplantation, subject peripheral blood samples are evaluated for increases in the number of HPC, HLSC, and more mature blood cells at various time points by standard flow cytometry and cell sorting techniques. (Talmadge, et al., supra).
  • In another aspect of the present invention, a method of increasing in vitro and ex vivo MSC and lineage-specific mesenchymal cell proliferation by exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0066] 2 type 2 receptor agonists is disclosed. Experimental conditions for the isolation, purification and in vitro growth of lineage-specific mesenchymal cells, such as bone-marrow derived stromal cells, have been reported (Johnson and Dorshkind, Blood 68(6):1348-1354 (1986); hereby incorporated by reference in its entirety). Other reports describe different conditions for culturing lineage-specific mesenchymal cells in vitro (Bab, et al., J. Cell Sci. 84:139-151 (1986); Benayahu, et al., J. Cellular Physiology 140:1-7 (1989); both references hereby incorporated by reference in their entirety).
  • In one embodiment of the present invention, MSC are isolated from bone marrow aspirates from the posterior iliac crest and/or femoral head cancellous bone, purified, resuspended in appropriate growth medium, counted and diluted to an appropriate concentration to seed in tissue culture plates (Caplan and Haynesworth, U.S. Pat. No. 5,486,359). Purified MSC are cultured in an appropriate growth medium and growth conditions in a humidified atmosphere. The cells are allowed sufficient time to attach to the tissue culture dish, whereupon non-attached cells are discarded. Adherent cells are placed in growth medium at 37° C. in a humidified atmosphere in the presence of, preferably, between about 0.1 ng/ml and about 1 mg/ml angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0067] 2 type 2 receptor agonists. The cells are expanded for a period of between 2 and 21 days and cellular proliferation is assessed at various time points during this time period. Subsequent medium changes are performed as needed. When the primary cultures are nearly confluent, the cells are harvested for reinfusion into a subject. Cells are examined microscopically to verify the absence of contamination. The cells are rinsed to remove all traces of culture fluid, resuspended in an appropriate medium and then pelleted and rinsed several times. After the final rinse, the cells are resuspended at between 0.7×106 and 50×106 cells per ml in an appropriate medium and reinfused into a subject through intravenous infusions. Subjects are evaluated for MSC proliferation in vivo by means of a repeat diagnostic bone marrow aspirate and biopsy to be compared with the original aspirate and biopsy. In a preferred embodiment, in vivo proliferation is assessed by reactivity to an antibody directed against a protein known to be present in higher concentrations in proliferating cells than in non-proliferating cells, such as proliferating cell nuclear antigen (PCNA, or cyclin). Such antibodies are commercially available from a number of sources, including Zymed Laboratories (South San Francisco, Calif., USA).
  • In a preferred embodiment, isolated MSC, are placed into Dulbecco's medium MEM (DMEM-LG) (Gibco, Grand Island, N.Y., USA). The cells are purified by a series of steps. Initially, the cells are pelleted and resuspended in Complete Medium. The cells are centrifuged through a 70% Percoll (Sigma Corporation, St. Louis, Mo., USA) gradient at 460×g for 15 minutes, the top 25% of the gradients are transferred to a tube containing 30 ml of Complete Medium and centrifuged to pellet the cells, which will then be resuspended in Complete Medium, counted and diluted to seed in 100-mm plates at 50×10[0068] 6 nucleated cells per plate.
  • In a further preferred embodiment, purified MSC are cultured in Complete Medium at 37° C. in a humidified atmosphere containing 95% air and 5% CO[0069] 2 and the cells are allowed to attach for 3 days, whereupon non-adherent cells are removed by changing the culture medium. Cellular proliferation of adherent cells and the presence of normal MSC morpholgy are assessed by phase microscopy at various time points during the subsequent growth period. Subsequent medium changes are performed every four days. When the primary cultures are nearly confluent, the cells are detached with 0.25% trypsin containing 0.1 mM EDTA (Gibco) and either diluted and replated as second passage cells, or used for reinfusion into a subject. Preferably, cells are rinsed free of culture fluid using Tyrode's solution (Gibco). After the final rinse, cells are placed in Tyrode's solution and placed in an incubator at 37° C. for one hour in order to shed serum proteins. The Tyrode's solution is removed and the cells are preferably placed into TC199 medium (Gibco) supplemented with 1% serum albumin. The cells are rinsed a number of times with this medium and after the final rinse MSC are resuspended in TC199 plus 1% serum albumin. Subsequently, MSC are injected slowly intravenously over 15 minutes. Evaluation of subsequent bone marrow aspirates are conducted up to 8 weeks after injection.
  • In a preferred embodiment, assessment of the in vivo proliferative effect (of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0070] 2 type 2 receptor agonists on MSC and mesenchymal lineage-specific cells is done by histochemical evaluations of various tissues. In a preferred embodiment, in vivo proliferation of MSC and mesenchymal lineage-specific cells is assessed by reactivity to an antibody directed against a protein known to be present in higher concentrations in proliferating cells than in non-proliferating cells, such as proliferating cell nuclear antigen (PCNA, or cyclin; Zymed Laboratories).
  • In a further aspect of the present invention, the effect of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0071] 2 type 2 receptor agonists on HPC, HLSC differentiation are assessed by examination of changes in gene expression, phenotype, morphology, or any other method that distinguishes a cell undergoing differentiation from a lineage-specific cell. In a preferred embodiment, macrophage differentiation to an elicited or activated state is assessed after exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists, as described above. The macrophages are assessed for phagocytotic ability by any of the well known art methods, including but not limited to determination of the number of macrophages that have ingested opsonized yeast particles, and the number of yeast per macrophage ingested. (Rodgers and Xiong, Fundamental and Applied Toxicology 33:100-108 (1996)).
  • In another preferred embodiment, the respiratory burst activity of leukocytes is assessed after exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0072] 2 type 2 receptor agonists as described above. The leukocytes are assessed for respiratory burst activity by any method; known in the art including, but not limited to, the ability to generate hydrogen peroxide via the respiratory burst system. (Rodgers et al., International Journal of Immunopharmacology 10:111-120 (1988); Rodgers, In: Modern Methods in Immunotoxicology (ed. G. Burleson), Wiley Liss 2:67-77 (1995)).
  • Macrophage activation by the compounds of the present invention can be utilized for treating viral and microbial infections, stimulating macrophages to produce cytokines made by activated macrophages, improving macrophage presentation to T cells, and augmenting anti-tumor immunity in a mammal. [0073]
  • In another aspect of the present invention angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or. AII AT[0074] 2 type 2 receptor agonists are used to increase in vivo HPC, HLSC, MSC and mesenchymal lineage-specific cell proliferation. For use in increasing proliferation of HPC, HLSCP, MSC and mesenchymal lineage-specific cells, angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists may be administered by any suitable route, including orally, parentally, by inhalation spray, rectally, or topically in dosage unit formulations containing conventional pharmaceutically acceptable carriers, adjuvants, and vehicles. The term parenteral as used herein includes, subcutaneous, intravenous, imtramuscular, intrasternal, intratendinous, intraspinal, intracranial, intrathoracic, infusion techniques or intraperitoneally.
  • In a further embodiment of the present invention, a method of increasing in vivo HPC, HLSC, MSC and lineage-specific mesenchymal cell proliferation by exposure to angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AI fragments and analogues thereof and/or AII AT[0075] 2 type 2 receptor agonists is disclosed, either in the presence or absence of other growth factors and cytokines. Examples of such growth factors and cytokines include, but are not limited to lymphokines, interleukins-1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, granulocyte colony-stimulating factor, granulocyte/macrophage colony stimulating factor, macrophage colony-stimulating factor, tumor necrosis factor, epidermal growth factor, fibroblast growth factor, platelet derived growth factor, transforming growth factor beta, and stem cell factor.
  • The angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0076] 2 type 2 receptor agonists may be made up in a solid form (including granules, powders or suppositories) or in a liquid form (e.g., solutions, suspensions, or emulsions). The compounds of the invention may be applied in a variety of solutions. Suitable solutions for use in accordance with the invention are sterile, dissolve sufficient amounts of the peptide, and are not harmful for the proposed application. In this regard, the compounds of the present invention are very stable but are hydrolyzed by strong acids and bases. The compounds of the present invention are soluble in organic solvents and in aqueous solutions at pH 5-8.
  • The angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0077] 2 type 2 receptor agonists may be subjected to conventional pharmaceutical operations such as sterilization and/or may contain conventional adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers, buffers etc. * While angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more other agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions that are given at the same time or different times, or the therapeutic agents can be given as a single composition.
  • For administration, the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0078] 2 type 2 receptor agonists are ordinarily combined with one or more adjuvants appropriate for the indicated route of administration. The compounds may be admixed with lactose, sucrose, starch powder, cellulose esters of alkanoic acids, stearic acid, talc, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulphuric acids, acacia, gelatin, sodium alginate, polyvinylpyrrolidine, and/or polyvinyl alcohol, and tableted or encapsulated for conventional administration. Alternatively, the compounds of this invention may be dissolved in saline, water, polyethylene glycol, propylene glycol, carboxymethyl cellulose colloidal solutions, ethanol, corn oil, peanut oil cottonseed oil, sesame oil, tragacanth gum, and/or various buffers. Other adjuvants and modes of administration are well known in the pharmaceutical art. The carrier or diluent may include time delay material, such as glyceryl monostearate or glyceryl distearate alone or with a wax, or other materials well known in the art.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin (e.g., liniments, lotions, ointments, creams, or pastes) and drops suitable for administration to the eye, ear, or nose. [0079]
  • The dosage regimen for increasing in vivo proliferation or differentiation of HPC, HLSCP, MSC and lineage-specific mesenchymal cell with angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0080] 2 type 2 receptor agonists is based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined; routinely by a physician using standard methods. Dosage levels of the order of between 0.1 ng/kg and 1 mg/kg angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists per body weight are useful for all methods of use disclosed herein.
  • The treatment regime will vary depending on the disease being treated, based on a variety of factors, including the type of injury, the age, weight, sex, medical condition of the individual, the severity of the condition, the route of administration, and the particular compound employed. For example, angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0081] 2 type 2 receptor agonists are administered to an oncology patient for up to 30 days prior to a course of radiation therapy. The therapy is administered for 1 to 6 times per day at dosages as described, above. In another example, in order to mobilize, hematopoietic lineage-specific cells for donation by plasmapheresis the therapy is for up to 30 days for 1 to 6 times per day.
  • In a preferred embodiment of the present invention, the angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0082] 2 type 2 receptor agonist is administered topically. A suitable topical dose of active ingredient of angiotensinogen, AI, AI analogues; AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT2 type 2 receptor agonists is preferably between about 0.1 ng/ml and about 1 mg/ml administered twice daily. For topical administration, the active ingredient may comprise from 0.0001% to 10% w/w, e.g., from 1% to 2% by weight of the formulation, although it may comprise as much as 10% w/w, but preferably not more than 5% w/w, and more preferably from 0.1% to 1% of the formulation.
  • In another aspect of the present invention, an improved cell culture medium is provided for the proliferation and differentiation of hematopoietic and mesenchymal stem and lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0083] 2 type 2 receptor agonists, as described above. Any cell culture media that can support the growth of hematopoietic and mesenchymal stem and lineage-specific cells can be used with the present invention. Such cell culture media include, but are not limited to Basal Media Eagle, Dulbecco's Modified Eagle Medium, Iscove's Modified Dulbecco's Medium, McCoy's Medium, Minimum Essential Medium, F-10 Nutrient Mixtures, Opti-MEMO Reduced-Serum Medium, RPMI Medium, and Macrophage-SFM Medium or combinations thereof The improved cell culture medium can be supplied in either a concentrated (ie: 10×) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophilizate. The cell culture may be either chemically defined, or may contain a serum supplement. Culture media is commercially available from many sources, such as GIBCO BRL (Gaithersburg, Md.) and Sigma (St. Louis, Mo.)
  • In a further aspect, the present invention provides kits for the propagation of hematopoietic and mesenchymal stem and lineage-specific cells, wherein the kits comprise an effective amount of angiotensinogen, AI, AI analogues, AI fragments and analogues thereof, AII, AII analogues, AII fragments and analogues thereof and/or AII AT[0084] 2 type 2 receptor agonists, and instructions for its use as a cell culture media supplement.
  • In a preferred embodiment, the kits further comprise cell culture growth medium. Any cell culture media that can support the growth of hematopoietic and mesenchymal stem and lineage-specific cells can be used with the present invention. Examples of such cell culture media are described above. The cell culture medium can be supplied in either a concentrated (ie: 10×) or non-concentrated form, and may be supplied as either a liquid, a powder, or a lyophalizate. The cell culture may be either chemically defined, or may contain a serum supplement. [0085]
  • In a further preferred embodiment, the kit further comprises a sterile container, which can comprise either a sealed container, such as a cell culture flask, a roller bottle, or a centrifuge tube, or a non-sealed container, such as a cell culture plate or microtiter plate (Nunc; Naperville, Ill.). [0086]
  • In another preferred embodiment, the kit further comprises an antibiotic supplement for inclusion in the reconstituted cell growth medium. Examples of appropriate antibiotic supplements include, but are not limited to actimonycin D, Fungizone®, kanamycin, neomycin, nystatin, penicillin, streptomycin, or combinations thereof (GIBCO). [0087]
  • The present invention, by providing a method for enhanced proliferation of HPC and HLSC, will greatly increase the clinical benefits of HPC transplantation. This is true both for increased “self-renewal”, which will provide a larger supply of PC capable of reconstituting the entire hematopoietic system, and for proliferation with differentiation, which will provide a larger supply of lineage-specific cells, for more rapid reconstitution of mature, functioning blood cells. Similarly, methods that increase in vivo proliferation of HPC will enhance the utility of HPC transplantation therapy by rapidly increasing local concentrations of HPC (and HLSC) in the bone marrow, and thereby more rapidly producing functioning blood cells. [0088]
  • Similarly, methods that increase the proliferation of MSC and mesenchymal lineage-specific cells, will greatly increase the utility of MSC therapy in the repair of skeletal tissues such as bone, cartilage, tendon and ligament. More rapid proliferation of large numbers of MSC and mesenchymal lineage-specific cells will permit more efficient delivery of high densities of these cells to a defect site and more rapid in vivo amplification in the local concentration of stem and lineage-specific cells at an appropriate repair site. [0089]
  • The method of the present invention also increases the potential utility of HPC and HLSC as vehicles for gene therapy in hematopoietic disorders, as well as MSC and mesenchymal lineage-specific cells as vehicles for gene therapy in skeletal disorders by more efficiently providing a large number of such cells for transfection, and also by providing a more efficient means to rapidly expand transfected HPC, HLSC, MSC and mesenchymal lineage-specific cells. [0090]
  • The present invention may be better understood with reference to the accompanying examples that are intended for purposes of illustration only and should not be construed to limit the scope of the invention, as defined by the claims appended hereto.[0091]
  • EXAMPLE 1 Macrophage Differentiation: Phagocytosis
  • Resident peritoneal macrophages have very little phagocytic activity. Exposure of macrophages to inflammatory or activating agents will increase this macrophage function. Resident peritoneal macrophages were harvested from C57B1/6 mice or Sprague Dawley rats and resuspended at a concentration of 1×10[0092] 6 cells/ml. in phosphate buffered saline (PBS). Five separate 0.5 ml cell aliquots were placed on a glass coverslip in a 35 mm petri dish. Prior to incubation, either 0.5 ml of PBS, AII, or AII analogues or fragments at between 1-1000 ug/ml final concentration was added to the individual cover slips. The dishes containing the cover slips were then incubated at 37° C. for 4 hours, after which the cover slips were washed 3 to 6 times with PBS. Opsonized yeast particles (Sigma Chemical Co.) (yeast opsonized with adult serum from the same species as that under study) were added to the cover slips and incubated for 2 hours, after which the cover slips were again washed with PBS and inverted onto a glass slide. The number of macrophages that ingested yeast and the number of yeast per macrophage ingested was then determined microscopically. At least 100 macrophages per coverslip were counted. The data from this study are summarized in Tables 3 and 4 and FIGS. 1 and 2. Table 5 describes the AII analogues and fragments used in these studies.
  • Exposure to 10 μg/ml or greater AII tremendously increased the phagocytic capability of peritoneal macrophages. Less than 1% of cells were phagocytic in the resident population (0.01 yeast per cell observed). After exposure to AII this increased to over 25% phagocytic at the highest concentration with on average 1 yeast observed per macrophage (25 fold increase in the number of macrophages able to phagocytose and a 100-fold increase in the number of particles phagocytized). [0093]
  • As shown in Table 4, both concentration of the peptides tested (with the exception of GSD 28) elevated that phagocytic capability of rat macrophages. However, none of the analogues resulted in the magnitude of an effect observed with AII. This suggests that AII and, to a lesser extent, AII analogues will stimulate macrophage differentiation to an elicited or activated state, which leads to the ingestion and clearance of bacteria and cellular debris. [0094]
    TABLE 1
    Effect of All on the Phagocytic Capability of Murine Peritoneal
    Macrophages
    Concentration of All (μg/ml)
    0 1 10 100 1000
    Yeast # # Macrophages with Yeast
    0 100 100 100 100 100
    1-3 1 1 3 10 20
    4-6 0 0 4 10 20
    7-9 0 0 0 0 0
    % Ingesting 1 1 6.5 16.7 25.6
    # Yeast/MO 0.01 0.01 0.24 0.58 1.00
  • [0095]
    TABLE 4
    Effect of AII on the Phagocytic Capability of Rat Peritoneal Macrophages
    Number of Macrophages with Yeast
    (Peptide) (Yeast Number)
    μg/ml 0 1-3 4-6 ≧7 % w/Yeast #/Cell
    AII-0 88 2 0 0 2.2 0.04
    100 90 30 20 0 35.7 1.14
    1000 50 54 40 25 70.4 2.86
    AII (1-7)-0 90 3 0 0 3.2 0.06
    100 100 21 5 0 20.6 0.53
    1000 90 30 10 0 30.8 0.85
    GSD22A-0 90 1 0 0 1.1 0.02
    100 100 26 5 0 23.7 0.59
    1000 100 22 1 0 18.7 0.40
    GSD 24-B-0 100 2 0 0 2.0 0.04
    100 100 25 5 0 23.1 0.58
    1000 100 10 0 0 9.1 0.18
    GSD28-0 100 1 0 0 1.0 0.02
    100 100 1 0 0 1.0 0.02
    1000 100 1 0 0 1.0 0.02
  • [0096]
    TABLE 5
    Designation for Analogues
    Name Abbreviation Sequence
    GSD
    28 Ile8-AII DRVYIHPI
    GSD
    24B Pro3-AII DRPYLHPF
    GSD 22A Ala4-AII RVYAHPF
    AII(1-7) DRVYIHP
  • EXAMPLE 2 Leukocyte Differentiation: Respiratory Burst
  • The respiratory burst of leukocytes (macrophages and polymorphonuclear neutrophils) is one component of the mediator system used to kill bacteria. As with phagocytosis, the level of this respiratory burst activity in resident macrophages is low. With differentiation, either to an elicited (inflammatory) or activated state, this functional activity is significantly elevated. Studies were conducted to assess the effect of in vitro exposure of murine or rat peritoneal macrophages and human peripheral blood mononuclear cells, (PBMC) to various concentrations of AII on the capacity to generate hydrogen peroxide via the respiratory burst system. For the human studies, five different donors were examined. [0097]
  • The murine or rat peritoneal cells were harvested by lavage with 5-15 ml of cold PBS with 0.5% bovine serum albumin. The human PBMC were harvested by venipuncture from normal human volunteers and isolated from peripheral blood by Ficoll Hypaque density centrifugation. After isolation, the cells were resuspended at 1×10[0098] 6 cells/ml and placed at 100 μl per well into 96 well plates. The cells were incubated with various concentrations of AII or AII analogues for 4 hours at 37° C. The cells were then preloaded with a fluorescent probe for hydrogen peroxide, 2,7 dichlorofluorescein acetate, which is nonfluorescent in the absence of hydrogen peroxide. Fifteen minutes later, 10 ng/ml of phorbol myristate acetate (+PMA) or PBS (−PMA) was added to stimulate the production of hydrogen peroxide. One hour after stimulation the level of fluorescence produced was measured on a Cytofluor 2350 multiwell fluorometer. Representative results from this study are shown in FIGS. 3-4.
  • In the absence of PMA or peptide, no hydrogen peroxide production is observed. Some variability in the response to AII was seen (i.e. the concentration of AII necessary to increase the level of this function); however, in all studies AII increased the ability of leukocytes to generate hydrogen peroxide both alone and in response to stimulation with PMA. Further, the effect of pre-exposure to analogues of AII ([0099] GSD 22A, GSD 24B, GSD 28 and AII(1-7)) on the respiratory burst activity of PBMC was assessed (FIGS. 5-8). For all analogues, a much higher concentration of the peptide was needed to increase the respiratory burst activity. Up to 100 times more of these analogues were necessary; however, an increase in the respiratory burst capacity was observed for all analogues tested. The analogues were able to stimulate that function both in the presence and absence of PMA. These data indicate that AII was able to stimulate the differentiation of monocytes/macrophages from three species.
  • EXAMPLE 3 Proliferative Response of Human Lymphocytes
  • Upon stimulation of lymphocytes with mitogen or antigen, these cells undergo blastogenesis and proliferation. In the absence of such stimuli, proliferation is seldom observed. One method to measure cellular proliferation in a short term assay is via measurement of the amount of the nucleotide thymidine that is incorporated into newly synthesized DNA. The effect of AII on the proliferation of human PBMC in the presence and absence of pokeweed mitogen (“PWM”) was assessed. [0100]
  • Human PBMC were collected from normal volunteers and isolated via Ficoll Hypaque (Sigma Chemical, St. Louis) density centrifugation. After isolation of the buffy coat, the cells were washed 3× to remove the Ficoll Hypaque, counted in trypan blue (0.01%) and resuspended at a concentration of 1×10[0101] 6 cells/ml in RPMI 1640 containing 10% fetal calf serum and antibiotics. A 100 μl aliquot of cells was added to each well. Thereafter, various concentrations (0.1 to −1000 μg/ml final concentration) of AII in RPMI 1640 containing 10% FCS and antibiotics were added to various wells in triplicate. To the appropriate wells, PWM (20 μg/ml final concentration) was added. These plates were incubated at 37° C. in 5% CO2 for 48 hours. Subsequently, 0.5 μCi of 3H-thymidine was added to each well, which were incubated at 37° C. for an additional 24 hours prior to harvest by a multiwell automated sample harvester (Skatron) onto glass fiber filters. These filters were dried, placed in scintillation fluid and the amount of thymidine incorporated was determined by beta counting. The results are shown in FIG. 9 and Table 6.
  • In the absence of mitogen, no increase in thymidine incorporation was observed after exposure to AII. However, in two separate experiments (cells from 2 different donors) AII was shown to increase the amount of thymidine incorporated in response to PWM. These data show that AII is able to increase the proliferation of cells from the hematopoietic lineage (e.g. lymphocytes). [0102]
    TABLE 6
    Effect of AII on the Proliferation of Human PBMC
    Proliferation Index
    cpm 3H-Thymidine Incorporated (SIa)
    AII Concentration Experiment 1b Experiment 2
    0 1146.3 (3.63) 2022.0 (31.11)
    0.1 1329.0 (3.54) 2135.0 (38.13)
    1 1411.2 (5.88) 2996.0 (62.42)
    10 1427.0 (5.34) 3946.0 (75.88)
    100 1552.1 (11.41) 3747.0 (69.39)
    1000 1551.5 (15.48) 4302.0 (89.63)
  • EXAMPLE 4 Rat Mesenchymal Stem Cell Proliferation
  • These studies were conducted to determine the effect that AII would have on the proliferation of MSC. Bone marrow cells were harvested from the femur and tibia of female Sprague Dawley rats by flushing the bones with Dulbecco's Minimal Essential Medium-High Glucose (“DMEM-HG”) with a syringe and an 18 gauge needle. These cells were cultured in 24 well plates at 5×10[0103] 3 cells/mm2 in DMEM-HG containing selected lots of fetal calf serum (FCS) and antibiotics (complete medium) at 37?C incubator containing 5% CO2 in air. Twenty-four hours after the initiation of the cultures the medium and nonadherent cells were aspirated and fresh medium was added. To each of these several wells, complete medium with (1 to 100 μg/ml) or without AII was added. The migration of cells from the clones and their proliferation was followed by microscopic examination. Every 4 days the old medium was removed and fresh medium was added to the cultures.
  • The data from these experiments are shown in FIGS. 10 and 11. Addition of AII to the cultures significantly increased the number of sites from which the MSC were migrating (CFU) and the size (number of cells) of the colonies formed. This occurred only in the presence of serum that in itself would support MSC growth, albeit to a lesser extent. As can be seen, AII caused an increase in the number of MSC in a concentration dependent manner at all time points examined in the presence of two different serum lots. These data support the hypothesis that AII can increase the proliferation of rat MSC. [0104]
  • EXAMPLE 5 Effect of Angiotensin II on Murine Lineage-Specific Cells
  • HPC were harvested from C57B1/6 mice by immunomagnetic affinity chromatography and placed in semi-solid cultures with optimal growth medium. At various times after initiation of culture, the formation of colonies and the size of the colonies (number of cells/colony) was assessed microscopically. [0105]
  • Female C57B1/6 mice were purchased from Simonson and used as a source of bone marrow cells in this study. The bone marrow was harvested, either from untreated mice or from mice injected with 5-fluorouracil,(5-FU) (3 mg/mouse; approximately 150 mg/kg) in the tail vein 48 hours before harvest, from the femur and tibia of mice by flushing with phosphate buffered saline (PBS), pH 7.4, containing 2% fetal bovine serum (FBS) with a 21-gauge needle. The eluant from the flushing was centrifuged and the pellet was resuspended at 4×10[0106] 7 nucleated cells/ml in PBS containing PBS containing 2% FBS and 5% normal rat serum.
  • The reagents for immunomagnetic labeling were purchased from Stem Cell Technologies, Inc. (Vancouver, BC). Biotin-labeled monoclonal antibodies to the following murine lineage-specific cell surface antigens were included in a cocktail for HPC enrichment and used according to the manufacturer's instructions: CD5 (Ly-1), CD45-R (B220), CD11b (Mac-1), Myeloid Differentiation Antigen (Gr-1) and Erythroid Cells (TER 119) Ten μl of antibody cocktail was added for each of the 2 sets of bone marrow (normal and 5-FU-treated), mixed and allowed to incubate at 4° C. for 15 minutes. The cells were then resuspended at 4×10[0107] 7 cells/ml in PBS containing 2% FBS. The antibody cocktail was then washed out and 100 μl anti-biotin tetramer was added for each ml of cells. The suspension was mixed and incubated at 4° C. for 15 minutes. Sixty μl of magnetic colloid was then added for each ml of cells, the combination was mixed and incubated at 4° C. for 15 minutes to yield the immunomagnetically-labeled bone marrow cells.
  • A column containing a stainless steel matrix was prepared by washing the matrix with PBS followed by washing with PBS containing 2% protein. The immunomagnetically-labeled bone marrow cells were loaded onto the column and unlabeled cell-containing medium (enriched HPC) was collected in the flow through fraction at a flow rate of 0.2 ml/minute. Medium was added to the top of the column so that it did not run dry until 8 to 10 ml of enriched HPC were harvested. Approximately 2% of the cells loaded onto the column were isolated-in the enriched HPC fractions. [0108]
  • The enriched HPC cell fractions were diluted into a semi-solid medium containing 0.9% methylcellulose in alpha minimal essential medium (alpha MEM), 30% fetal calf serum, 1% bovine serum albumin, 10[0109] −4 M 2-mercaptoethanol, 2 mM L-glutamine, and 2% conditioned medium containing colony stimulating factors. The conditioned medium was supernatant from splenocyte cultures (1×106 cells/ml) incubated for 48 hours in RPMI 1640 containing 10 μg/ml pokeweed mitogen (PWM), 10% FCS, and antibiotics. Various concentrations of AII, between 0 and 100 μg/ml were added in a small volume to the wells of microtiter dishes, to which between 2×110 cells/ml for the normal and 2.5×104 cells/ml for the 5-FU treated cells. The cells were incubated at 37° C. and 5% CO2 for 14 days. At day 14 only, macroscopic cell colonies were observed in the wells containing enriched HPC from untreated (normal) mice treated with 10 μg/ml (18 macroscopic colonies) and 100 μg/ml AII (10 macroscopic colonies). Microscopic evaluation of the cells was performed at various days after initiation of incubation, and the results are summarized in FIGS. 12-17.
  • FIGS. [0110] 12-14 and 16 represent the number of colonies containing more than a certain number of cells/colony as a function of the duration and concentration of AII exposure (FIGS. 12-14 for normal cells; FIG. 16 for 5-FU treated cells.) FIGS. 15 and 17 represents the number of cells per colony seen after incubation of bone marrow from normal or 5-FU treated mice with various concentrations of AII as a function of time. The results clearly demonstrate that HPC colony size increases proportionately with exposure to increased concentrations of AII, and thus that AII increases HPC proliferation.
  • EXAMPLE 6 Effect of AII Analogues and Fragments on Rat Mesenchymal Stem Cell Proliferation
  • These studies were conducted to determine the effect that inclusion of AII analogues and fragments in the cell culture of MSC would have on the proliferation of these cells. Bone marrow cells were harvested from the femur and tibia of female Sprague Dawley rats by flushing the bones with Dulbecco's Minimal Essential Medium-High Glucose (“DMEM-HG”) with a syringe and an 18 gauge needle. These cells were cultured in 24 well plates at 5×10 cells/mm[0111] 2 in DMEM-HG containing selected lots of fetal calf serum (FCS) and antibiotics (complete medium) at 37° C. incubator containing 5% CO2 in air. Twenty-four hours after the initiation of the cultures the medium and nonadherent cells were aspirated and fresh medium was added. To each of these several wells, complete medium with (1 to 100 μg/ml) or without AII analogues and fragments (see Table 5) was added. The migration of cells from the clones and their proliferation was followed by microscopic examination. Every. 4 days the old medium was removed and fresh medium was added to the cultures.
  • Addition of AII analogues or AII fragments to these cultures had a profound effect on the number of sites from which MSC were migrating (CFU) and the size (number of cells) of the colonies formed. The results from these studies can be seen in FIGS. [0112] 18-21. As can be seen, AII analogues and fragments caused an increase in the number of MSC at all time points examined. These data indicate that AI analogues and fragments can increase the proliferation of rat MSC.
  • EXAMPLE 7 Proliferation of MSC Lineage-Specific Cells in the Presence of AII
  • Mesenchymal stem cells isolated from bone marrow and grown under appropriate conditions can express characteristics of multiple cell types, including cells involved in the aspiration of bone, cartilage, muscle and tendons. Osteogenic cells (cells that can form bone tissue) express the enzyme alkaline phosphatase when cultured in medium that drives them toward their osteogenic differentiation. [0113]
  • Bone marrow from female Sprague Dawley rats were harvested by flushing the femur with medium. The cells were placed in [0114] culture dishes 9 cm2 in diameter, allowed to adhere overnight, and then placed in DMEM-LG medium containing antibiotics and 10% fetal calf serum together with varying concentrations of AII. At various times after culture initiation, the cells were washed with Tyrode's buffer and placed in osteogenic medium (DMEM-LG containing 10% fetal calf serum, 100 nM dexamethasone and 0.05 mM ascorbic acid) for 4 days prior to assessment of the level of alkaline phosphatase activity per well. Briefly, the wells were washed with Tyrode's buffer and 1 ml alkaline phosphatase substrate solution (50 mM glycine, pH 10.5, containing 1 mM magnesium chloride and 2.5 mM p-nitrophenyl phosphate) to each well. Fifteen minutes after addition of this aqueous substrate, the buffer was removed from the culture and mixed with 1 ml of 1N sodium hydroxide to stop the reaction. The absorbance of the resultant mixture at 405 nm was then determined via spectroscopy. The level of alkaline phosphatase activity is expressed as the level of absorbance per culture dish. These data are shown in FIG. 22 and demonstrate that AII can accelerate the proliferation of cells that express alkaline phosphatase when placed in medium appropriate to induce osteogenic differentiation.
  • EXAMPLE 8 Effect of Angiotensin II on Murine Lineage-Specific Cells
  • HPC were harvested from C57B1/6 mice by immunomagnetic affinity chromatography and placed in semi-solid cultures with optimal growth medium in the absence of colony stimulating factors. At various times after initiation of culture, the formation of colonies and the size of the colonies. (number of cells/colony) was assessed microscopically. [0115]
  • Female C57B1/6 mice were purchased from Simonson and used as a source of bone marrow cells in this study. The bone marrow was harvested from the femur and tibia of mice by flushing with phosphate buffered saline (PBS), pH 7.4, containing 2% fetal bovine serum (FBS) with a 21-gauge needle. The eluant from the flushing was centrifuged and the pellet was resuspended at 4×10[0116] 7 nucleated cells/ml in PBS containing PBS containing 2% FBS and 5% normal rat serum.
  • The reagents for immunomagnetic labeling were purchased from Stem Cell Technologies, Inc. (Vancouver, BC). Biotin-labeled monoclonal antibodies to the following murine lineage-specific cell surface antigens were included in a cocktail for HPC enrichment and used according to the manufacturer's instructions: CD5 (Ly-[0117] 1), CD45-R (B220), CD11b (Mac-1), Myeloid Differentiation Antigen (Gr-1) and Erythroid Cells (TER 119) Ten μl of antibody cocktail was added to the bone marrow, mixed and allowed to incubate at 4° C. for 15 minutes. The cells were then resuspended at 4×107 cells/ml in PBS containing 2% FBS. The antibody cocktail was then washed out and 100 μl anti-biotin tetramer was added for each ml of cells. The suspension was mixed and incubated at 4° C. for 15 minutes. Sixty ill of magnetic colloid was then added for each ml of cells, the combination was mixed and incubated at 4° C. for 15 minutes to yield the immunomagnetically-labeled bone marrow cells.
  • A column containing a stainless steel matrix was prepared by washing the matrix with PBS followed by washing with PBS containing 2% protein. The immunomagnetically-labeled bone marrow cells were loaded onto the column and unlabeled cell-containing medium (enriched HPC) was collected in the flow through fraction at a flow rate of 0.2 ml/minute. Medium was added to the top of the column so that it did not run dry until 8 to 10 ml of enriched HPC were harvested. Approximately 2% of the cells loaded onto the column were isolated in the enriched HPC fractions. [0118]
  • The enriched HPC cell fractions were diluted into a semi-solid medium containing 0.9% methylcellulose in alpha minimal essential medium (alpha MEM), 30% fetal calf serum, 1% bovine serum albumin, 10[0119] −4 M 2-mercaptoethanol, 2 mM L-glutamine, and 2% conditioned medium. The conditioned medium was supernatant from splenocyte cultures (1×106 cells/ml) incubated for 48 hours in RPMI 1640 containing 10 μg/ml pokeweed mitogen PWM), 10% FCS, and antibiotics. Various concentrations of AII, between 0 and 100;g/ml, or AII analogues or AII fragments were added in a small volume to the wells of microtiter dishes, which contained approximately either 3,000 cells per well or 30,000 cells per well. The cells were incubated at 37° C. and 5% CO2 for 14 days. Microscopic evaluation of the cells was performed at various days after initiation of incubation, and the results are summarized in FIGS. 23-33. The results clearly demonstrate that HPC colony size increases proportionately with exposure to increased concentrations of AII, AII analogues and AII fragments, and thus that AII increases HPC proliferation in the absence of colony stimulating factors.
  • It is to be understood that the invention is not to be limited to the exact details of operation, or to the exact compounds, compositions, methods, procedures or embodiments shown and described, as obvious modifications and equivalents will be apparent to one skilled in the art, and the invention is therefore to be limited only by the full scope of the appended claims. [0120]
  • 1 37 1 8 PRT Artificial Sequence Description of Artificial SequenceAII 1 Asp Arg Val Tyr Ile His Pro Phe 1 5 2 7 PRT Artificial Sequence Description of Artificial SequenceAII (2-8) 2 Arg Val Tyr Ile His Pro Phe 1 5 3 6 PRT Artificial Sequence Description of Artificial SequenceAII (3-8) 3 Val Tyr Ile His Pro Phe 1 5 4 7 PRT Artificial Sequence Description of Artificial SequenceAII (1-7) 4 Asp Arg Val Tyr Ile His Pro 1 5 5 6 PRT Artificial Sequence Description of Artificial SequenceAII (2-7) 5 Arg Val Tyr Ile His Pro 1 5 6 5 PRT Artificial Sequence Description of Artificial SequenceAII (3-7) 6 Val Tyr Ile His Pro 1 5 7 4 PRT Artificial Sequence Description of Artificial SequenceAII (5-8) 7 Ile His Pro Phe 1 8 6 PRT Artificial Sequence Description of Artificial SequenceAII (1-6) 8 Asp Arg Val Tyr Ile His 1 5 9 5 PRT Artificial Sequence Description of Artificial SequenceAII (1-5) 9 Asp Arg Val Tyr Ile 1 5 10 4 PRT Artificial Sequence Description of Artificial SequenceAII (1-4) 10 Asp Arg Val Tyr 1 11 3 PRT Artificial Sequence Description of Artificial SequenceAII (1-3) 11 Asp Arg Val 1 12 7 PRT Artificial Sequence Description of Artificial SequenceAII analogue 12 Arg Xaa Tyr Ile His Pro Phe 1 5 13 7 PRT Artificial Sequence Description of Artificial SequenceAII analogue 13 Arg Val Tyr Xaa His Pro Phe 1 5 14 3 PRT Artificial Sequence Description of Artificial SequenceAII (6-8) 14 His Pro Phe 1 15 5 PRT Artificial Sequence Description of Artificial SequenceAII (4-8) 15 Tyr Ile His Pro Phe 1 5 16 7 PRT Artificial Sequence Description of Artificial SequenceAII analogue class 16 Xaa Xaa Tyr Xaa His Pro Phe 1 5 17 7 PRT Artificial Sequence Description of Artificial SequenceAII analogue 17 Arg Val Tyr Gly His Pro Phe 1 5 18 7 PRT Artificial Sequence Description of Artificial SequenceAII analogue 18 Arg Val Tyr Ala His Pro Phe 1 5 19 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 1 19 Asp Arg Val Tyr Val His Pro Phe 1 5 20 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 2 20 Asn Arg Val Tyr Val His Pro Phe 1 5 21 11 PRT Artificial Sequence Description of Artificial SequenceAII analogue 3 21 Ala Pro Gly Asp Arg Ile Tyr Val His Pro Phe 1 5 10 22 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 4 22 Glu Arg Val Tyr Ile His Pro Phe 1 5 23 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 5 23 Asp Lys Val Tyr Ile His Pro Phe 1 5 24 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 6 24 Asp Arg Ala Tyr Ile His Pro Phe 1 5 25 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 7 25 Asp Arg Val Thr Ile His Pro Phe 1 5 26 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 8 26 Asp Arg Val Tyr Leu His Pro Phe 1 5 27 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 9 27 Asp Arg Val Tyr Ile Arg Pro Phe 1 5 28 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 10 28 Asp Arg Val Tyr Ile His Ala Phe 1 5 29 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 11 29 Asp Arg Val Tyr Ile His Pro Tyr 1 5 30 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 12 30 Pro Arg Val Tyr Ile His Pro Phe 1 5 31 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 13 31 Asp Arg Pro Tyr Ile His Pro Phe 1 5 32 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 14 32 Asp Arg Val Tyr Ile His Pro Phe 1 5 33 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 15 33 Asp Arg Xaa Tyr Ile His Pro Phe 1 5 34 8 PRT Artificial Sequence Description of Artificial SequenceAII analogue 16 34 Asp Arg Val Tyr Xaa His Pro Phe 1 5 35 9 PRT Artificial Sequence Description of Artificial SequenceAII analogue 17 35 Asp Arg Val Xaa Tyr Ile His Pro Phe 1 5 36 8 PRT Artificial Sequence Description of Artificial Sequencep-aminophenylalanine 6 AII 36 Asp Arg Val Tyr Ile Xaa Pro Phe 1 5 37 10 PRT Artificial Sequence Description of Artificial Sequenceangiotensin I 37 Asp Arg Val Tyr Ile His Pro Phe His Leu 1 5 10

Claims (60)

We claim:
1. A method of accelerating the proliferation of mesenchymal stem cells comprising contacting the mesenchymal stem cells with an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
2. A method of accelerating the proliferation of hematopoietic lineage-specific cells comprising contacting the hematopoietic lineage-specific cells with an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4 —R5—R6—R7-R8
in which R1 R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), He and Tyr, excluding sequences including R4 as a terminal Tyr group.
3. A method of accelerating the proliferation of mesenchymal lineage-specific cells comprising contacting the mesenchymal lineage-specific cells with an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
4. A method of accelerating the proliferation of hematopoietic lineage-specific cells comprising contacting the hematopoietic lineage-specific cells with an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous ammo acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
5. A method of increasing the differentiation of hematopoietic stem or lineage-specific cells comprising contacting the hematopoietic stem or lineage-specific cells with an amount effective to increase differentiation of the hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
6. The method of claim 5, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting of macrophages and monocytes.
7. The method of claim 1, 2, 3, 4, 5, or 6 wherein the active agent is selected from the group consisting of AII, AIII, AII(2-8), SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, and SEQ ID NO:31.
8. The method of claims 1, 2, 3, 4, 5, or 6 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
9. An improved cell culture medium for growth of mesenchymal stem cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence, consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
10. An improved cell culture medium for growth of hematopoietic lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and. Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
11. An improved cell culture medium for growth of mesenchymal lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
12. An improved cell culture medium for growth of hematopoietic lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
13. An improved cell culture medium for differentiation of hematopoietic stem or lineage-specific cells, wherein the improvement comprises addition to the cell culture medium an amount effective to increase differentiation of the hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
14. The method of claim 13, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting essentially of macrophages and monocytes.
15. The improved cell culture medium of claim 9, 10, 11, 12, 13, or 14 wherein the active agent is selected from the group consisting of AII, AIII, AII(2-8), SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5,, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, and SEQ ID NO:31.
16. The improved cell culture medium of claim 9, 10, 11, 12, 13, or 14 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
17. A kit for the propagation of mesenchymal stem cells, comprising:
(a) an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala, and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
18. The kit of claim 17, further comprising mesenchymal stem cell growth medium.
19. A kit for the propagation of hematopoietic lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
20. The kit of claim 19, further comprising hematopoietic lineage-specific cell growth medium.
21. A kit for the propagation of mesenchymal lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
22. The kit of claim 21, further comprising mesenchymal lineage-specific cell growth medium.
23. A kit for the propagation of hematopoietic lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
f R3 is selected from the group consisting of Val, Ala, Leu, norLeu, le, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
24. The kit of claim 23, further comprising hematopoietic lineage-specific cell growth medium.
25. A kit for the differentiation of hematopoietic stem or lineage-specific cells, comprising:
(a) an amount effective to accelerate differentiation of hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R1-R8 in the sequence of general formula I
R1—R2—R3—R4—R5—R6—R7-R8
in which R1 and R2 together form a group of formula
X—RA—RB—,
wherein X is H or a one to three peptide group and a peptide bond between RA and RB is labile to aminopeptidase A cleavage;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, excluding sequences including R4 as a terminal Tyr group.
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
26. The kit of claim 25, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting of macrophages and monocytes.
27. The kit of claim 25 or 26, further comprising hematopoietic lineage-specific cell growth medium.
28. The kit of claim 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, or 27 further comprising a sterile container.
29. The kit of claim 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, or 27 wherein the active agent is selected from the group consisting of AII, AIII, AII(2-8), SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID NO:12, SEQ ID NO:13, and SEQ ID NO:31.
30. The kit of claim 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, or 27 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
31. A method of accelerating the proliferation of mesenchymal stem cells comprising contacting the mesenchymal stem cells with an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
32. A method of accelerating the proliferation of hematopoietic lineage-specific cells comprising contacting the hematopoietic lineage-specific cells with an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), He and Tyr.
33. A method of accelerating the proliferation of mesenchymal lineage-specific cells comprising contacting the mesenchymal lineage-specific cells with an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
34. A method of accelerating the proliferation of hematopoietic lineage-specific cells comprising contacting the hematopoietic lineage-specific cells with an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
35. A method of increasing the differentiation of hematopoietic stem or lineage-specific cells comprising contacting the hematopoietic stem or lineage-specific cells with an amount effective to increase differentiation of the hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Try.
36. The method of claim 35, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting of macrophages and monocytes.
37. The method of claim 31, 32, 33, 34, 35, or 36 wherein the active agent is selected from the group consisting of SEQ ID NO:2, SEQ ID NO:17, and SEQ ID NO:18.
38. The method of claims 31, 32, 33, 34, 35, or 36 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
39. An improved cell culture medium for growth of mesenchymal stem cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
40. An improved cell culture medium for growth of hematopoietic lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
41. An improved cell culture medium for grown of mesenchymal lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
42. An improved cell culture medium for growth of hematopoietic lineage-specific cells, wherein the improvement comprises addition to the cell culture medium of an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr,
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
43. An improved cell culture medium for differentiation of hematopoietic stem or lineage-specific cells, wherein the improvement comprises addition to the cell culture medium an amount effective to increase differentiation of the hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr.
44. The method of claim 43, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting of macrophages and monocytes.
45. The improved cell culture medium of claim 39, 40 41, 42, 43, or 44 wherein the active agent is selected from the group consisting SEQ ID NO:2, SEQ ID NO:17, and SEQ ID NO:18.
46. The improved cell culture medium of claim 39, 40, 41, 42, 43, or 44 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
47. A kit for the propagation of mesenchymal stem cells, comprising:
(a) an amount effective to accelerate proliferation of mesenchymal stem cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
48. The kit of claim 47, further comprising mesenchymal stem cell growth medium.
49. A kit for the propagation of hematopoietic lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous
amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr; and
;(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
50. The kit of claim 49, further comprising hematopoietic lineage-specific cell growth medium.
51. A kit for the propagation of mesenchymal lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of mesenchymal lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr, and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
52. The kit of claim 51, further comprising mesenchymal lineage-specific cell growth medium.
53. A kit for the propagation of hematopoietic lineage-specific cells, comprising:
(a) an amount effective to accelerate proliferation of hematopoietic lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Ser(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
54. The kit of claim 53, further comprising hematopoietic lineage-specific cell growth medium.
55. A kit for the differentiation of hematopoietic stem or lineage-specific cells, comprising:
(a) an amount effective to accelerate differentiation of hematopoietic stem or lineage-specific cells of at least one active agent comprising a sequence consisting of at least three contiguous amino acids of groups R2-R8 in the sequence of general formula II
R2—R3—R4—R5—R6—R7—R8
in which R2 is selected from the group consisting of H, Arg, Lys, Ala, Orn, Se r(Ac), Sar, D-Arg and D-Lys;
R3 is selected from the group consisting of Val, Ala, Leu, norLeu, Ile, Gly, Pro, Aib, Acpc and Tyr;
R4 is selected from the group consisting of Tyr, Tyr(PO3)2, Thr, Ser, homoSer and azaTyr;
R5 is selected from the group consisting of Ile, Ala, Leu, norLeu, Val and Gly;
R6 is His, Arg or 6-NH2-Phe;
R7 is Pro or Ala; and
R8 is selected from the group consisting of Phe, Phe(Br), Ile and Tyr; and
(b) instructions for using the amount effective of active agent as a cell culture medium supplement.
56. The kit of claim 55, wherein the hematopoietic stem or lineage-specific cells are selected from the group consisting of macrophages and monocytes.
57. The kit of claim 55 or 56, further comprising hematopoietic lineage-specific cell growth medium.
58. The kit of claim 47, 48, 49, 50, 51, 52, 53, 54, 55, 56 or 57 further comprising a sterile container.
59. The kit of claim 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, or 57 wherein the active agent is selected from the group consisting of SEQ ID NO:2, SEQ ID NO:17, and SEQ ID NO:18.
60. The kit of claim 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, or 57 wherein the concentration of active agent is between about 0.1 ng/kg and about 1.0 mg/kg.
US10/360,274 1997-01-28 2003-02-07 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation Abandoned US20040006003A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/360,274 US20040006003A1 (en) 1997-01-28 2003-02-07 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US3650797P 1997-01-28 1997-01-28
US4685997P 1997-05-08 1997-05-08
US6368497P 1997-10-28 1997-10-28
US6391097P 1997-10-31 1997-10-31
US6561297P 1997-11-18 1997-11-18
US6659397P 1997-11-26 1997-11-26
US09/012,400 US6335195B1 (en) 1997-01-28 1998-01-23 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US71688600A 2000-11-20 2000-11-20
US10/360,274 US20040006003A1 (en) 1997-01-28 2003-02-07 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US71688600A Continuation 1997-01-28 2000-11-20

Publications (1)

Publication Number Publication Date
US20040006003A1 true US20040006003A1 (en) 2004-01-08

Family

ID=27556310

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/012,400 Expired - Lifetime US6335195B1 (en) 1997-01-28 1998-01-23 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US09/564,045 Expired - Lifetime US7118748B1 (en) 1997-01-28 2000-05-03 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US10/360,274 Abandoned US20040006003A1 (en) 1997-01-28 2003-02-07 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US11/442,624 Expired - Fee Related US7744927B2 (en) 1997-01-28 2006-05-26 Methods of promoting hematopoietic and mesenchymal cell proliferation and differentiation

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/012,400 Expired - Lifetime US6335195B1 (en) 1997-01-28 1998-01-23 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US09/564,045 Expired - Lifetime US7118748B1 (en) 1997-01-28 2000-05-03 Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/442,624 Expired - Fee Related US7744927B2 (en) 1997-01-28 2006-05-26 Methods of promoting hematopoietic and mesenchymal cell proliferation and differentiation

Country Status (7)

Country Link
US (4) US6335195B1 (en)
EP (1) EP0973539A2 (en)
JP (2) JP3662029B2 (en)
AU (1) AU737058B2 (en)
CA (1) CA2278737A1 (en)
MX (1) MXPA99007026A (en)
WO (1) WO1998032457A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090069246A1 (en) * 1997-01-28 2009-03-12 University Of Southern California Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US7523131B2 (en) * 2005-02-10 2009-04-21 Oracle International Corporation Techniques for efficiently storing and querying in a relational database, XML documents conforming to schemas that contain cyclic constructs
US20130316361A1 (en) * 2010-12-06 2013-11-28 Thd S.P.A. Method for the diagnosis of a carcinoma and uses thereof
US9259443B2 (en) 2010-10-25 2016-02-16 The Children's Hospital Of Philadelphia Compositions and methods for the generation of platelets and methods of use thereof
US20160118821A1 (en) * 2014-10-22 2016-04-28 Mitsumi Electric Co., Ltd. Battery protection circuit, battery protection apparatus, and battery pack
US20170357603A1 (en) * 2016-06-14 2017-12-14 Oracle International Corporation System and method for filtering field programmable gate array input/output

Families Citing this family (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1047441A1 (en) * 1997-11-26 2000-11-02 University Of Southern California Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
AU755225B2 (en) * 1997-12-12 2002-12-05 University Of Southern California Wound healing compositions
US6239109B1 (en) * 1998-02-09 2001-05-29 University Of Southern California Method of promoting erythropoiesis
AU2590399A (en) * 1998-02-09 1999-08-23 Gere Dizerega Method of promoting hepatic cell proliferation
WO2000018885A1 (en) * 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
AU2680999A (en) * 1998-02-19 1999-09-06 University Of Southern California Method of promoting embryonic stem cell proliferation
EP1054684A1 (en) * 1998-02-19 2000-11-29 University Of Southern California Method of promoting neuronal cell proliferation and differentiation
US7173011B2 (en) * 2000-11-20 2007-02-06 University Of Southern California Radiation therapy methods
AU3077299A (en) 1998-03-11 1999-09-27 University Of Southern California Method of promoting production of living tissue equivalents
DE69924715T2 (en) 1998-05-11 2006-01-26 University Of Southern California, Los Angeles USE OF ANGIOTENSIN ANALOGUE FOR THE PREPARATION OF A MEDICAMENT FOR INCREASING THE LIFE OF BLOOD CELLS AND MOBILIZING HAEMATOPOETIC STEM CELLS AFTER CHEMOTHERAPEUTIC TREATMENT
US6762167B1 (en) 1998-05-11 2004-07-13 University Of Southern California Methods for treating a patient undergoing chemotherapy
US6916783B2 (en) 1998-07-13 2005-07-12 University Of Southern California Methods for accelerating bone and cartilage growth and repair
DE69911609T2 (en) 1998-07-13 2004-07-01 University Of Southern California, Los Angeles PROCESS FOR ACCELERATING THE GROWTH AND HEALING OF BONES AND CARTILES
EP1105149A1 (en) 1998-08-13 2001-06-13 University Of Southern California Methods to increase blood flow to ischemic tissue
US6730775B1 (en) 1999-03-23 2004-05-04 University Of Southern California Methods for limiting scar and adhesion formation
US7338938B2 (en) * 1999-05-10 2008-03-04 University Of Southern California Methods for treating a patient undergoing chemotherapy
US7670628B2 (en) * 1999-07-07 2010-03-02 Angioblast Systems, Inc. Mesenchymal precursor cell
AUPQ147799A0 (en) 1999-07-07 1999-07-29 Medvet Science Pty. Ltd. Mesenchymal precursor cell
US8062675B2 (en) * 1999-07-07 2011-11-22 Angioblast Systems, Inc. Mesenchymal precursor cell
AU2003901668A0 (en) * 2003-03-28 2003-05-01 Medvet Science Pty. Ltd. Non-haemopoietic precursor cells
US20050158289A1 (en) * 1999-07-07 2005-07-21 Simmons Paul J. Mesenchymal precursor cell and use thereof in the repair of bone defects and fractures in mammals
US6821953B1 (en) 1999-12-16 2004-11-23 University Of Southern California Methods for treating and preventing damage to mucosal tissue
US6747008B1 (en) 2000-06-19 2004-06-08 University Of Southern California Methods for treating and preventing alopecia
US20030032179A1 (en) 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
AU2002220209B2 (en) * 2000-12-06 2006-05-25 Robert J. Hariri Method of collecting placental stem cells
US7311905B2 (en) * 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
JP2005514926A (en) * 2002-01-14 2005-05-26 ザ・ボード・オブ・トラスティーズ・オブ・ザ・ユニバーシティ・オブ・イリノイ Novel mammalian multipotent stem cells and compositions, methods for their preparation and administration
IL152904A0 (en) * 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) * 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
EP2186407A1 (en) 2002-02-13 2010-05-19 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
WO2003078567A2 (en) * 2002-03-18 2003-09-25 Gamida-Cell Ltd. Methods of inducing differentiation in ex vivo expanded stem cells
MXPA04009997A (en) * 2002-04-12 2004-12-13 Celgene Corp Modulation of stem and progenitor cell differentiation, assays, and uses thereof.
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
JP2005528105A (en) * 2002-05-30 2005-09-22 セルジーン・コーポレーション Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative diseases and myelodysplastic syndromes
WO2004009767A2 (en) * 2002-07-23 2004-01-29 Boston Scientific Limited Cell therapy for regeneration
US20040126879A1 (en) * 2002-08-29 2004-07-01 Baylor College Of Medicine Heart derived cells for cardiac repair
US20050054097A1 (en) * 2002-11-17 2005-03-10 Tony Peled EX-VIVO expansion of hematopoietic system cell populations in mononuclear cell cultures
KR101042448B1 (en) * 2002-11-26 2011-06-16 안트로제네시스 코포레이션 Cytotherapeutics, cytotherapeutic units and methods for treatments using them
NZ566132A (en) * 2003-02-13 2009-09-25 Anthrogenesis Corp Use of umbilical cord blood to treat inflammation, ParkinsonÆs disease or diabetes
CA2517959A1 (en) * 2003-03-07 2004-09-16 Gamida-Cell Ltd. Expansion of renewable stem cell populations using modulators of pi 3-kinase
US20060162014A1 (en) * 2003-07-07 2006-07-20 Jaffe Eileen K Alternate morpheeins of allosteric proteins as a target for the development of bioactive molecules
US8153410B2 (en) * 2003-07-07 2012-04-10 Fox Chase Cancer Center Alternate morpheein forms of allosteric proteins as a target for the development of bioactive molecules
AU2004263865B2 (en) 2003-08-08 2007-05-17 Sangamo Therapeutics, Inc. Methods and compositions for targeted cleavage and recombination
US7888121B2 (en) 2003-08-08 2011-02-15 Sangamo Biosciences, Inc. Methods and compositions for targeted cleavage and recombination
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
KR100534215B1 (en) * 2003-11-11 2005-12-08 (주)히스토스템 Method of isolating and culturing mesenchymal stem cell derived from cryopreserved umbilical cord blood
KR100560340B1 (en) * 2003-11-11 2006-03-14 한훈 Method of isolating and culturing mesenchymal stem cell derived from umbilical cord blood
WO2005055929A2 (en) * 2003-12-02 2005-06-23 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
ZA200607800B (en) 2004-03-26 2009-02-25 Celgene Corp System and methods for providing a stem cell bank
EP1745125A1 (en) * 2004-05-14 2007-01-24 Becton, Dickinson and Company Cell culture environments for the serum-free expansion of mesenchymal stem cells
US20070048253A1 (en) * 2004-06-30 2007-03-01 Goodwin Thomas J Natively glycosylated mammalian biological molecules produced by electromagnetically stimulating living mammalian cells
WO2006030442A2 (en) * 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
KR101598947B1 (en) 2004-09-24 2016-03-08 메소블라스트, 아이엔씨. Method of enhancing proliferation and/or survival of mesenchymal precursor cells (mpc)
EP1812555B1 (en) 2004-10-25 2015-04-22 Cellerant Therapeutics, Inc. Methods of expanding myeloid cell populations and uses thereof
AU2006272634B2 (en) 2005-07-26 2013-01-24 Sangamo Therapeutics, Inc. Targeted integration and expression of exogenous nucleic acid sequences
ES2452595T3 (en) 2005-10-13 2014-04-02 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
KR20210122908A (en) * 2005-12-29 2021-10-12 안트로제네시스 코포레이션 Placental stem cell populations
AU2006332679A1 (en) 2005-12-29 2007-07-12 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
EP1984491B1 (en) 2006-02-14 2016-12-07 Cellerant Therapeutics, Inc. Compositions for enhancing engraftment of hematopoietic stem cells
PT2548951E (en) 2006-03-23 2016-06-14 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
EP2091335A4 (en) * 2006-11-09 2012-07-04 Gamida Cell Ltd Use of ex-vivo cultured hematopoietic cells for treatment of peripheral vascular diseases
DK2120977T3 (en) 2007-02-12 2013-08-12 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
KR101614823B1 (en) * 2007-09-11 2016-04-22 훗카이도 코리츠 다이가쿠 호진 삿포르 이카 다이가쿠 Cell growth method and pharmaceutical preparation for tissue repair and regeneration
KR101644659B1 (en) * 2007-09-26 2016-08-01 안트로제네시스 코포레이션 Angiogenic cells from human placental perfusate
KR20210022148A (en) * 2007-09-28 2021-03-02 안트로제네시스 코포레이션 Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
KR20160040739A (en) * 2007-11-07 2016-04-14 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
WO2009114461A2 (en) * 2008-03-10 2009-09-17 University Of Southern California Angiotensin (1-7) dosage forms and uses thereof
KR101903049B1 (en) * 2008-08-20 2018-11-07 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
KR20110050521A (en) * 2008-08-20 2011-05-13 안트로제네시스 코포레이션 Improved cell composition and methods of making the same
KR20110050688A (en) * 2008-08-22 2011-05-16 안트로제네시스 코포레이션 Methods and compositions for treatment of bone defects with placental cell populations
AU2009316541B2 (en) * 2008-11-19 2015-08-06 Celularity Inc. Amnion derived adherent cells
ES2731340T3 (en) * 2008-11-21 2019-11-15 Celularity Inc Treatment of diseases, disorders or lung conditions using placental cells
WO2011017315A2 (en) 2009-08-03 2011-02-10 Recombinetics, Inc. Methods and compositions for targeted gene modification
EP3284818B1 (en) * 2010-01-26 2022-03-09 Celularity Inc. Treatment of bone-related cancers using placental stem cells
KR20230054905A (en) 2010-04-07 2023-04-25 셀룰래리티 인코포레이티드 Angiogenesis using placental stem cells
WO2011127113A1 (en) 2010-04-08 2011-10-13 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
ES2666746T3 (en) 2010-07-13 2018-05-07 Anthrogenesis Corporation Methods to generate natural cytolytic lymphocytes
US9725689B2 (en) 2010-10-08 2017-08-08 Terumo Bct, Inc. Configurable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
CA2793515C (en) 2011-02-02 2015-05-12 University Of Southern California Methods for treating diabetic foot ulcers
WO2012115940A2 (en) * 2011-02-21 2012-08-30 Duke University A method of inducing hematopoietic reconstruction
PL2714059T3 (en) 2011-06-01 2019-04-30 Celularity Inc Treatment of pain using placental stem cells
JP6214530B2 (en) 2011-07-15 2017-10-18 ザ ジェネラル ホスピタル コーポレイション Method for assembling a transcription activator-like effector
FR2979110B1 (en) * 2011-08-16 2013-09-27 Etat Francais Ministere De La Defense Service De Sante Des Armees IN VITRO MODELING OF HEMATOPOIETIC STEM CELL MEDICINAL NICHES: A TOOL FOR STUDYING REGULATION OF HEMATOPOIESE, EVALUATING THE POTENTIAL OF NICHING A HEMATOPOIETIC GRAFT AND TESTING THE PHARMACO-TOXICOLOGY OF MEDICINAL PRODUCTS.
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
US8889630B2 (en) * 2011-12-23 2014-11-18 Carlos Lopez Method for hair regrowth using Granulocyte-Colony Stimulating Factor
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
EP2849773A1 (en) 2012-05-14 2015-03-25 University of Southern California Methods for limiting development of a skin wound
US8557958B1 (en) 2012-06-18 2013-10-15 Tarix Pharmaceuticals Ltd. Compositions and methods for treatment of diabetes
US9567569B2 (en) * 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US8633158B1 (en) 2012-10-02 2014-01-21 Tarix Pharmaceuticals Ltd. Angiotensin in treating brain conditions
AU2014207618A1 (en) 2013-01-16 2015-08-06 Emory University Cas9-nucleic acid complexes and uses related thereto
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US9623084B2 (en) 2013-03-15 2017-04-18 University Of Southern California Methods for treating multiple sclerosis
EP3935944A1 (en) 2013-03-15 2022-01-12 University Of Southern California, USC Stevens Compounds for the treatment of musculoskeletal diseases
CA2916701A1 (en) 2013-07-03 2015-01-08 The Arizona Board Of Regents On Behalf Of The University Of Arizona Method for treating cognitive dysfunction
WO2015073913A1 (en) 2013-11-16 2015-05-21 Terumo Bct, Inc. Expanding cells in a bioreactor
US9333233B2 (en) 2014-02-25 2016-05-10 Tarix Pharmaceuticals Ltd. Methods and compositions for the delayed treatment of stroke
WO2015148704A1 (en) 2014-03-25 2015-10-01 Terumo Bct, Inc. Passive replacement of media
US10183055B2 (en) 2014-07-21 2019-01-22 Arizona Board Of Regents On Behalf Of The University Of Arizona Ang-(1-7) derivative oligopeptides for the treatment of pain and other indications
WO2016014342A2 (en) 2014-07-21 2016-01-28 Arizona Board Of Regents For The University Of Arizona Ang-(1-7) derviative oligopeptides and methods for using and producing the same
CN106715676A (en) 2014-09-26 2017-05-24 泰尔茂比司特公司 Scheduled feed
US10117911B2 (en) 2015-05-29 2018-11-06 Agenovir Corporation Compositions and methods to treat herpes simplex virus infections
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
KR20180048617A (en) 2015-07-20 2018-05-10 앤지오크린 바이오사이언스 인코포레이티드 Methods and compositions for stem cell transplantation
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
WO2018002673A1 (en) 2016-07-01 2018-01-04 N4 Pharma Uk Limited Novel formulations of angiotensin ii receptor antagonists
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
US11702634B2 (en) 2017-03-31 2023-07-18 Terumo Bct, Inc. Expanding cells in a bioreactor

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5015629A (en) * 1989-06-26 1991-05-14 University Of Southern California Tissue repair
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5629292A (en) * 1993-09-24 1997-05-13 The University Of Southern California Use of angiotensin III and analogs thereof in tissue repair
US5693616A (en) * 1992-07-14 1997-12-02 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5716935A (en) * 1993-09-24 1998-02-10 University Of Southern California Use of antiotensin II analogs in tissue repair
US6096709A (en) * 1993-09-24 2000-08-01 University Of Southern California Use of angiotensin II fragments and analogs thereof in tissue repair
US6110895A (en) * 1996-12-16 2000-08-29 University Of Southern California Method of promoting healing in skin grafts
US6165978A (en) * 1997-12-12 2000-12-26 University Of Southern California Wound healing composition
US6177407B1 (en) * 1998-08-13 2001-01-23 University Of Southern California Methods to increase blood flow to ischemic tissue
US6239109B1 (en) * 1998-02-09 2001-05-29 University Of Southern California Method of promoting erythropoiesis
US6248587B1 (en) * 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6258778B1 (en) * 1998-07-13 2001-07-10 University Of Southern California Methods for accelerating bone and cartilage growth and repair
US6334195B1 (en) * 1995-12-29 2001-12-25 Lsi Logic Corporation Use of hot spare drives to boost performance during nominal raid operation
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6444646B1 (en) * 1995-06-06 2002-09-03 University Of Southern California Use of angiotensin II type 2 receptor agonists in tissue repair
US6455500B1 (en) * 1998-03-10 2002-09-24 University Of Southern California Radiation therapy methods
US6475988B1 (en) * 1998-05-11 2002-11-05 University Of Southern California Methods to increase white blood cell survival after chemotherapy
US6498138B1 (en) * 1998-03-11 2002-12-24 University Of Southern California Method of promoting production of living tissue equivalents
US6747008B1 (en) * 2000-06-19 2004-06-08 University Of Southern California Methods for treating and preventing alopecia
US6762167B1 (en) * 1998-05-11 2004-07-13 University Of Southern California Methods for treating a patient undergoing chemotherapy
US6821953B1 (en) * 1999-12-16 2004-11-23 University Of Southern California Methods for treating and preventing damage to mucosal tissue

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997034627A2 (en) 1996-03-19 1997-09-25 Societe De Conseils De Recherches Et D'applications Scientifiques S.A. Protection of hemopoietic cells during chemotherapy or radiotherapy
US7173011B2 (en) 2000-11-20 2007-02-06 University Of Southern California Radiation therapy methods
US6916783B2 (en) * 1998-07-13 2005-07-12 University Of Southern California Methods for accelerating bone and cartilage growth and repair
US6730775B1 (en) * 1999-03-23 2004-05-04 University Of Southern California Methods for limiting scar and adhesion formation
US7338938B2 (en) * 1999-05-10 2008-03-04 University Of Southern California Methods for treating a patient undergoing chemotherapy
KR101316423B1 (en) * 2007-08-09 2013-10-08 엘지이노텍 주식회사 Semiconductor light emitting device and fabrication method thereof

Patent Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5015629A (en) * 1989-06-26 1991-05-14 University Of Southern California Tissue repair
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5693616A (en) * 1992-07-14 1997-12-02 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
US5629292A (en) * 1993-09-24 1997-05-13 The University Of Southern California Use of angiotensin III and analogs thereof in tissue repair
US5716935A (en) * 1993-09-24 1998-02-10 University Of Southern California Use of antiotensin II analogs in tissue repair
US6096709A (en) * 1993-09-24 2000-08-01 University Of Southern California Use of angiotensin II fragments and analogs thereof in tissue repair
US6444646B1 (en) * 1995-06-06 2002-09-03 University Of Southern California Use of angiotensin II type 2 receptor agonists in tissue repair
US6334195B1 (en) * 1995-12-29 2001-12-25 Lsi Logic Corporation Use of hot spare drives to boost performance during nominal raid operation
US6110895A (en) * 1996-12-16 2000-08-29 University Of Southern California Method of promoting healing in skin grafts
US6335195B1 (en) * 1997-01-28 2002-01-01 Maret Corporation Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6248587B1 (en) * 1997-11-26 2001-06-19 University Of Southern Cailfornia Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6165978A (en) * 1997-12-12 2000-12-26 University Of Southern California Wound healing composition
US6239109B1 (en) * 1998-02-09 2001-05-29 University Of Southern California Method of promoting erythropoiesis
US6455500B1 (en) * 1998-03-10 2002-09-24 University Of Southern California Radiation therapy methods
US6498138B1 (en) * 1998-03-11 2002-12-24 University Of Southern California Method of promoting production of living tissue equivalents
US6475988B1 (en) * 1998-05-11 2002-11-05 University Of Southern California Methods to increase white blood cell survival after chemotherapy
US6566335B1 (en) * 1998-05-11 2003-05-20 University Of Southern California Methods for mobilizing hematopoietic progenitor cells from bone marrow into peripheral blood in a patient in need of chemotherapy
US6762167B1 (en) * 1998-05-11 2004-07-13 University Of Southern California Methods for treating a patient undergoing chemotherapy
US6258778B1 (en) * 1998-07-13 2001-07-10 University Of Southern California Methods for accelerating bone and cartilage growth and repair
US6177407B1 (en) * 1998-08-13 2001-01-23 University Of Southern California Methods to increase blood flow to ischemic tissue
US6821953B1 (en) * 1999-12-16 2004-11-23 University Of Southern California Methods for treating and preventing damage to mucosal tissue
US6747008B1 (en) * 2000-06-19 2004-06-08 University Of Southern California Methods for treating and preventing alopecia

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090069246A1 (en) * 1997-01-28 2009-03-12 University Of Southern California Methods for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US7744927B2 (en) 1997-01-28 2010-06-29 University Of Southern California Methods of promoting hematopoietic and mesenchymal cell proliferation and differentiation
US7523131B2 (en) * 2005-02-10 2009-04-21 Oracle International Corporation Techniques for efficiently storing and querying in a relational database, XML documents conforming to schemas that contain cyclic constructs
US9259443B2 (en) 2010-10-25 2016-02-16 The Children's Hospital Of Philadelphia Compositions and methods for the generation of platelets and methods of use thereof
US20130316361A1 (en) * 2010-12-06 2013-11-28 Thd S.P.A. Method for the diagnosis of a carcinoma and uses thereof
US9347953B2 (en) * 2010-12-06 2016-05-24 Thd S.P.A. Method for the diagnosis of a carcinoma and uses thereof
US20160118821A1 (en) * 2014-10-22 2016-04-28 Mitsumi Electric Co., Ltd. Battery protection circuit, battery protection apparatus, and battery pack
US20170357603A1 (en) * 2016-06-14 2017-12-14 Oracle International Corporation System and method for filtering field programmable gate array input/output

Also Published As

Publication number Publication date
US7744927B2 (en) 2010-06-29
MXPA99007026A (en) 2004-08-31
JP2002500624A (en) 2002-01-08
JP3662029B2 (en) 2005-06-22
JP4171727B2 (en) 2008-10-29
WO1998032457A2 (en) 1998-07-30
WO1998032457A3 (en) 1998-11-12
US7118748B1 (en) 2006-10-10
AU737058B2 (en) 2001-08-09
US20090069246A1 (en) 2009-03-12
JP2005160472A (en) 2005-06-23
AU6648598A (en) 1998-08-18
EP0973539A2 (en) 2000-01-26
US6335195B1 (en) 2002-01-01
CA2278737A1 (en) 1998-07-30

Similar Documents

Publication Publication Date Title
US6335195B1 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
US6248587B1 (en) Method for promoting mesenchymal stem and lineage-specific cell proliferation
US6239109B1 (en) Method of promoting erythropoiesis
US7776828B2 (en) Radiation therapy methods
US6455500B1 (en) Radiation therapy methods
EP1047441A1 (en) Method for promoting hematopoietic and mesenchymal cell proliferation and differentiation
CA2659664C (en) Improved radiation therapy methods
AU2680999A (en) Method of promoting embryonic stem cell proliferation
EP1410801A1 (en) Method for promoting hematopoietic cell proliferation and differentiation
WO1999039743A2 (en) Method of promoting hepatic cell proliferation
MXPA00007509A (en) Method of promoting erythropoiesis
MXPA00008070A (en) Method of promoting embryonic stem cell proliferation
MXPA00008843A (en) Improved radiation therapy methods

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION