WO1997018306A1 - Crf analogs and their use in photoaffinity labeling of crf receptors - Google Patents

Crf analogs and their use in photoaffinity labeling of crf receptors Download PDF

Info

Publication number
WO1997018306A1
WO1997018306A1 PCT/EP1996/005011 EP9605011W WO9718306A1 WO 1997018306 A1 WO1997018306 A1 WO 1997018306A1 EP 9605011 W EP9605011 W EP 9605011W WO 9718306 A1 WO9718306 A1 WO 9718306A1
Authority
WO
WIPO (PCT)
Prior art keywords
crf
analog
binding
receptor
protein
Prior art date
Application number
PCT/EP1996/005011
Other languages
French (fr)
Inventor
Joachim Spiess
Andreas RÜHMANN
Andreas Köpke
Original Assignee
MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
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 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. filed Critical MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
Priority to EP96939046A priority Critical patent/EP0866856A1/en
Priority to JP9518589A priority patent/JP2000500968A/en
Publication of WO1997018306A1 publication Critical patent/WO1997018306A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/57509Corticotropin releasing factor [CRF] (Urotensin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/575Hormones
    • G01N2333/5751Corticotropin releasing factor [CRF] (Urotensin)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/72Assays involving receptors, cell surface antigens or cell surface determinants for hormones
    • G01N2333/726G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH

Definitions

  • the present invention refers to CRF or analogs thereof bearing a photoactivatable moiety and a label and their use in detecting CRF receptors and binding proteins and identifying and characterizing their binding site.
  • Corticotropin-releasing factor appears to integrate the endocrine, autonomic, immunologic and behavioral responses to stress in the CNS.
  • the 41 residue polypeptide (1) was originally characterized on the basis of its hypophysiotropic activity stimulating the release of adrenocorticotropic hormone (ACTH) (2), which is known to stimulate the secretion of glucocorticoids from the adrenal cortex.
  • ACTH adrenocorticotropic hormone
  • CRF is the main regulator of the hypothalamus-pituitary-adrenal (HPA) axis leading to the release of glucocorticoids after exposure to stress.
  • CRF CRF receptor protein
  • CRF exhibits its activity through G protein-coupled receptors.
  • CRF receptor, type 1 (CRFR1) mainly found in pituitary and brain was independently cloned from human, mouse and rat brain, and a human Cushing's corticotropic cell tumor (3-6).
  • urocortin Ucn
  • CRF binding protein Besides the CRF receptor, a 37 kDa CRF binding protein has been characterized. This protein which is not homologous to any known splice variant of CRFRl or CRFR2 was demonstrated to bind human/rat CRF (h/rCRF) but not ovine CRF (oCRF) with high affinity (12).
  • Labeling through monofunctional photoaffinity probes is expected to provide higher yields than labeling with chemical cross-linking methods using bifunctional reagents. Additionally, photoactivation is assumed to be superior over thermal activation, because highly reactive species such as carbenes and nitrenes can be selectively formed after irradiation uder mild conditions. The carbenes or nitrenes formed can insert into X-H bonds and thereby attack groups that are normally inert to chemical affinity labeling (21).
  • PAL photoaffinity labeling
  • the technical problem underlying the present invention is to provide CRF or analogs thereof which bind efficiently and with high affinity to the receptor resulting in an irreversible labeling of the receptor.
  • the present invention provides CRF or analogs thereof bearing a photoactivatable moiety and a label.
  • analog encompasses any variant or fragment of CRF which retains CRF ligand binding activity.
  • the photoactivatable moiety and the label are adjacent to each other.
  • the photoactivatable moiety should preferably be of such quality that the photoaffinity labeling can be performed under mild conditions at a suitable wavelength.
  • Examples of the photoactivatable moiety are the 4-(1-azi-2,2,2-trifluoroethyl)-benzoyl residue or the phenylalanine analog thereof.
  • the label can be a radioactive marker, e.g. 125 I, or a fluorescent marker, e.g. fluorescein, or via biotin which interacts with avidin carrying a fluorescent group.
  • a radioactive marker e.g. 125 I
  • a fluorescent marker e.g. fluorescein
  • Preferred embodiments of the invention are a CRF agonist, 4-(1-azi-2,2,2-trifluoroethyl)benzoyl-[ 125 I]-tyrosine 0 oCRF (compound 3), and CRF antagonists based on the amino acid sequence of astressin carrying the 4-(1-azi- 2,2,2-trifluoroethyl)-benzoyl (ATB) residue and a histidine or tyrosine by choice for specific radiolabeling, e.g.
  • ATB-cyclo(30-33) [ 125 I-His 13 , Nle 21,38 , Glu 30 , Ala 32 ,Lys 33 ]h/rCRF-(13-41) (compound 6) and ATB-cyclo(30- 33) [Nle 21,38 , Glu 30 , 125 I-Tyr 32 , Lys 33 ]h/rCRF-(13-41) (compound 7).
  • the synthesis of the compounds of the invention can be performed by linking the photoactivatable moiety, e.g. ATB, to the CRF or CRF analog and subsequent labeling, e.g. iodination.
  • the synthesis of compounds 4 and 5 is performed by linking 4-(1-Azi-2,2,2-trifluoroethyl)benzoic acid to cyclo(30-33) [Nle 21,38 , Glu 30 , Ala 32 , Lys 33 ]h/rCRF- (13-41) and cyclo(30-33) [Nle 21,38 , Glu 30 , Tyr 32 , Lys 33 ]h/rCRF-(13-41).
  • Cyclization of the peptides on the resin prior to coupling of the phenyldiazirine to the N-terminus of the peptides is chosen because of the probable sensitivity of the diazirine group towards tetrakistriphenylphosphine palladium (0) (23). Subsequent iodination with 125 I at histidine 13 or tyrosine 32 furnishes compounds 6 and 7 with a specific activity of 82 TBq/mmol, respectively.
  • the [ 125 I]Tyr 0 oCRF analog bears the 4-(1-azi-2,2,2-trifluoroethyl)benzoyl residue at its N-terminus, where the disturbance of ligand binding is supposed to be minimal (2, 24, 25).
  • the immediate proximity of the photoactivatable part to the radioactive tracer in the molecule facilitates the identification and purification of peptide fragments after photoaffinity labeling experiments.
  • CRF-R1 with a molecular weight of approximately 75kDa was detected with the new CRF analog in HEK 293 cells, permanently transfected with the CRFR1 gene.
  • the compounds of the invention can be used for the detection of CRF receptors and binding proteins and for the identification of the binding site of these proteins.
  • the photoaffinity labeling technique of the present invention is advantageous towards chemical cross-linking methods when identifying the ligand binding site within a receptor molecule as on irradiation of the photoactivatable ligand, a highly reactive short living species is formed, which then irreversibly binds with high yield to its receptor.
  • the affinity tagged receptor polypeptide identified by the label is stable so that it can be further purified, e.g. by HPLC. It can then be cleaved into fragments, and the binding site can be identified by amino acid sequence analysis.
  • FIG. 1 Synthetic route for the photoactivatable diazirine 1 according to document (26) and its linkage to Tyr 0 oCRF 1-41 to generate 2 and its 125 iodinated analog 3.
  • FIG. 2. (A.) Displacement of [ 125 I-Tyr°]oCRF bound to membranes from transfected HEK 293 cells by oCRF ( ⁇ ) or ovine photoCRF 2 ( ⁇ ). Data are the mean of triplicates of a representative experiment. (Inset) Scatchard plots of the binding of oCRF ( ⁇ ) and ovine photoCRF 2 ( ⁇ ).
  • Fig. 3 Stimulation of intracellular cAMP accumulation in Y79 cells by oCRF ( ⁇ ), ovine photoCRF 2 ( ⁇ ), and ovine photoCRF 2 (x) in the presence of 100 nM recombinant human [D-Phe 12 , Nle 21,38 ]CRF-(12-41). Data is the mean ⁇ SEM values (bars) of duplicates of a representative experiment.
  • FIG. 4 Photoaffinity cross-linking of ovine 125 I-photoCRF 3 to HEK 293 cell membrane homogenates. Lanes: 1-5, extracts of cells stably transfected with cDNA coding for rCRFRl; 6 and 7, extracts of nontransfected HEK 293 cells. Radioactive ovine photoCRF was bound in the absence of oCRF (lanes 1, 5, and 6) or in the presence of 100 nM (lane 2), 1 ⁇ M (lane 3), 10 ⁇ M (lanes 4 and 7) oCRF or 1 ⁇ M vasoactive intestinal peptide (lane 5).
  • FIG 5. (A) Plot of radioactivity of membrane components covalently labeled with ovine 125 I-photo CRF 3 and purified with RPHPLC. (B) Pooled fractions were subjected to SDS/PAGE in 7.5% gels.
  • a permanent cell line was established from HEK 293 cells stably transfected with cDNA coding for rCRFRl.
  • a pool of HEK cell clones was employed in the following experiments. Binding results obtained with individual HEK cell clones did not differ significantly from the results of binding experiments with the cell clone pool. Scatchard analysis indicated that oCRF was bound with a K d value of 7.8 ⁇ 6.3 nM at a high-affinity site and a K d value of 137 ⁇ 90 nM at a low-affintiy site.
  • EC50 values of 0.5 ⁇ 0.2 nM and 0.4 ⁇ 0.1 nM were determined for oCRF and photoCRF, respectively (Fig. 2B).
  • Non-transfected cells did not show significant binding or cAMP accumulation. This observation was also confirmed by photoaffinity-labeling experiments.
  • Ovine 125 I-photoCRF 3 did not bind to membranes of non-transfected HEK 293 cells (see Fig. 4).
  • a K i value of 101 ⁇ 92 nM, 51 ⁇ 52 nM and 497 ⁇ 72 nM for astressin and compounds 4 and 5 were obtained.
  • Non-transfected cells did not show significant binding or cAMP accumulation. This observation was also confirmed by photoaffinity labeling experiments.
  • Compound 7 did not bind to membranes of non-transfected HEK 293 cells.
  • Statistical analysis of the described binding and cAMP data with the program ANOVA revealed no significant differences between the K d and K i values for astressin and compound 4. Both peptides exhibited high potency to reduce the stimulatory potency of oCRF to produce cAMP in transfected HEK 293 cells and Y79 cells.
  • the freshly prepared tracer 7 was stored free of any carrier protein, and the photoaffinity labeling experiments were performed in buffer solutions in the absence of BSA.
  • a 66 kDa cross-link was identified with SDS PAGE after irradiation at 360 nm of a mixture of compound 7 and membranes of HEK 293 cells permanently transfected with rCRFR1. No cross-link could be identified without light activation at 360 nm.
  • Binding of compound 7 to the receptor could be efficiently inhibited by addition of 1 ⁇ M ATB-cyclo(30-33) [Nle 21,38 , Glu 30 , Tyr 32 , Lys 33 ]h/rCRF-(31-41) (compound 5) but not 1 ⁇ M vasoactive intestinal peptide (VIP) in agreement with the assumed specificity of this photoprobe.
  • VIP vasoactive intestinal peptide
  • the compounds of the invention can be used for the specific irreversible labeling and tracking of receptors in various tissue membranes, of CRF binding proteins, as well as in cytological investigations using a fluorescent analog of 2, 4 or 5, e.g. on cell sorting, receptor internalization, trafficking.
  • the resin was filtered off, washed three times with 0.5 ml of NMP, added to 750 ⁇ l of cleavage mixture (75 ⁇ g of crystalline phenol, 25 ⁇ l of EDT, 50 ⁇ l of thioanisole and 50 ⁇ l of dH 2 O, 1 ml of TFA) and stirred for 1.4 hr.
  • the resin was filtered off and the peptide precipitated in 20 ml of ice cold ether. After filtration, the crude peptide was dissolved in 2 ml of TFA and 50 ml of 20% MeCN in 0.1 % TFA/water and lyophilized.
  • the mixture was pipetted onto a Bond Elut C 18 cartridge (Varian Associates), prewetted with 5 ml of MeOH, then 5 ml of 0.1 % TFA/water.
  • Five milliliters of dH 2 O followed by 5 ml of 0.1 % TFA/water were passed through the column in order to separate the iodinated peptide from free iodine and BSA.
  • the iodinated peptide was then eluted from the column by the addition of 5 ml of 80% MeCN in 0.1 % of TFA/H 2 O.
  • the specific activity of the peptide 82 TBq/mmol.
  • the peak tubes of radioactivity were pooled and ⁇ -mercaptoethanol was added to a final concentration of 0.5 M.
  • the iodinated tracer 3 (Fig. 1) was stored in aliquots at -20°C and typically used for binding assays and photoaffinity labeling experiments for 2 months.
  • the CRF peptides were synthesized with Fmoc chemistry on TentaGel S RAM resin (0.1 mmole scale, Rapp, Tubingen, F.R.G.) with a model ABI 433A peptide synthesizer (Applied Biosystems). After cleavage of the peptides from the resin, the crude peptides were purified by preparative reverse-phase HPLC (RPHPLC) performed on a Waters Prep Nova-Pak HR C 18 silica gel column (5 ⁇ 30 cm, 6- ⁇ m particle size, 6-nm pore size) with a mixture of aqueous 0.1% trifluoroacetic acid (TFA) and MeCN. The mass spectra of the purified peptides were measured with ESI (electrospray ion) MS on a Micromass AutoSpec-T tandem mass spectrometer.
  • ESI electrospray ion
  • the purified CRF peptides were subjected to analytical RPHPLC on a Vydac C 18 silica gel column (0.46 ⁇ 25 cm, 5- ⁇ m particle size, 30-nm pore size) with solvents A (0.1% TFA in water) and B (80% MeCN in 0.1% TFA in water) at a flow rate of 1 ml/min.
  • the samples were eluted with 5% B for 5 min. and then with a linear gradient of 5-95% B in 30 min.
  • the rat CRFRl gene fragment (1284 bp, BamHI, EcII26II fragment) was subcloned into the vector pcDNA3 (Invitrogen, San Diego, Ca, USA).
  • the recombinant plasmid (pCDNA3-rCRF 1 ) was isolated, and purified with the Qiagen plasmid preparation system (Qiagen, Hilden, Germany). The ligation sites were verified by DNA sequence analysis.
  • HEK 293 cells were transfected with pCDNA3-rCRF-R1 utilizing the calcium/BBS transfection method (32). Sixteen hours after transfection, the medium was removed and replaced by selection medium (600 ⁇ g/ml Geneticin in medium). Cells were grown until confluent and split 1:2 with further selection. Following one to two weeks of growing under selection conditions, all cells were geneticin-resistant and grew normally.
  • the cells obtained according to Example 5 were dislodged from the cell culture flasks with a cell scraper into ice cold PBS buffer. The cells were precipitated at 150 g for 10 min. at 4°C, resuspended in 1 ⁇ PBS buffer and recentrifuged. The supernatant was entirely removed and the wet weight of the cell pellet was determined.
  • the cells were suspended in 3 ml/g cells of CRF membrane buffer (50 mM Tris/Cl, 5 mM MgCl 2 , 2 mM EGTA, 500 ⁇ l Trasylol (FBA, New York, USA), 1 mM DTT, pH 7.4) and treated for 10 strokes (each 2 s) with the medium sized polytron tool at power level 5.
  • the nuclei were precipitated for 5 min at 600 g in the cold. The supernatant was carefully removed with a Pasteur pipette and collected on ice. The pellet was reextracted with the same amount of membrane buffer using some strokes of the polytron. The nuclei were again precipitated from this suspension as described.
  • the combined supernatant ⁇ were centrifuged at 10,000 g for 15 min to precipitate the membranes.
  • the pellet was resuspended with 3 ml/g of cells in storage buffer (membrane buffer containing 20% glycerol) with 10 strokes of a glass Teflon homogenizer.
  • a micro BCA assay (Pierce, Rockford, USA) was performed with 2 ⁇ l and 4 ⁇ l of the suspension to estimate the total protein concentration (about 2.5 ⁇ g/ ⁇ l).
  • the membranes were frozen in liquid nitrogen and stored at -70° C until use.
  • Photoaffinity labeling experiments were in principle performed in the same manner as mentioned above except that the incubation buffer used was without BSA.
  • a concentration series of either oCRF (0, 100 nM, 1 ⁇ M, 10 ⁇ M) or VIP (1 ⁇ M) and 180,000 cpm of 3 per tube HEK 293 membrane homogenates of either transfected or non-transfected cells (75 ⁇ g of protein/tube) were added and incubated for the indicated time. Before photolysis, the pellets were washed three times, resuspended in 300 ⁇ l of buffer and irradiated at 360 nm for 30 min (4°C, 8 cm distance from the lamps).
  • HEK 293 and human Y79 retinoblastoma cells were incubated with different CRF analogs in the presence of 1 or 5 mM 3-isobutyl-1-methylxanthine (37°C, 30 min), respectively.
  • the incubation medium of the Y79 cells contained additionally 1 mg/ml BSA and 0.05 mg/ml ascorbic acid. When compound 2 or the photoactivatable astressin analogs were used, all experiments were performed in the dark. After removal of the medium, cells were lyzed with aqueous 6% trichloroacetic acid (100°C, 5 min).
  • the cell lysates were stored at -70°C until assayed with a RIA kit (Amersham, Little Chalfont). Data analysis was achieved with the sigmoidal dose-response curve fitting programs ALLFIT. Statistical significance was determined across groups by one-way ANOVA.
  • Membrane protein (250 ⁇ g) was labeled with 1.1 ⁇ 10 7 cpm of 3 (2.82 pmol).
  • One-tenth of the sample was dissolved in 50% ethanolic formic acid (100 ⁇ l) and subjected to RPHPLC using a Vydac C 4 silica gel column (0.46 ⁇ 25 cm, 5 ⁇ m particle size, 30 nm pore size.) Elution was accomplished with a mixture of aqueous 0.5% trifluoroacetic acid and EtOH.

Abstract

The present invention refers to CRF or analogs thereof bearing a photoactivatable moiety and a label and their use in detecting CRF receptor and binding proteins and identifying their binding site.

Description

CRF Analogs and their use in Photoaffinity Labeling of CRF
Receptors
The present invention refers to CRF or analogs thereof bearing a photoactivatable moiety and a label and their use in detecting CRF receptors and binding proteins and identifying and characterizing their binding site.
Corticotropin-releasing factor (CRF) appears to integrate the endocrine, autonomic, immunologic and behavioral responses to stress in the CNS. The 41 residue polypeptide (1) was originally characterized on the basis of its hypophysiotropic activity stimulating the release of adrenocorticotropic hormone (ACTH) (2), which is known to stimulate the secretion of glucocorticoids from the adrenal cortex. It is generally accepted that CRF is the main regulator of the hypothalamus-pituitary-adrenal (HPA) axis leading to the release of glucocorticoids after exposure to stress.
The various functions of CRF in the endocrine, autonomic and immunologic system are mediated by a variety of receptor proteins. Many of these receptors have been studied and investigations concerning the binding affinity and biopotency of several homologues of the CRF family have been performed.
CRF exhibits its activity through G protein-coupled receptors. CRF receptor, type 1 (CRFR1), mainly found in pituitary and brain was independently cloned from human, mouse and rat brain, and a human Cushing's corticotropic cell tumor (3-6). cDNAs coding for two splice variants of CRF receptor, type 2 (CRFR2a and CRFR2b), were cloned from brain, heart, and skeletal muscle (7-10). Recently, it has been proposed that urocortin (Ucn), a naturally occurring CRF analog, is the endogenous ligand to CRFR2 (11).
Besides the CRF receptor, a 37 kDa CRF binding protein has been characterized. This protein which is not homologous to any known splice variant of CRFRl or CRFR2 was demonstrated to bind human/rat CRF (h/rCRF) but not ovine CRF (oCRF) with high affinity (12). The very potent CRF antagonist astressin, cyclo(30-33) [D-Phe12, Nle21,38, Glu30, Lys33]h/rCRF-(12-41), with its amino acid sequence based on h/rCRF exhibited similar binding affinity to CRFRl as found for h/rCRF but did not bind to the CRF binding protein (13). The biopotency of astressin to inhibit CRF mediated ACTH release in an in vitro pituitary cell culture assay was attributed to the built-in lactam bridge at the end of the presumable α-helical part of the peptide stretching from amino acid 5-36 in h/rCRF (13, 14). Surprisingly, the potency of h/rCRF to stimulate ACTH secretion in pituitary cells was not significantly increased when the same lactam bridge motif was introduced into the peptide. It was therefore assumed that the N-terminus of CRF is responsible for CRF receptor activation and induction of α-helicity along the whole molecule (13) . Recently, in binding studies with COSM6 cells transiently expressing chimeric receptors of rCRFR1 and the rat growth hormone releasing factor receptor (rGRFR), it was shown high affinity binding of astressin to the Nyterminus of rCRFRl (rCRFRN). The CRF peptide agonists h/rCRF and urocortin still produced cAMP production when bound to rCRFRN/rGRFR but to a lower extent than CRF stimulation of rCRFRl (15). A study on the characterization of another seven transmembrane spanning G protein-coupled receptor clearly indicated different binding sites for agonist and antagonist binding of Gonadotropin-releasing hormone by site-directed mutagenesis (16). Considering the numerous important functions of CRF and in order to further investigate agonist and antagonist binding of CRF to its receptors, it would be helpful to identify the amino acid sequence directly involved in CRF binding and to investigate the cell biological fate of the CRF receptor and the binding protein after ligand linkage by means of a CRF analog serving as a label covalently linked to proteins binding CRF with high affinity.
Chemical cross-linking with [125I]Tyr0 oCRF has been proved not to be suitable to characterize the actual binding site since the cross-linking efficiency is very low and subsequent chemical and enzymatic cleavages result in the removal of the label from the cross-linked CRF receptor.
Several CRF receptor cross-links with molecular weights in the range of 58,000-75,000 have been characterized applying bifunctional reagents to membranes of bovine anterior pituitary membranes (17), AtT-20 mouse pituitary tumor cells (18), rat brain, and anterior pituitary (19,20). However, all CRF cross-links reported to date were obtained with an extremely low yield (<1%).
Labeling through monofunctional photoaffinity probes is expected to provide higher yields than labeling with chemical cross-linking methods using bifunctional reagents. Additionally, photoactivation is assumed to be superior over thermal activation, because highly reactive species such as carbenes and nitrenes can be selectively formed after irradiation uder mild conditions. The carbenes or nitrenes formed can insert into X-H bonds and thereby attack groups that are normally inert to chemical affinity labeling (21).
A prerequisite for all experiments using a photoaffinity labeling (PAL) technique is that the photoactivatable ligand binds with high affinity to the receptor and that the receptor is not destroyed or deactivated by the light used to activate the label (21,22). Recently, a new class of photoactivatable compounds, the aryldiazirines, has been introduced, which allows photochemical decomposition under mild conditions (23).
Thus, the technical problem underlying the present invention is to provide CRF or analogs thereof which bind efficiently and with high affinity to the receptor resulting in an irreversible labeling of the receptor.
The solution to said technical problem is provided by the embodiments characterized in the claims.
Accordingly, the present invention provides CRF or analogs thereof bearing a photoactivatable moiety and a label.
In this context, the term "analog" encompasses any variant or fragment of CRF which retains CRF ligand binding activity.
In a specific embodiment the photoactivatable moiety and the label are adjacent to each other.
The photoactivatable moiety should preferably be of such quality that the photoaffinity labeling can be performed under mild conditions at a suitable wavelength. Examples of the photoactivatable moiety are the 4-(1-azi-2,2,2-trifluoroethyl)-benzoyl residue or the phenylalanine analog thereof.
The label can be a radioactive marker, e.g. 125I, or a fluorescent marker, e.g. fluorescein, or via biotin which interacts with avidin carrying a fluorescent group.
Preferred embodiments of the invention are a CRF agonist, 4-(1-azi-2,2,2-trifluoroethyl)benzoyl-[125I]-tyrosine0oCRF (compound 3), and CRF antagonists based on the amino acid sequence of astressin carrying the 4-(1-azi- 2,2,2-trifluoroethyl)-benzoyl (ATB) residue and a histidine or tyrosine by choice for specific radiolabeling, e.g. ATB-cyclo(30-33) [125I-His13, Nle21,38, Glu30, Ala32,Lys33]h/rCRF-(13-41) (compound 6) and ATB-cyclo(30- 33) [Nle21,38, Glu30, 125I-Tyr32, Lys33]h/rCRF-(13-41) (compound 7).
The synthesis of the compounds of the invention can be performed by linking the photoactivatable moiety, e.g. ATB, to the CRF or CRF analog and subsequent labeling, e.g. iodination.
For example, the synthesis of compounds 4 and 5 is performed by linking 4-(1-Azi-2,2,2-trifluoroethyl)benzoic acid to cyclo(30-33) [Nle21,38, Glu30, Ala32, Lys33]h/rCRF- (13-41) and cyclo(30-33) [Nle21,38, Glu30, Tyr32, Lys33]h/rCRF-(13-41). Cyclization of the peptides on the resin prior to coupling of the phenyldiazirine to the N-terminus of the peptides is chosen because of the probable sensitivity of the diazirine group towards tetrakistriphenylphosphine palladium (0) (23). Subsequent iodination with 125I at histidine13 or tyrosine32 furnishes compounds 6 and 7 with a specific activity of 82 TBq/mmol, respectively.
In a preferred embodiment of the invention the [125I]Tyr0oCRF analog bears the 4-(1-azi-2,2,2-trifluoroethyl)benzoyl residue at its N-terminus, where the disturbance of ligand binding is supposed to be minimal (2, 24, 25). The immediate proximity of the photoactivatable part to the radioactive tracer in the molecule facilitates the identification and purification of peptide fragments after photoaffinity labeling experiments. CRF-R1 with a molecular weight of approximately 75kDa was detected with the new CRF analog in HEK 293 cells, permanently transfected with the CRFR1 gene.
The compounds of the invention can be used for the detection of CRF receptors and binding proteins and for the identification of the binding site of these proteins. The photoaffinity labeling technique of the present invention is advantageous towards chemical cross-linking methods when identifying the ligand binding site within a receptor molecule as on irradiation of the photoactivatable ligand, a highly reactive short living species is formed, which then irreversibly binds with high yield to its receptor. The affinity tagged receptor polypeptide identified by the label is stable so that it can be further purified, e.g. by HPLC. It can then be cleaved into fragments, and the binding site can be identified by amino acid sequence analysis.
Brief description of the figures
FIG. 1. Synthetic route for the photoactivatable diazirine 1 according to document (26) and its linkage to Tyr0oCRF 1-41 to generate 2 and its 125iodinated analog 3.
FIG. 2. (A.) Displacement of [ 125I-Tyr°]oCRF bound to membranes from transfected HEK 293 cells by oCRF (●) or ovine photoCRF 2 (□). Data are the mean of triplicates of a representative experiment. (Inset) Scatchard plots of the binding of oCRF (●) and ovine photoCRF 2 (▭).
(B). Stimulation of intracellular cAMP accumulation in transfected HEK 293 cells by oCRF (●) and ovine photoCRF 2 (□). Data are the mean from duplicate of a representative experiment. The error bars represent the SEM and are not shown when smaller than the symbol size.
Fig. 3. Stimulation of intracellular cAMP accumulation in Y79 cells by oCRF (●), ovine photoCRF 2 (▭), and ovine photoCRF 2 (x) in the presence of 100 nM recombinant human [D-Phe12, Nle21,38]CRF-(12-41). Data is the mean ± SEM values (bars) of duplicates of a representative experiment.
FIG. 4. Photoaffinity cross-linking of ovine 125I-photoCRF 3 to HEK 293 cell membrane homogenates. Lanes: 1-5, extracts of cells stably transfected with cDNA coding for rCRFRl; 6 and 7, extracts of nontransfected HEK 293 cells. Radioactive ovine photoCRF was bound in the absence of oCRF (lanes 1, 5, and 6) or in the presence of 100 nM (lane 2), 1 μM (lane 3), 10 μM (lanes 4 and 7) oCRF or 1 μM vasoactive intestinal peptide (lane 5). Fifty micrograms of total membrane protein was labeled with approximately 100,000 cpm of ovine 125I-photoCRF and incubated (37°C, 30 min.) in the presence (lane 9) or absence (lane 8) of 2000 units of PNGase.
FIG 5. (A) Plot of radioactivity of membrane components covalently labeled with ovine 125I-photo CRF 3 and purified with RPHPLC. (B) Pooled fractions were subjected to SDS/PAGE in 7.5% gels.
Agonist binding studies using compounds 2 and 3
A. Preliminary experiments
Preliminary experiments with the diazirine function of 1 were performed in order to optimize the photo-affinity labeling experiments with 3 on CRFR1 transfected HEK 293 cell membranes. The photolysis proceeded with a half-life of 100 s, and after 12 min all diazirine was converted to its carbene or diazo valence isomer (80% carbene, 20% diazo valence isomer (26)). The photolysis was performed at a wavelength of 360 nm using a UV Stratalinker (Stratagene) equipped with five 15 watts lamps and monitored with a UV spectrophotometer (Beckman DU650 spectrometer, Fullerton). At a distance of 14 cm from the lamps, 1 was photolyzed (c = 1 mM in ethanol, V = 380 μl, 1 ml quartz cuvette) with first order kinetic and a half-life of 100 s at 4°C. The same results were obtained when photolyzing 3 after incubation with membranes in different time intervals and analyzing the photoproduct with SDS/PAGE. B. Binding and cAMP assay
For the determination of the binding affinity and the biological potency of ovine photoCRF 2, a permanent cell line was established from HEK 293 cells stably transfected with cDNA coding for rCRFRl. A pool of HEK cell clones was employed in the following experiments. Binding results obtained with individual HEK cell clones did not differ significantly from the results of binding experiments with the cell clone pool. Scatchard analysis indicated that oCRF was bound with a Kd value of 7.8 ± 6.3 nM at a high-affinity site and a Kd value of 137 ± 90 nM at a low-affintiy site. The Bjnax values of 30 fmol/μg and 347 fmol/μg of protein, respectively, indicated a high efficiency of expression. A similar Kd value of 5.6 ± 2.6 nM (Bmax = 12 fmol/μg of protein) was found for ovine photoCRF 2 displacing [125I-Tyr0]oCRF. Scatchard analysis indicated only binding of ovine photoCRF to the high-affinity site (Fig. 2A). Application of oCRF or ovine photoCRF to the transfected HEK 293 cells stimulated the accumulation of cAMP in a dose-dependent manner. EC50 values of 0.5 ± 0.2 nM and 0.4 ± 0.1 nM were determined for oCRF and photoCRF, respectively (Fig. 2B). Non-transfected cells did not show significant binding or cAMP accumulation. This observation was also confirmed by photoaffinity-labeling experiments. Ovine 125I-photoCRF 3 did not bind to membranes of non-transfected HEK 293 cells (see Fig. 4). In experiments with membrane preparations from human Y79 retinoblastoma cells, known to carry an endogenous functional CRF receptor (27), a Kd value of 2 nM (Bmax = 0.19 fmol/μg of protein) was found for oCRF or ovine photoCRF 2. In Y79 cells, only a high-affinity site was detected for either CRF analog. Ovine photoCRF and oCRF stimulated cAMP accumulation in Y79 cells with EC50 values of 2.3 ± 0.5 nM and 1.3 ± 0.6 nM, respectively (Fig. 3). Statistical analysis of the described binding and cAMP data with the program ANOVA revealed no significant differences between the Kd and EC50 values for oCRF and photoCRF. The specificity of the stimulatory action of ovine photoCRF was further demonstrated by the observation that this peptide exhibited lower stimulatory potencies in the presence of the specific CRF antagonist recombinant human [D-Phe12,
Nle21,38]CRF-(12-41). For the antagonist an apparent inhibitory constant (Ki) of 10.3 ± 5.0 nM was found (Fig.
3).
C. Photoaffinity-Labeling Experiments
Since it had been found (17,28) that BSA interferes with the labeling of the receptor, freshly prepared tracer 3 was stored free of any carrier protein, and photoaffinity-labeling experiments were performed in buffer solutions in the absence of BSA. A 75 kDa cross-link was identified with SDS/PAGE after irradiation at 360 nm of a mixture of ovine 125I-photoCRF 3 and membranes of HEK 293 cells permanently transfected with rCRFRl (Fig. 4). No cross-link could be identified without light activation at 360 nm. Using commercially available [125l-Tyr0]oCRF and disuccinimidyltartrate, a 75 kDa protein was labeled in chemical cross-linking experiments. Binding of ovine 125I-photoCRF to the receptor could be efficiently inhibited by addition of 1 μM oCRF but not 1 μM vasoactive intestinal peptide, in agreement with the assumed specificity of this photoprobe. As mentioned above, no photoaffinity cross-linking of ovine 125I-photoCRF to nontransfected HEK 293 membranes was detected. Deglycosylation of the 75 kDa protein cross-link with PNGase generated a 46 kDa protein detected by SDS/PAGE (Fig. 4).
In a preparative photoaffinity-labeling experiment, membrane proteins cross-linked to ovine 125I-photoCRF were purified by RPHPLC. It was found by SDS/PAGE analysis that the radioactive fractions that were eluted after the void volume contained the 75 kDa CRFR1 protein cross-link (Fig. 5). To calculate the yield of the cross-linking procedure, labeled receptor was divided by the radioactivity of ovine 125I-photoCRF specifically bound to the HEK cell membranes that served as starting material. On this basis, a yield of at least 20-30% was estimated.
Antagonist binding studies using compounds 4 to 7
A. Binding and cAMP assay
For the determination of the binding affinity and the biological potency of the photoactivatable CRF antagonists 4 and 5, a HEK 293 cell line, stably transfected with cDNA coding for rCRFR1, and the human Y79 retinoblastoma cell line, expressing an endogenous CRF receptor (CRFR1), were used. The results are shown in Table I. Scatchard analysis indicated high and low affinity binding of oCRF (Kd1 = 1.1 ± 0.7 nM; Kd2 = 1.1 ± 1.3 μM) and astressin (Kd1 = 0.9 ± 1.0 nM; Kd2 = 1.6 ± 1.6 μM) to membrane homogenates of Y79 cells. Compound 4 exhibited similar binding characteristics as astressin (Kd1 = 0.6 ± 0.5 nM; Kd2 = 3.4 ± 2.2 μM). Compound 5 showed decreased binding affinity to CRFRl in this cell line (Kd1 = 26 ± 23 nM). Similar results were obtained when oCRF, astressin and compounds 4 and 5 were bound to membrane homogenates of transfected HEK 293 cells with a Kd value of 3.3 ± 0.5 nM, 7.7 ± 2.6 nM, 3.2 ± 2.7 nM and 12 ± 3.6 nM, respectively. Only oCRF showed binding to a low affinity site with a Kd value of 147 ± 78 nM in this cell line. Application of oCRF to the Y79 cells and HEK 293 cells stimulated the accumulation of cAMP in a dose dependent manner with EC50 values of 3.8 ± 2.6 nM and 0.4 ± 0.1 nM, respectively. Ovine CRF stimulated cAMP production could be efficiently inhibited in the presence of 5 nM antagonist in Y79 cells. An inhibitory constant (Ki) of 0.5 ± 0.3 nM, 1.0 ± 0.3 nM and 6.0 ± 2.8 nM was determined for astressin and compound 4 and 5, respectively. Similar results were obtained when oCRF stimulated cAMP accumulation in transfected HEK 293 cells was inhibited in the presence of 100 nM CRF antagonist. A Ki value of 101 ± 92 nM, 51 ± 52 nM and 497 ± 72 nM for astressin and compounds 4 and 5 were obtained. Application of a higher dosis of CRF antagonist to observe significant reduction of oCRF stimulated cAMP production in HEK 293 was necessary because of a fifty times higher expression of high affinity receptors in transfected HEK 293 cells (oCRF: Bmax1 = 16 ± 6 fmol/μg; Bmax2 = 197 ± 15 fmol/μg) when compared with the Y79 cells (oCRF: Bmax1 = 0.3 ± 0.3 fmol/μg; Bmax2 = 35 ± 57 fmol/μg). Non-transfected cells did not show significant binding or cAMP accumulation. This observation was also confirmed by photoaffinity labeling experiments. Compound 7 did not bind to membranes of non-transfected HEK 293 cells. Statistical analysis of the described binding and cAMP data with the program ANOVA revealed no significant differences between the Kd and Ki values for astressin and compound 4. Both peptides exhibited high potency to reduce the stimulatory potency of oCRF to produce cAMP in transfected HEK 293 cells and Y79 cells. Compound 5, however, revealed 5-10 times lower potency to inhibit cAMP production in both cell lines when compared to astressin or compound 4 which was consistent with its decreased binding affintiy to CRFR1.
B. Photoaffinity labeling experiments
As described above, the freshly prepared tracer 7 was stored free of any carrier protein, and the photoaffinity labeling experiments were performed in buffer solutions in the absence of BSA. A 66 kDa cross-link was identified with SDS PAGE after irradiation at 360 nm of a mixture of compound 7 and membranes of HEK 293 cells permanently transfected with rCRFR1. No cross-link could be identified without light activation at 360 nm. Binding of compound 7 to the receptor could be efficiently inhibited by addition of 1 μM ATB-cyclo(30-33) [Nle21,38, Glu30, Tyr32, Lys33]h/rCRF-(31-41) (compound 5) but not 1 μM vasoactive intestinal peptide (VIP) in agreement with the assumed specificity of this photoprobe. As mentioned above, no photoaffinity cross-linking of compound 7 to non-transfected HEK 293 membranes was detected. Deglycosylation of the 66 kDa protein cross-link with PNGase generated a 38 kDa protein detected by SDS PAGE.
Thus, the compounds of the invention can be used for the specific irreversible labeling and tracking of receptors in various tissue membranes, of CRF binding proteins, as well as in cytological investigations using a fluorescent analog of 2, 4 or 5, e.g. on cell sorting, receptor internalization, trafficking.
The invention is illustrated by the following examples.
Example 1
Synthesis of 4-(1-azi-2,2,2-trifluoroethyl)benzoic acid ( 1) .
In the dark, 420 mg of 4-(1-azi-2,2,2-trifluoroethyl)benzyl alcohol (1.9 mmol; 44 % overall yield starting with 4-bromobenzyl alcohol in a seven step synthesis) (26) was dissolved in 1.4 ml of dioxane and 12 ml of 0.2 N aqueous KOH. Then, KMnO4 (462 mg; 2.9 mmol) was added in portions and the mixture was stirred for 2 hr at ambient temperature. The precipitated Mn02 was removed by filtration, washed several times with methanol and the combined filtrates were concentrated under reduced pressure. The residual alkaline solution was extracted with ether, acidified to pH 2-3 with IN aqueous H2SO4 and extracted again with ether. The organic layer was washed neutral with water, dried with anhydrous Na2SO4 and the solvent was evaporated in vacuo. The product was crystallized from hexane and yielded 230 mg of 1 (1.0 mmol; 53%) :m.p. 123-125°C, decomp. with foam (N2); 1H-NMR (CDCl3, TMS) 7.72 (AABB, 4H, Ar-H); 13C-NMR (CDCl3, TMS) 28.46 (m, J = 41 Hz), 121.85 (m, J = 274 Hz), 126.49 (m, J = 1.3 Hz), 130.32 (m, J = 2.9 Hz), 130.54 (s), 134.78 (s), 170.81 (s); 19F-NMR (DMSO-d6, CFCl3)-64.00; UV (ethanol) λ (ε )348 nm (248); MS m/z (rel. intensity) 229 (100, [M-H]+), 201 (21, [M-N2]+), 157 (51), 137 (8); HRMS calcd. for C9H5N2F3O2 229.0249, found 229.0228.
Example 2
Synthesis of 4-(1-Azi-2,2,2-trifluoroethyl)benzoyl-tyrosine0oCRF 1-41 (2).
In the dark, 26 mg of 1 (0.11 mmol) in 0.2 ml of NMP were activated by 0.2 ml of 0.45 M HBTU/HOBt in DMF (6 min.) and 0.1 ml of 2 M of DIEA in NMP (2 min.). 83 mg of peptide resin (7.00 μmol side chain protected [Tyr0]oCRF 1-41 on TentaGel S RAM resin; capacity 0.22 mmol/g) were added and the mixture was reacted for 15 min. The resin was filtered off, washed three times with 0.5 ml of NMP, added to 750 μl of cleavage mixture (75 μg of crystalline phenol, 25 μl of EDT, 50 μl of thioanisole and 50 μl of dH2O, 1 ml of TFA) and stirred for 1.4 hr. The resin was filtered off and the peptide precipitated in 20 ml of ice cold ether. After filtration, the crude peptide was dissolved in 2 ml of TFA and 50 ml of 20% MeCN in 0.1 % TFA/water and lyophilized. 21 mg of 38 mg crude product was purified by preparative reversed-phase HPLC, and yielded 2.7 mg of 2 (0.54 μmol, 14%):ESI MS calcd. 5045.7; found 5045.1. Analytical RP-HPLC was performed on a Vydac C18 silica gel column (0.46 × 25 cm, 5 μm particle size, 30 nm pore size) with solvents A: 0.1% TFA/water and B: 80% MeCN in 0.1% TFA/water, flow rate: 1 ml/min, 40% B for 5 min, then 40-90% B for 25 min . Rt = 19.62 min). Example 3
Synthesis of 4-(1-Azi-2,2,2-trifluoroethyl)benzoyl-[125I]- tyrosine0oCRF 1-41 (3).
2 was iodinated with slight modifications according to literature (29). To a tube containing 4 μl of a 100 μM solution of 2 in 0.01N HOAc in dH2O, the following reagents were added in a certain order: 10 μl of 0.5 M phosphate buffer, pH 7.4, approximately 20 MBq of 125I (IMS 30, Amersham, UK), 12.5 μg of chloramine T in 5 μl of 0.05 M phosphate buffer, 15 s later the reaction was stopped by adding 10 mg of BSA in 100 μl of 0.5 M phosphate buffer and 1 mg of KI in 100 μl of 0.05 M phosphate buffer. The mixture was pipetted onto a Bond Elut C18 cartridge (Varian Associates), prewetted with 5 ml of MeOH, then 5 ml of 0.1 % TFA/water. Five milliliters of dH2O followed by 5 ml of 0.1 % TFA/water were passed through the column in order to separate the iodinated peptide from free iodine and BSA. The iodinated peptide was then eluted from the column by the addition of 5 ml of 80% MeCN in 0.1 % of TFA/H2O. The volume of the peptide fraction was reduced to approximately 200 μl with a Speed Vac (Christ) and loaded onto a Vydac C18 silica gel column (0.46 × 25 cm, 5 μm particle size, 30 nm pore size) and eluted with solvents A (0.1 % TFA/water) and B (80% MeCN in 0.1 % TFA/water) and a flow rate of 1 ml/min. Elution was performed with 45% B for 5 min, then 45-95% B for 25 min. The retention time for 3 was Rt = 17.36 min. A Beckman 171 Radioisotope Detector equipped with a liquid scintillator flow cell was used. The specific activity of the peptide: 82 TBq/mmol. The peak tubes of radioactivity were pooled and β-mercaptoethanol was added to a final concentration of 0.5 M. The iodinated tracer 3 (Fig. 1) was stored in aliquots at -20°C and typically used for binding assays and photoaffinity labeling experiments for 2 months. Example 4
Synthesis of ovine CRF, cyclo (30-33) [D-Phe12, Nle21,38, Glu30, Lys33] h/rCRF-(12-41) (Astressin), ATB-cyclo(30-33) [Nle21,38, Glu30, Ala32, Lys33]h/rCRF-(13-41) (Compound 4), ATB-cyclo (30-33) [Nle21,38, Glu30, Tyr32, Lys33]h/rCRF- (13-41) (Compound 5)
The CRF peptides were synthesized with Fmoc chemistry on TentaGel S RAM resin (0.1 mmole scale, Rapp, Tubingen, F.R.G.) with a model ABI 433A peptide synthesizer (Applied Biosystems). After cleavage of the peptides from the resin, the crude peptides were purified by preparative reverse-phase HPLC (RPHPLC) performed on a Waters Prep Nova-Pak HR C18 silica gel column (5 × 30 cm, 6-μm particle size, 6-nm pore size) with a mixture of aqueous 0.1% trifluoroacetic acid (TFA) and MeCN. The mass spectra of the purified peptides were measured with ESI (electrospray ion) MS on a Micromass AutoSpec-T tandem mass spectrometer.
For the synthesis of the cyclized CRF analogs, amino acid derivatives Fmoc-Glu(OAl)-OH and Fmoc-Lys (Aloe)-OH (PerSeptive Biosystems GmbH, Hamburg, F.R.G.) were used. The side-chain protected peptides were reacted with Pd [PPh3]4 in HOAc/N-methylaniline/dichloromethane (v/v; 2:1:40) for three hours and then cyclized with HOBt/HBTU in DMF and DIEA in NMP for eight hours. After removal of the N-terminal Fmoc group with piperidine in NMP, 4-(1-azi-2,2,2-trifluoroethyl)benzoic acid was linked to the N-terminus of the peptide resin with HOBt/HBTU in DMF and DIEA in NMP in the dark. The peptides were then cleaved from the resin and purified by preparative RPHPLC. The purified CRF peptides were subjected to analytical RPHPLC on a Vydac C18 silica gel column (0.46 × 25 cm, 5-μm particle size, 30-nm pore size) with solvents A (0.1% TFA in water) and B (80% MeCN in 0.1% TFA in water) at a flow rate of 1 ml/min. The samples were eluted with 5% B for 5 min. and then with a linear gradient of 5-95% B in 30 min. (oCRF: ESI MS calcd 4670.4, found 4669.2, Rt = 25.9 min; astressin: ESI MS calcd 3565.1, found 3563.1, Rt = 24.8 min; 4: ESI MS calcd 3562.1, found 3561.1, Rt = 30.2 min; 5: ESI MS calcd 3654.2, found 3653.7, Rt = 29.6 min).
ATB-cyclo (30-33) [125I-His13, Nle21,38, Glu30, Ala32, Lys33]h/rCRF- (13-41) (Compound 6 ) and ATB-cyclo (30- 33) [Nle21,38, Glu30, 125I-Tyr32, Lys33] h/rCRF-(13-41) (Compound 2)
Compounds 6 and 7 were iodinated as described (29,30). The peptides were partially purified with a Bond Elut C18 cartridge (Analytichem, Harbor City, CA, USA) and subsequently with RPHPLC performed on a Vydac C18 silica gel column (0.46 × 25 cm, 5 μm particle size, 30 nm pore size) with solvents A (0.1% TFA in water) and B (80% MeCN in 0.1% TFA in water) at a flow rate of 1 ml/min. The samples were eluted with 45% B for 5 min. and then with a linear gradient of 45-95% B in 25 min (6: Rt = 21.9 min; 7:Rt = 20.4 min). A Beckman 171 Radioisotope Detector equipped with a liquid scintillation flow cell (Beckman, Fullerton, CA, USA) was used to monitor radioactivity. The specific activity of the peptides was 82 TBq/mmol.
Example 5
Transfection of HEK 293 cells
Human embryonic kidney cells 293 (Graham, Smiley, Russell & Nairn, 1977) (supplied by Dr. C. Stevens and G. Sharraa, The Salk Institute, La Jolla) were grown in Dulbecco's modified eagle medium (GIBCO BRL, Gaitherburg, MD, USA, cat. no.: 041-01885M) supplemented with 10% fetal calf serum (Sigma, St. Louise, MO, USA, cat. no.: F-7524) and brought to a final concentration of 4 mM L-glutamine (GIBCO BRL, cat. no.: 043-05030), 0.45% glucose. They were maintained as described (31). The rat CRFRl gene fragment (1284 bp, BamHI, EcII26II fragment) was subcloned into the vector pcDNA3 (Invitrogen, San Diego, Ca, USA). The recombinant plasmid (pCDNA3-rCRF1) was isolated, and purified with the Qiagen plasmid preparation system (Qiagen, Hilden, Germany). The ligation sites were verified by DNA sequence analysis.
HEK 293 cells were transfected with pCDNA3-rCRF-R1 utilizing the calcium/BBS transfection method (32). Sixteen hours after transfection, the medium was removed and replaced by selection medium (600 μg/ml Geneticin in medium). Cells were grown until confluent and split 1:2 with further selection. Following one to two weeks of growing under selection conditions, all cells were geneticin-resistant and grew normally.
Example 6
Preparation of Crude Membranes
The cells obtained according to Example 5 were dislodged from the cell culture flasks with a cell scraper into ice cold PBS buffer. The cells were precipitated at 150 g for 10 min. at 4°C, resuspended in 1 × PBS buffer and recentrifuged. The supernatant was entirely removed and the wet weight of the cell pellet was determined. The cells were suspended in 3 ml/g cells of CRF membrane buffer (50 mM Tris/Cl, 5 mM MgCl2, 2 mM EGTA, 500 μl Trasylol (FBA, New York, USA), 1 mM DTT, pH 7.4) and treated for 10 strokes (each 2 s) with the medium sized polytron tool at power level 5. The nuclei were precipitated for 5 min at 600 g in the cold. The supernatant was carefully removed with a Pasteur pipette and collected on ice. The pellet was reextracted with the same amount of membrane buffer using some strokes of the polytron. The nuclei were again precipitated from this suspension as described. The combined supernatantε were centrifuged at 10,000 g for 15 min to precipitate the membranes. The pellet was resuspended with 3 ml/g of cells in storage buffer (membrane buffer containing 20% glycerol) with 10 strokes of a glass Teflon homogenizer. A micro BCA assay (Pierce, Rockford, USA) was performed with 2 μl and 4 μl of the suspension to estimate the total protein concentration (about 2.5 μg/μl). The membranes were frozen in liquid nitrogen and stored at -70° C until use.
Example 7
Binding assays with oCRF, astressin and compounds 2 , 4. and 5
To a tube containing the peptides (c = 0-1 μM) and 100,000 or 200,000 cpm, respectively, of [125I-Tyr0]oCRF in 200 μl incubation buffer (membrane buffer supplemented with BSA to 1 mg/ml), 100 μl of membrane suspension containing 25 μg of protein (HEK 293 cells) or 100 μg of protein (Y79 cells) was added. After incubation (1 hr, 23°C), membrane buffer (1 ml) was added. After centrifugation at 14,000 × g (4°C, 5 min), the pellet was washed twice with 1 ml of membrane buffer. Radioactivity was measured with a 1470 WIZARD automatic gamma counter (Berthold, Hannover). Data analysis was achieved with the non-linear curve fitting program LIGAND.
Example 8 a) Photoaffinity labeling experiments with 3
Photoaffinity labeling experiments were in principle performed in the same manner as mentioned above except that the incubation buffer used was without BSA. To a concentration series of either oCRF (0, 100 nM, 1 μM, 10 μM) or VIP (1 μM) and 180,000 cpm of 3 per tube, HEK 293 membrane homogenates of either transfected or non-transfected cells (75 μg of protein/tube) were added and incubated for the indicated time. Before photolysis, the pellets were washed three times, resuspended in 300 μl of buffer and irradiated at 360 nm for 30 min (4°C, 8 cm distance from the lamps). After photolysis, 1 ml of buffer was added and the pellets were spinned out at 15,000 rpm for 5 min. The pellet was resuspended in 15 μl of dH2O and 15 μl of 2xSDS sample buffer and heated at 100°C for 5 min. The samples were subjected to electrophoresis in a 7.5% SDS gel and autoradiography developed on a BAS-IP NP 2040P imaging plate with a Fujix BAS 2000 scanner (Raytest). Apparent molecular masses were estimated from gel mobilities relative to those of commercial markers (SDS-PAGE high range markers, BioRad). Gel documentation was performed with the programs TINA (Straubenhardt) and WINCAM (Cybertech). b) Photoaffinity labeling experiments with compound 7
The photoaffintiy labeling experiments were carried out like the binding assay except that no BSA was use. Samples (25 μg of protein/tube) were irradiated at 360 nm for 30 min (4°C, 8 cm distance from the lamps) after incubation with ligand (1 hr, 23°C). In some experiments the photolabeled receptor was deglycosylated with PNGase (New England Biolabs, Schwalbach). Samples were then heated (100°C, 5 min) and subjected to SDS PAGE. Autoradiography was carried out on a BAS-IP NP 2040P imaging plate. Radioactivity was monitored with a Fujix BAS 2000 scanner (Raytest, Straubenhardt). Gel documentation was accomplished with the program TINA (Raytest).
Example 9 cAMP stimulation
HEK 293 and human Y79 retinoblastoma cells (American Type Cell Culture, Rockville) were incubated with different CRF analogs in the presence of 1 or 5 mM 3-isobutyl-1-methylxanthine (37°C, 30 min), respectively. The incubation medium of the Y79 cells contained additionally 1 mg/ml BSA and 0.05 mg/ml ascorbic acid. When compound 2 or the photoactivatable astressin analogs were used, all experiments were performed in the dark. After removal of the medium, cells were lyzed with aqueous 6% trichloroacetic acid (100°C, 5 min). The cell lysates were stored at -70°C until assayed with a RIA kit (Amersham, Little Chalfont). Data analysis was achieved with the sigmoidal dose-response curve fitting programs ALLFIT. Statistical significance was determined across groups by one-way ANOVA.
Example 10
Purification and characterization of the 75 kDa Protein Cross-Link
Membrane protein (250 μg) was labeled with 1.1 × 107 cpm of 3 (2.82 pmol). One-tenth of the sample was dissolved in 50% ethanolic formic acid (100 μl) and subjected to RPHPLC using a Vydac C4 silica gel column (0.46 × 25 cm, 5 μm particle size, 30 nm pore size.) Elution was accomplished with a mixture of aqueous 0.5% trifluoroacetic acid and EtOH.
Figure imgf000023_0001
References
1) Spiess, J., Rivier, J., Rivier, C. & Vale, W. (1981) Proc. Natl. Acad. Sci. USA 78, 6517-6521
2) Vale, W., Spiess, J., Rivier, C. & Rivier, J. (1981) Science 213, 1394-1397.
3) Vita, N., Laurent, P., Lefort, S., Chalon, P., Lelias, J.-M., Kaghad, M., Le Fur, G., Caput, D. & Ferrara, P.
(1993), FEBS Lett. 335, 1-5.
4) Chen, R., Lewis, K.A., Perrin, M. H. & Vale, W. (1993) Proc. Natl. Acad. Sci. USA 90, 8967-8971.
5) Perrin, M. H., Donaldson, C. J., Chen, R., Lewis, K. A.
& Vale, W. (1993) Endocrinology 133, 3058-3061.
6) Chang, C.P., Pearse II, R.V., O'Connell, S. & Rosenfeld, M.G. (1993) Neuron 11, 1187-1195.
7) Lovenberg, T. W., Liaw, C. W., Grigoriadis, D. E., Clevenger W., Chalmers, D. T., De Souza, E. B. & Oltersdorf, T. (1995) Proc. Natl. Acad. Sci. USA 92, 836-840.
8) Perrin, M., Donaldson, C., Chen, R., Blount, A., Berggren, T., Bilezikjian, L., Sawchenko, P. & Vale w. (1995) Proc. Natl. Acad. Sci. USA 92, 2969-2973.
9) Kishimoto, T., Pearse II, R.V., Lin, C. R. & Rosenfeld M. G. (1995) Proc. Natl. Acad. Sci. USA 92, 1108-1112.
10) Stenzel, P., Kesterson, R., Yeung, W., Cone, R. D., Rittenberg, M. B. & Stenzel-Poore, M. P. (1995) Molecular Endocrinology 9, 637-645. 11) Vaughan, J., Donaldson, C., Bittencourt, J., Perrin, M. H., Lewis, K., Sutton, S., Chan, R., Turnbull, A. V., Lovejoy, D., Rivier, C., Rivier, J., Sawchenko, P. E. & Vale, W. (1995) Nature 378, 287-292.
12) Sutton, S. W., Behan, D. P., Lahrichi, S. L., Kaiser, R., Corrigna, A., Lowry, P., Potter, E., Perrin, M. H., Rivier, J. & Vale, W. W. (1995) Endocrinology 136, 1097- 1102.
13) Gulyas, J., Rivier, C., Perrin, M., Koerber, S. C, Sutton, S., Corrigan, A., Lahrichi, S. L., Craig, A. G., Vale W. & Rivier, J. (1995) Proc. Natl. Acad. Sci. USA 92, 10575-10579.
14) Lovejoy, D.A. (1996) Biochem. Cell. Biol. 74, 1-7.
15) Perrin, M. H., Sutton, S. W., Berggren, W. T. & Vale, W.
W. (1996) Society for Neuroscience 22, poster 609.9.
16) Zhou, Wei, Rodic, V., Kitanovic, S., Flanagan, C. A., Chi, L., Weinstein, H., Maayani, S., Millar, R. P. & Sealfon S. C. (1995) J. Biol. Chem. 270, 18853-18857.
17) Nishimura, E., Billestrup, N., Perrin, M., & Vale, W.
(1987) J. Biol. Chem. 262, 12893-12896.
18) Rosendale, B. E., Jarrett, D. B. & Robinson, A. G.
(1987) Endocrinology 120, 2357-2366.
19) Grigoriadis, D. E. & DDe Souza E. B. (1988) J. Biol.
Chem. 263, 10927-10931.
20) Grigoriadis, D. E. & De Souza E. B. (1989) Endocrinology 125, 1877-1888. 21) Schuster, D. I., Probst, W. C., Ehrlich, G. K. & Singh, G. (1989) Photochem. Photobiol. 49, 785-804.
22) Guillory, R. J. (1989) Pharmac. Ther. 41, 1-25.
23) Bayley, H. (1987) in Chemistry of Diazirines, ed. Liu, M. T. H. (CRC Press, Boca Raton, FL), Vol. 2, pp. 75-99.
24) Rivier, J., Spiess, J. & Vale W. (1983) Proc. Natl.
Acad. Sci. USA 80, 4851-4855.
25) Rivier, J., Rivier, C. & Vale, W. (1984) Science 224, 889-891.
26) Nassal, M. (1983) Liebigs Ann. Chem. 1510-1523.
27) Olianas, M. C., Lampis, G. & Onali, P. (1995) J.
Neurochem. 64, 402-407
28) Rύhmann, A., Kόpke, A. K. E., Dautzenberg, F. M. & Spiess J. (1996) Proc. Natl. Acad. Sci. USA 93, 10609- 10613.
29) Rύckert, Y., Rhode, W. & Furkert, J. (1990) Exp. and Clin. Endocrinology 96, 129-137.
30) Vale, W., Vaughan, J., Yamamoto, G., Bruhn, T., Dgouglas, C., DAlton, D., Rivier, C. & Rivier, J. (1983) Meth. in Enzymol. 103, 565-577.
31) Graham, F.L., Smiley, J., Russell, W.C. & Nairn, R.
(1977) Journal of gen. Virology, 36, 59-72.
32) Sambrook, J., Fritsch, E.F. & Maniatis, T. (1989) Molecular Cloning, (Cold Spring Harbor Laboratory Press: Cold Spring Harbor) 2nd Ed., chapter 16.33.

Claims

CLAIMS :
1. A CRF or an analog thereof bearing a photoactivatable moiety and a label.
2. The CRF or analog of claim 1 wherein the photoactivatable moiety and the label are adjacent to each other.
3. The CRF or analog of claim 1 or 2 wherein the photoactivatable moiety is a 4-(1-azi-2,2,2- trifluoroethyl)-benzoyl residue.
4. The CRF or analog of any one of claims 1 to 3 wherein the label is a radioactive marker.
5. The CRF or analog of claim 4 wherein the label is 125I.
6. The CRF or analog of any one of claims 1 to 3 wherein the label is a fluorescent marker.
7. The CRF or analog of any one of claims 1 to 5 which is 4-(1-azi-2,2,2-trifluoroethyl)benzoyl-[125I]- tyrosine0oCRF.
8. The CRF or analog of any one of claimM 1 to 5 which is ATB-cyclo (30-33) [125I-His13, Nle21,38, Glu30, Ala32, Lys33]h/rCRF-(13-41).
9. The CRF or analog of any one of claims 1 to 5 which is ATB-cyclo(30-33) [Nle21,38, Glu30, 125I-Tyr32, Lys33]h/rCFR-(13-41).
10. Use of CRF or an analog thereof of any one of claims 1 to 9 for detecting CRF receptors and binding proteins.
11. The use of CRF or an analog thereof of any one of claims 1 to 9 for the identification of the binding site of a CRF receptor or binding protein.
12. The use of claim 10 or 11 wherein the receptor protein is detected in a tissue membrane.
13. The use of any one of claims 10 to 12 wherein the receptor protein is detected in the membrane of HEK 293 cells.
14. The use of any one of claims 10 to 13 wherein the CRF receptor protein has a molecular weight of 66 or 75 kDa.
15. A process for the purification of a CRF receptor protein which comprises reacting a membrane preparation containing the protein with CRF or an analog thereof of any one of claims 1 to 9, performing photolysis and purifying the resultant product by HPLC.
16. A process for the characterization of the binding site of a CRF receptor or binding protein which comprises purifying the CRF binding protein or the CRF receptor according to the process of claim 15, fragmenting the purified product and determining the amino acid sequence of the relevant fragment.
17. A CRF receptor or binding protein the binding site of which has been identified according to the process of claim 16.
PCT/EP1996/005011 1995-11-14 1996-11-14 Crf analogs and their use in photoaffinity labeling of crf receptors WO1997018306A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP96939046A EP0866856A1 (en) 1995-11-14 1996-11-14 Crf analogs and their use in photoaffinity labeling of crf receptors
JP9518589A JP2000500968A (en) 1995-11-14 1996-11-14 CRF analogs and their use in photoaffinity labeling of CRF receptors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP95117939.9 1995-11-14
EP95117939 1995-11-14

Publications (1)

Publication Number Publication Date
WO1997018306A1 true WO1997018306A1 (en) 1997-05-22

Family

ID=8219815

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1996/005011 WO1997018306A1 (en) 1995-11-14 1996-11-14 Crf analogs and their use in photoaffinity labeling of crf receptors

Country Status (4)

Country Link
EP (1) EP0866856A1 (en)
JP (1) JP2000500968A (en)
CA (1) CA2237548A1 (en)
WO (1) WO1997018306A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002095395A2 (en) * 2001-05-25 2002-11-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Benzophenone-linked crf and crf-like peptides for covalent labeling of corticotropin-releasing factor (crf) binding protein

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2628750A1 (en) * 1988-03-21 1989-09-22 Pasteur Institut VECTOR MODIFIED BY A SEQUENCE ENCODING A SEQUENCE OF AMINO ACIDS CONTAINED IN A MAMMALIAN PROTEIN HAVING A MEMBRANE RECEPTOR BIOLOGICAL ACTIVITY
US4986979A (en) * 1989-03-14 1991-01-22 Neorx Corporation Imaging tissue sites of inflammation
WO1992013074A1 (en) * 1991-01-15 1992-08-06 The Salk Institute For Biological Studies CORTICOTROPIN RELEASING FACTOR BINDING PROTEIN (CRF-bp)
WO1995000640A1 (en) * 1993-06-18 1995-01-05 The Salk Institute For Biological Studies Cloning and recombinant production of crf (corticotropin releasing factor) receptor(s)
WO1996018649A1 (en) * 1994-12-12 1996-06-20 The Salk Institute For Biological Studies Improved cyclic crf agonists

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2628750A1 (en) * 1988-03-21 1989-09-22 Pasteur Institut VECTOR MODIFIED BY A SEQUENCE ENCODING A SEQUENCE OF AMINO ACIDS CONTAINED IN A MAMMALIAN PROTEIN HAVING A MEMBRANE RECEPTOR BIOLOGICAL ACTIVITY
US4986979A (en) * 1989-03-14 1991-01-22 Neorx Corporation Imaging tissue sites of inflammation
WO1992013074A1 (en) * 1991-01-15 1992-08-06 The Salk Institute For Biological Studies CORTICOTROPIN RELEASING FACTOR BINDING PROTEIN (CRF-bp)
WO1995000640A1 (en) * 1993-06-18 1995-01-05 The Salk Institute For Biological Studies Cloning and recombinant production of crf (corticotropin releasing factor) receptor(s)
WO1996018649A1 (en) * 1994-12-12 1996-06-20 The Salk Institute For Biological Studies Improved cyclic crf agonists

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
EBERLE A N: "Peptides containing multiple photolabels: a new tool for the analysis of ligand-receptor interactions", J. RECEPT. RES., 1993, VOL. 13, NO. 1-4, PAGE(S) 27-37, XP000647454 *
EBERLE, ALEX N. ET AL: "Solid phase peptide synthesis and application of photoreactive peptides", INNOVATION PERSPECT. SOLID PHASE SYNTH. COLLECT. PAP., INT. SYMP., 2ND, 1992, PAGES 95-103, XP000646246 *
KOTZYBA-HIBERT F ET AL: "RECENT TRENDS IN PHOTOAFFINITY LABELING", ANGEWANDTE CHEMIE. INTERNATIONAL EDITION, vol. 34, no. 12, 7 July 1995 (1995-07-07), pages 1296 - 1312, XP000510821 *
NASSAL M: "4-(1-Azi-2,2,2-trifluoroethyl)benzoic acid, a highly photolabile carbene generating label readily fixable to biochemical agents", LIEBIGS ANN. CHEM., 1983,, NO. 9, PAGE(S) 1510-23, XP002029660 *
RESEK J F ET AL: "A new photo-crosslinking reagent for the study of protein-protein interactions", J. ORG. CHEM., 1993, VOL. 58, NO. 26, PAGE(S) 7598-601, XP002029659 *
RUEHMANN A ET AL: "Synthesis and characterization of a photoactivatable analog of corticotropin-releasing factor for specific receptor labeling", PROC. NATL. ACAD. SCI. U. S. A., 1996, VOL. 93, NO. 20, PAGE(S) 10609-10613, XP002029661 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002095395A2 (en) * 2001-05-25 2002-11-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Benzophenone-linked crf and crf-like peptides for covalent labeling of corticotropin-releasing factor (crf) binding protein
WO2002095395A3 (en) * 2001-05-25 2003-02-27 Max Planck Gesellschaft Benzophenone-linked crf and crf-like peptides for covalent labeling of corticotropin-releasing factor (crf) binding protein

Also Published As

Publication number Publication date
CA2237548A1 (en) 1997-05-22
JP2000500968A (en) 2000-02-02
EP0866856A1 (en) 1998-09-30

Similar Documents

Publication Publication Date Title
US5846747A (en) Method for detecting glucagon-like peptide-1 antagonists and agonists
Lindskog et al. The novel high-affinity antagonist, galantide, blocks the galanin-mediated inhibition of glucose-induced insulin secretion
WO1993019175A1 (en) Receptor for the glucagon-like-peptide-1 (glp-1)
CA2403447A1 (en) Peptide derivative
US5093233A (en) Antagonists with position 13 modification
Rühmann et al. Synthesis and characterization of a photoactivatable analog of corticotropin-releasing factor for specific receptor labeling.
EP1143000B1 (en) Screening method
KR20010085847A (en) Peptide derivative
Shults et al. Demonstration and distribution of kassinin‐like material (substance K) in the rat central nervous system
Bovy et al. Identification of structural requirements for analogs of atrial natriuretic peptide (ANP) to discriminate between ANP receptor subtypes
US7273710B2 (en) Method for screening MCH receptor antagonist/agonist
WO1997018306A1 (en) Crf analogs and their use in photoaffinity labeling of crf receptors
EP0524173B1 (en) Solubilization and purification of the gastrin releasing peptide receptor
US6238873B1 (en) Methods of screening for agonists and antagonists of the interaction between the human KIAA0001 receptor and ligands thereof
EP0742825B1 (en) Cloning and recombinant production of crf receptor(s)
US20030059856A1 (en) Methods of screening for agonists and agonists of the interaction between the AXOR8 and AXOR52 receptors and ligands thereof
RU2458068C1 (en) ALPHA-CONOTOXIN PnIA ANALOGUE SHOWING HIGH AFFINITY AND SELECTIVITY TO ACETYLCHOLINE-BINDING PROTEIN FROM APLYSIA CALIFORNICA
Bonk et al. Novel high‐affinity photoactivatable antagonists of corticotropin‐releasing factor (CRF) Photoaffinity labeling studies on CRF receptor, type 1 (CRFR1)
RU2455359C1 (en) Peptide having high affinity to type alpha7 human nicotinic receptor and use thereof
JP4780365B2 (en) MCH receptor antagonist / agonist screening method
Eng et al. Purification of peptide hormones from chinchilla pancreas by chemical assay
Weir et al. Characterization of binding sites in rat for A, B and C-type natriuretic peptides
US20040260071A1 (en) Benzophene-linked crf and crf-like peptides for covalent labeling of corticotropin-releasing factor crf binding protein
EP1131335A1 (en) 7tm receptor hlwar77
Lin The interactions of bivalent and trivalent ACTH analogs with ACTH receptors: Potency and mechanisms

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

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

Ref document number: 2237548

Country of ref document: CA

Ref country code: CA

Ref document number: 2237548

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 1997 518589

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1996939046

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1996939046

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1996939046

Country of ref document: EP