WO1997041154A1 - Eosinophil eotaxin receptor - Google Patents

Eosinophil eotaxin receptor Download PDF

Info

Publication number
WO1997041154A1
WO1997041154A1 PCT/US1997/006568 US9706568W WO9741154A1 WO 1997041154 A1 WO1997041154 A1 WO 1997041154A1 US 9706568 W US9706568 W US 9706568W WO 9741154 A1 WO9741154 A1 WO 9741154A1
Authority
WO
WIPO (PCT)
Prior art keywords
receptor
eosinophil eotaxin
eosinophil
nucleic acid
eotaxin
Prior art date
Application number
PCT/US1997/006568
Other languages
French (fr)
Inventor
Bruce L. Daugherty
Julie A. Demartino
Martin S. Springer
Salvatore J. Siciliano
Original Assignee
Merck & Co., Inc.
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
Priority claimed from GBGB9700894.0A external-priority patent/GB9700894D0/en
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Priority to EP97925399A priority Critical patent/EP1012190A4/en
Priority to JP53897097A priority patent/JP3689118B2/en
Priority to CA002252850A priority patent/CA2252850A1/en
Publication of WO1997041154A1 publication Critical patent/WO1997041154A1/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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7158Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for chemokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to an eosinophil eotaxin receptor ("CC CKR3"), in particular, the human eosinophil eotaxin receptor and nucleic acids encoding this receptor.
  • This invention further relates to assays which may be used to screen and identify compounds that bind to the eosinophil eotaxin receptor. Such compounds would be useful in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma.
  • Eosinophils play prominant roles in a variety of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma (for a reviews see e.g. Gleich, G. J., et al., Eosinophils. J. I. Gallin, I. M. Goldstein, R. Snyderman, Eds., Inflammation: Basic Principles and Clinical Correlates (Raven Press, Ltd., New York, 1992) and Seminario, M. C, et al. (1994) Current Opinion in Immunology 6, 860-864).
  • a pivotal event in the process is the accumulation of eosinophils at the involved sites.
  • chemokines a family of 8-10 kDa proteins are more restricted in the leukocyte subtypes they target and are potential candidates for the recruitment of eosinophils in atopic diseases and asthma (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179).
  • ⁇ -chemokines particularly eotaxin and RANTES, the receptor which mediates these actions has not been identified.
  • the chemokines contain four conserved cysteines, and are divided into two sub-families based on the arrangement of the first cysteine pair (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179).
  • ⁇ -chemokine family which includes IL-8, MGS A, NAP-2 and IP- 10
  • these two cysteines are separated by a single amino acid
  • RANTES regulated on activation T expressed and secreted
  • MCP-1 monocyte chemotactic protein
  • MCP-2 monocyte chemotactic protein
  • MCP-3 macrophage inflammatory protein
  • MlP-l ⁇ macrophage inflammatory protein
  • eotaxin eotaxin
  • the ⁇ -chemokines act primarily on neutrophils, and the ⁇ -chemokines on monocytes, lymphocytes, basophils and eosinophils (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179).
  • RANTES, MCP-2, MCP-3, and MlP-l ⁇ have been shown to activate eosinophils in vitro (Dahinden, C. A., et al. (1994) Journal of Experimental Medicine 179, 751-756; Ebisawa, M., et al. (1994) Journal of Immunology 153, 2153-2160; Weber, M., et al.
  • Eosinophils are attracted by a number of ⁇ -chemokines, the most potent of which are eotaxin (Griffiths- Johnson, D. A., et al. (1993) Bichemical and Biophysical Research Communications 197, 1167- 1172; Jose, P. J., et al. (1994) Journal of Experimental Medicine 179, 881-887; Rothenberg, et al. (1995) Proceedings ofthe National Academy of Sciences 92, 8960-8964) and RANTES (Dahinden, C. A., et al. (1994) Journal of Experimental Medicine 179, 751-756; Ebisawa, M., et al. (1994) Journal of Immunology 153, 2153-2160; Weber, M., et al. (1995) Journal of Immunology 154, 4166-4172;
  • ⁇ -chemokines While elucidation of the actions of ⁇ -chemokines on eosinophils has contributed greatly to the understanding of eosinophil biology, information regarding the cell surface receptors which mediate these effects remain sparse. Furthermore, there are no reports describing binding studies of any of the ⁇ -chemokines to primary eosinophils.
  • the known ⁇ -chemokine receptors are members of the G protein-coupled receptor superfamily. Two of these receptors, CC CKR1 (12, 13) and CC CKR2 (MCP-1R) (Charo, I. F., et al. (1994) Proceecing of the National Academy of Sciences 91, 2752-2756; Myers, S. J., et al.
  • CC CKR1 binds RANTES with high affinity, but binds eotaxin poorly, and while the effects of eotaxin on CC CKR2 have not been studied this receptor has no avidity for RANTES (Myers, S. J., et al. (1995) Journal of Biological Chemistry 270, 5786-5792).
  • a human eotaxin receptor has been reported by Ponath, P.D., et al. J. Exp. Med. 183, 2437-2448 (June 1996) and Gerard, C.J., et al., PCT Publication No. WO 96/22371 (July 25, 1996).
  • the sequence disclosed in this publication possesses an error in the assignment of threonine rather than serine at position # 276 of the receptor.
  • functionality of the receptor was not fully demonstrated.
  • HIV-1 human immunodeficiency virus
  • LAV lymphoid virus
  • HTLV-III human immunodeficiency syndrome
  • ARV advanced immune deficiency syndrome
  • Entry of HIV-1 into a target cell requires cell- surface CD4 and additional host cell cofactors. Fusin has been identified as a cofactor required for infection with virus adapted for growth in transformed T-cells, however, fusin does not promote entry of macrophagetropic viruses which are believed to be the key pathogenic strains of HIV in vivo.
  • chemokine receptors CCR-5 and CXCR-4, as well as the primary receptor CD4
  • the principal cofactor for entry mediated by the envelope glycoproteins of primary macrophage-trophic strains of HIV-1 is CCR5, a receptor for the ⁇ -chemokines RANTES, MlP-l ⁇ and MIP- l ⁇ (Deng, et al., Nature. 381. 661-666 (1996)). HIV attaches to the CD4 molecule on cells through a region of its envelope protein, gpl20.
  • CD-4 binding site on the gpl20 of HIV interacts with the CD4 molecule on the cell surface, and undergoes conformational changes which allow it to bind to another cell-surface receptor, such as CCR5 and/or CXCR-4.
  • CCR5 and/or CXCR-4 another cell-surface receptor
  • ⁇ -chemokine ligands prevent HIV-1 from fusing with the cell (Dragic, et al., Nature. 381. 667-673 (1996)).
  • chemokine receptors may be used by some strains of HIV-1 or may be favored by non-sexual routes of transmission. Although most HIV-1 isolates studied to date utilize CCR-5 or fusin, some can use both as well as the related CCR-2B and CCR-3 as co-receptors (Nature Medicine. 2(11), 1240-1243 (1996)). The determination that chemokine receptors are critical co-receptors for the entry of HIV into cells was pronounced a "1996 Breakthrough of the Year" by Science Magazine (Science. 274. 1987-1991 (Dec. 20, 1996)).
  • orally-active agents which modulate the action of the eosinophil eotaxin receptor would be a significant advance in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma. Further, agents which could block the eosinophil eotaxin receptor in humans who possess normal chemokine receptors should prevent infection in healthy individuals and slow or halt viral progression in infected patients.
  • the present invention relates to a novel receptor which is the eosinophil eotaxin receptor.
  • This receptor is a human ⁇ -chemokine receptor and has been designated "CC CKR3".
  • One aspect of the present invention is directed to the human eosinophil eotaxin receptor, free from receptor-associated proteins.
  • a further aspect of this invention is the human eosinophil eotaxin receptor which is isolated or purified.
  • Another aspect of this invention are eosinophil eotaxin receptors which are encoded by substantially the same nucleic acid sequences, but which have undergone changes in splicing or other RNA processing-derived modifications or mutagenesis induced changes, so that the expressed protein has a homologous, but different amino acid sequence from the native forms.
  • These variant forms may have different and/or additional functions in human and animal physiology or in vitro in cell based assays.
  • the present invention further provides the eosinophil eotaxin receptor, CC CKR3, which is a ⁇ -chemokine receptor and which was cloned from primary eosinophils, and expressed in AML14.3D10 cells.
  • This receptor binds the potent eosinophil attractants, eotaxin, RANTES and MCP-3 with high affinity.
  • eotaxin and RANTES and to a lessor extent MCP-3, induce Ca 2+ -fluxes in cells expressing CC CKR3.
  • Correlation with the binding properties of primary eosinophils provide conclusive evidence that CC CKR3 is the primary endogenous receptor which mediates the effects of ⁇ - chemokines on eosinophils.
  • the present invention further relates to assays which employ a novel receptor which is the eosinophil eotaxin receptor.
  • This receptor is a human ⁇ -chemokine receptor and has been designated "CC CKR3".
  • One aspect of the present invention is directed to assays employing the the human eosinophil eotaxin receptor, free from receptor-associated proteins.
  • a further aspect of this invention is directed to assays which employ the human eosinophil eotaxin receptor which is isolated or purified.
  • the present invention provides assays in which the eosinophil eotaxin receptor is expressed in an AML14.3D10 cell line. DETAILED DESCRIP ⁇ ON OF THE INVENTION
  • the present invention is directed to an eosinophil eotaxin receptor "CC CKR3" which is a G protein-coupled receptor and has been cloned from human eosinophils and which when stably expressed in AML14.3D10 cells binds eotaxin, RANTES and MCP-3 with high affinity.
  • CC CKR3 eosinophil eotaxin receptor
  • Competition binding studies against 125 ⁇ _human eotaxin gives Kd values of 0.1, 2.7, and 3.1 nM, respectively for the three ⁇ - chemokines.
  • CC CKR3 also binds MCP-1 with lower affinity, but does not bind MlP-l ⁇ or MIP-l ⁇ .
  • Competition binding studies on primary eosinophils give binding affinities for the different chemokines which are indistinguishable from those measured with CC CKR3. Since CC CKR3 is prominently expressed in eosinophils it is concluded that CC CKR3 is the eosinophil eotaxin receptor. Eosinophils also express a much lower level of a second chemokine receptor, CC CKR1, which appears to be responsible for the effects of MlP-l ⁇ .
  • the eosinophil eotaxin receptor is a protein containing various functional domains, including one or more domains which anchor the receptor in the cell membrane, and at least one ligand binding domain. As with many receptor proteins, it is possible to modify many of the amino acids, particularly those which are not found in the ligand binding domain, and still retain at least a percentage of the biological activity of the original receptor. In accordance with this invention, it is suggested that certain portions of the eosinophil eotaxin receptor are not essential for its activation by ⁇ -chemokines. Thus this invention specifically includes modified functionally equivalent eosinophil eotaxin receptors which have deleted, truncated, or mutated portions.
  • This invention also specifically includes modified functionally equivalent eosinophil eotaxin receptors which contain modified and/or deletions in other domains, which are not accompanied by a loss of functional activity. Additionally, it is possible to modify other functional domains such as those that interact with second messenger effector systems, by altering binding specificity and/or selectivity. Such functionally equivalent mutant receptors are also within the scope of this invention.
  • a further aspect of this invention are nucleic acids which encode an eosinophil eotaxin receptor or a functional equivalent from human or other species. These nucleic acids may be free from associated nucleic acids, or they may be isolated or purified. For most cloning purposes, cDNA is a preferred nucleic acid, but this invention specifically includes other forms of DNA as well as RNAs which encode an eosinophil eotaxin receptor or a functional equivalent.
  • Yet another aspect of this invention relates to vectors which comprise nucleic acids encoding an eosinophil eotaxin receptor or a functional equivalent.
  • These vectors may be comprised of DNA or RNA; for most cloning purposes DNA vectors are preferred.
  • Typical vectors include plasmids, modified viruses, bacteriophage and cosmids, yeast artificial chromosomes and other forms of episomal or integrated DNA that can encode an eosinophil eotaxin receptor. It is well witnin the skill of the ordinary artisan to determine an appropriate vector for a particular gene transfer or other use.
  • a further aspect of this invention are host cells which are transformed with a gene which encodes an eosinophil eotaxin receptor or a functional equivalent.
  • the host cell may or may not naturally express an eosinophil eotaxin receptor on the cell membrane.
  • the host cells are able to express the eosinophil eotaxin receptor or a functional equivalent on the cell membrane.
  • the receptors of this invention were cloned from RNA isolated from eosinophils.
  • PCR was used with primers designed from both CCCKR1 and CCCKR2, and clones screened by expression in the AML14.3D10 cell line.
  • the cloning was made difficult by several factors. First, prior to this invention there was very little information available about the biochemical characteristics and intracellular signalling/effector pathways used by these receptors making screening procedures uncertain. Second, this receptor could not be expressed and/or functionally coupled in the cell lines normally used for clomng receptors such, as COS, CHO, HEK293. After repeated failures using standard lines, an obscure eosinophilic- like cell line, AML14.3D10, was tried and found to suitable for expression of the receptors described in this invention.
  • the present invention further relates to assays which employ a novel receptor which is the eosinophil eotaxin receptor.
  • This receptor is a human ⁇ -chemokine receptor and has been designated "CC CKR3".
  • One aspect of the present invention is directed to assays employing the the human eosinophil eotaxin receptor, free from receptor-associated proteins.
  • a further aspect of this invention is directed to assays which employ the human eosinophil eotaxin receptor which is isolated or purified.
  • the present invention provides assays in which the eosinophil eotaxin receptor is expressed in an AML14.3D10 cell line.
  • a particular embodiment of this invention is directed to an assay to determine the presence of a compound which binds to the eosinophil eotaxin receptor.
  • this invention also comprises a method to determine the presence of a compound which binds to an eosinophil eotaxin receptor comprising: (a) introducing a nucleic acid which encodes an eosinophil eotaxin receptor into a cell under conditions so that eosinophil eotaxin receptor is expressed;
  • the eosinophil eotaxin receptor is expressed in AML14.3D10 cells.
  • the binding of the compound suspected of binding to the eosinophil eotaxin receptor is compared to the binding or the influence of eotaxin, RANTES and MCP-3.
  • a further embodiment of this invention is directed to an assay to determine the presence of a compound which antagonizes the binding of a known ligand to the eosinophil eotaxin receptor.
  • this invention further comprises a method to determine the presence of a compound which antagonizes the eosinophil eotaxin receptor comprising:
  • the eosinophil eotaxin receptor is expressed in AML14.3D10 cells.
  • the known ligand of the eosinophil eotaxin receptor is eotaxin, RANTES and MCP-3.
  • One aspect of this invention is the development of a sensitive, robust, reliable and high-throughput screening assay which may be used to detect ligands which bind to the eosinophil eotaxin receptor, in particular, antagonists of the action of chemokines on eosinophils.
  • a typical protocol of such an assay is as follows. Assay buffer (50 mM Hepes, pH 7.2 w/ 0.5% BSA, 5 mM MgCl2, 1 mM CaCl2, 100 uM PMSF and 10 ug/ml phosphoramidon, leupeptin, aprotinin and chymostatin), test compound (or equivalent volume of solvent), 20 pM 125 ⁇ _ n uman eotaxin (2000 Ci/mmol), 25 ng unlabeled human eotaxin (non-specific binding wells only), and AML14.3D10 cells expressing eotaxin receptor cells, or eosinophils, are added sequentially in 96-well, round-bottom, polystyrene plates to a final volume of 250 uL.
  • Assay buffer 50 mM Hepes, pH 7.2 w/ 0.5% BSA, 5 mM MgCl2, 1 mM CaCl2, 100 uM PMSF and 10
  • Assay plates are then mixed and incubated for 60 minutes at 31°C. After incubation, assay plates are harvested onto Packard 96-well GF/C Unifilter plates treated with 0.33% polyethylenimine (PEI) using Packard Filtermate 196 cell harvester. Wells and filters are washed with 200 uL 50 mM Hepes, pH 7.2 with 0.5M NaCl and 0.02% NaN3- After filtration, GF/C plates are dried and sealed. 25 uL Packard Microscint-O scintillant are then added to each well and counted for 2 minutes on Packard Topcount (liquid 125j setting).
  • PKI polyethylenimine
  • Ligands detected using assays described herein may be used in the treatment and prevention of conditions which would be benefited by the modification of the activity of the eosinophil eotaxin receptor, such as in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma.
  • a further aspect of this invention is directed to novel ligands which are identified using the subject assays.
  • the eosinophil eotaxin receptor and fragments are immunogenic.
  • another aspect of this invention is antibodies and antibody fragments which can bind to eosinophil eotaxin receptor or an eosinophil eotaxin receptor fragment.
  • These antibodies may be monoclonal antibodies and produced using either hybridoma technology or recombinant methods. They may be used as part of assay systems or to deduce the function of an eosinophil eotaxin receptor present in a cell.
  • a further aspect of this invention are antisense oligonucleotides nucleotides which can bind to eosinophil eotaxin receptor nucleotides and modulate receptor function or expression.
  • a further aspect of this invention is a method of increasing the amount of eosinophil eotaxin receptor in a cell comprising, introducing into the cell a nucleic acid encoding an eosinophil eotaxin receptor, and allowing expression of the eosinophil eotaxin receptor.
  • the receptor protein Free from receptor-associated proteins—the receptor protein is not in a mixture or solution with other membrane receptor proteins.
  • nucleic acid Free from associated nucleic acids—the nucleic acid is not covalently linked to DNA which it is naturally covalently hnked in the organism's chromosome. Isolated receptor- the protein is not in a mixture or solution with any other proteins.
  • Isolated nucleic acid the nucleic acid is not in a mixture or solution with any other nucleic acid.
  • Functional equivalent a receptor which does not have the exact same amino acid sequence of a naturally occurring eosinophil eotaxin receptor, due to alternative splicing, deletions, mutations, or additions, but retains at least 1%, preferably 10%, and more preferably 25% of the biological activity of the naturally occurring receptor.
  • Such derivatives will have a significant homology with the natural eosinophil eotaxin receptor and can be detected by reduced stringency hybridization with a DNA sequence obtained from an eosinophil eotaxin receptor.
  • the nucleic acid encoding a functional equivalent has at least about 50% homology at the nucleotide level to a naturally occurring receptor nucleic acid.
  • Purified receptor the receptor is at least about 95% pure.
  • Purified nucleic acid the nucleic acid is at least about 95% pure.
  • the 5' genomic DNA flanking sequence encoding the human eosinophil eotaxin receptor further comprises the region beginning with nucleotide 1 and ending with nucleotide 3586 as depicted below (SEQ ID NO:3):
  • sequence for the cDNA encoding human eosinophil eotaxin receptor further comprises the terminator region beginning with nucleotide 4652 and ending with nucleotide 5099 as depicted below (SEQ ID NO:4):
  • this invention also includes alternative base sequences wherein a codon (or codons) are replaced with another codon such that the amino acid sequence translated by the DNA sequences remains unchanged.
  • a sequence bearing one or more such replaced codons will be defined as a degenerate variation.
  • mutations exchanges of individual amino acids which one skilled in the art would expect to have no effect on functionality, such as valine for leucine, arginine for lysine, and asparagine for glutamine.
  • CC CKR3 The amino acid sequence of CC CKR3 shares some sequence homology with CC CKR1 (Neote, K., et al. (1993) Cell 72, 415-425), CC CKR2B (Charo, I. F., et al. (1994) Proceecing ofthe National Academy of Sciences 91, 2752-2756), CC CKR4 (Power, C. A., et al. (1995) Journal of Biological Chemistry 270, 19495-19500) and V28 (Raport, C. J., et al. (1995) Gene 163, 295-299).
  • the sequence of this protein, designated CC CKR3 is comparable to that previously reported by Combadiere et al.
  • Genomic cloning provided confirmation of the subject sequence, including lysine at position 107.
  • the sequence discrepency which results from a substitution of G to T at the third position of the codon for residue 107, could represent a genetic polymorphism. This is highly unlikely, however, because all ⁇ - and ⁇ -chemokine receptors analyzed to date contain lysine in that position including the recently described basophilic ⁇ -chemokine receptor (Power, C. A., et al.
  • CC CKR3 An unusual feature of CC CKR3 is the cluster of negatively charged amino acids (ETEELFEET) distal to TMIV in the second extracellular loop. Expression of the Human CC CKR3 in AML14.3D10 Cells
  • the CC CKR3 transfected AML14.3D10 cell line has been placed on restricted deposit with American Type Culture Collection in Rockville, Maryland as ATCC No. CRL- 12079, on April 5, 1996.
  • Stable clones were selected for neomycin resistance, and a number were chosen for further analysis.
  • a western blot was performed using antisera generated against a peptide derived from the predicted C-terminus of CC CKR3. Immunoreactive bands migrating at approximately 45-55 kd are present in primary eosinophils and the 3.16 clone, indicating that CC CKR3 is indeed expressed in these cells. There was no immunoreactive bands present in neutrophils indicating that the antisera was indeed identifying an eosinophil-specific protein.
  • CC CKR3 is a high affinity receptor for eotaxin.
  • Kd 60 nM
  • MlP-l ⁇ , and MIP-l ⁇ failed to compete at all (See Table 1).
  • the ⁇ -chemokine IL-8 did not inhibit eotaxin binding.
  • CC CKR3 is functionally coupled in AML14.3D10 cells
  • CC CKR3 The selectivity of CC CKR3 for the various ⁇ -chemokines mirrors the effectiveness of these ligands as eosinophil chemoattractants suggesting that CC CKR3 is the primary mediator of chemokine induced eosinophil chemotaxis.
  • the affinites for RANTES and MCP-3 were also identical to those measured on CC CKR3, and as with CC CKR3, neither MlP-l ⁇ , or MIP-l ⁇ , showed any ability to compete with radiolabeled eotaxin (see Table 1).
  • the Kd's obtained by competition against 125 ⁇ _MCP-3 on eosinophils are effectively indistinguishable to those measured against cloned CC CKR3 (see Table 1).
  • MCP-1 binds with much lower affinity, while MlP-l ⁇ and MlP-l ⁇ fail to bind at all.
  • the selectivity of CC CKR3 mirrors most of the binding activity of primary eosinophils. In fact, when measured by competition against 125 ⁇ _ eotaxin, the binding affinities on eosinophils for all of these ⁇ - chemokines are indistinguishable from those obtained with cloned CC CKR3.
  • CC CKR3 was cloned from eosinophils, and as shown by Western blotting is heavily expressed in these cells.
  • the abilities of the different chemokines to activate CC CKR3 are consistent with the binding data as eotaxin, RANTES, and to a lessor extent MCP- 3 all stimulate Ca ⁇ + fluxes in clones which express the receptor, while MCP-1, MlP-l ⁇ and MIP-l ⁇ do not, even at concentrations as high as 1 ⁇ M.
  • CC CKR3 is the eosinophil eotaxin receptor.
  • the primers used for PCR corresponded to a consensus sequence encoded in transmembrane domains (TM) II and VII: 5 * -PCR primer (TMII) (SEQ ID NO:5):
  • the resultant PCR product of -700 bp was subcloned into plasmid pNoTA (Five Prime, Three Prime, Ine) and sequenced using Sequenase (USB).
  • the remaining 5' and 3' sequence encoding CC CKR3 was cloned by rapid amplification of cDNA ends (RACE) using both the 5'-RACE and 3'-RACE kits (Clontech) with the following primer sequences: (5 -RACE) (SEQ ID NO:7): 5'-TCTCGCTGTACAAGCCTGTGTG-3';
  • 5 -CCTTCTCTCTTCCTATCAATCC-3 * The resultant PCR products (5'-RACE, -450 bp; 3 * -RACE, -700 bp) were subcloned into pCRII (Invitrogen) and sequenced. Upon identification of the 5'-end of the cDNA containing the initiator ATG codon and the 3'-end containing the termination codon TAG, a new set of PCR primers were designed to reamplify the entire coding region from eosinophil total RNA for expression of CC CKR3. The primer sequences used for RT/PCR were: 5'-PCR primer (SEQ ID NO:9):
  • Plasmid pD5/Igh/Neo (Daugherty, B.L., et al. (1991) Nucleic Acids Research 19, 2471-2476) was digested with the restriction enzyme Sail, filled in with E. coli DNA polymerase I Klenow fragment to create a blunt end and subsequently digested with the restriction enzyme Notl.
  • CM VIE intron A fragment from plasmid p89-l 1 was digested with Clal, filled in to create a blunt end and subsequently digested with Hindill. These fragments were used in a three-way ligation with a Hindill and Notl fragment of the human C5a receptor cDNA.
  • the C5a receptor fragment was excised with Hindill and Notl and replaced with the eotaxin receptor cDNA of 1105 bp obtained by RT/PCR with oligonucleotides SEQ ID NO:9 and SEQ ID NO: 10 after digestion with Hindill and Notl.
  • Several clones were sequenced and one clone comprising the consensus sequence was chosen for expression of CC CKR3 in heterologous cells.
  • AML14.3D10 cells (Paul, C. C, et al. (1995) Blood 86, 3737-3744) were cultured in RPMI-1640, 10% FBS, 1 mM sodium pyruvate, 0.5 ⁇ M ⁇ -mercaptoethanol and 2 mM L-glutamine (complete medium). Cells were harvested at a density of 0.3 x 10°7mL, washed once in PBS, resuspended in RPMI at 10 7 /mL, and 25 ⁇ g of plasmid was added. Electroporation was carried out at 300 V, 960 ⁇ F using a Gene Pulser (BioRad).
  • eosinophils were isolated from granulophoresis preparations obtained from allergic and asthmatic donors (Bach, M. K., et al. (1990) Journal of Immunological Methods 130, 277-281). The leukocytes were mixed with equal volumes of HBSS and layered over LSM (Organon Teknika) as described (Rollins, T. E., et al. (1988) Journal of Biological Chemistry 263, 520-526). After lysis of erythrocytes with NH4CI, the granulocytes were subsequently treated with anti-CD 16 microbeads followed by MACS separation (Miltenyl Biotech) (Hansel, T. T., et al. (1991) Journal of Immunological Methods 145, 105-110). Typically the eosinophil preparations were >99% pure as determined using the LeukoStat staining kit (Fisher).
  • Polyclonal rabbit antisera was generated to CC CKR3 using the C-terminal decapeptide sequence TAEPELSIVF. Peptide synthesis, coupling to thyroglobulin and production of antisera was performed (Miller, D. K., et al. (1993) Journal of Biological Chemistry 268, 18062-18069). Whole cells were boiled and sonicated in Laemli sample buffer (Laemmli, U. K.
  • MCP-3, MCP-1, RANTES, murine and human eotaxin were obtained from Peprotech (Princeton, NJ).
  • 125 ⁇ _ MCP-3 and 125 ⁇ _MIP-l ⁇ were obtained from New England Nuclear (Boston, MA), and ⁇ i-human-eotaxin was obtained from Amersham.
  • Binding of 125 ⁇ _ ⁇ a beled ligands (typically a total of 2 x 104 cpm) in the presence of varying concentrations of unlabeled ligands to intact cells (typically 1.5 x 104 , 10 ⁇ , or 10 ⁇ for experiments with labeled eotaxin, MCP-3, or MlP-l ⁇ , respectively) were performed at 32°C (Van Riper, G., et al. (1993) Journal of Experimantal Medicine 111, 851-856).
  • CC CKR3 Binding Assay Assay buffer 50 mM Hepes, pH 7.2 w/ 0.5% BSA, 5 mM
  • AML14.3D10 expressing eotaxin receptor (CCCKR3) cells are incubated with test compound and stimulated with eotaxin, RANTES, or MCP-3, pelleted and lysed in 1 mL lysis buffer (1% Nonidet P-40, 100 mM NaCl, 20 mM Tris, pH 7.4, 10 mM iodoacetamide, 46 mM b- glyceraphosphate, 10 mM NaF, 1 mM PMSF, 1 ug/mL leupeptin, 1 ug/mL chymostatin, 1 ug/mL antipain, 1 ug/mL pepstatin A, and 1 mM sodium orthovanadate).
  • lysis buffer 1% Nonidet P-40, 100 mM NaCl, 20 mM Tris, pH 7.4, 10 mM iodoacetamide, 46 mM b- glyceraphosphate, 10 mM NaF
  • Ly sates are then pre-cleared for 1 hr with uncoupled protein A Affi-Gel beads. Immunoprecipitation is then performed with p85 polyclonal antiserum (1 ul/mL lysate; Upstate Biologies, New York, NY), coupled to protein A Affi-Gel beads (Bio- Rad) at 4°C for 2 hr.
  • Immunoprecipitates are washed and subjected to in vitro lipid kinase assays by using a lipid mixture, 100 ul 0.1 mg/ml Ptdlns and 0.1 mg/ml phosphatidylserine dispersed by sonication into solution in 20 mM HEPES, pH 7.0, and 1 mM EDTA.
  • the reaction is initiated by the addition of 100 mM ATP and 20 uCi [gamma- 32 P]ATP (3000 Ci/mmol) in 20 ul kinase buffer.
  • the reaction is then terminated after 15 min and the phosphoinositide lipids are separated by thin layer chromatography (TLC) and visualized by exposure to iodine vapor autoradiography.
  • TLC thin layer chromatography
  • AML14.3D10 expressing eotaxin receptor cells are isolated by centrifugation (van Riper, G., et al. (1994) J. Immunol. 152, 4055- 4061) for 15 min at 150 X g, washed and resuspended at 10' cells/ml in HBSS (pH 7.4) containing 1 mM CaCl2 and 1 mM MgCl2 (chemotaxis buffer). The chemotaxis experiments is then performed in Transwell dishes (6.5 mm, Costar, Cambridge, MA).
  • the lower chamber contains 0.6 ml of chemotaxis buffer and is separated from the upper chamber containing 10" cells by a 5-mm pore Nucleopore polycarbonate membrane (Nucleopore Co ⁇ oration, Pleasanton, CA). After a 15 min preincubation at 37°C, test compound and eotaxin, RANTES, or MCP-3 are added to the lower chamber to a final concentration of 300 nM. After 2 hrs at 37°C, the upper chamber inserts are removed, and the cells that migrate to the lower chamber are enumerated by a Coulter Counter (Coulter Electronics, Hialeah, FL).
  • Coulter Counter Coulter Electronics, Hialeah, FL.
  • AML14.3D10 expressing eotaxin receptor cells are labeled with [ 3 H] inositol (10 uCi/ml) for 24 hrs as described (Wu, D., et al. (1993) Science 261, 101-103).
  • Test compound and arious concentrations of eotaxin, MCP-3, or RANTES are then added to the cells for 30 min.
  • the cells are lysed in 10% perchloric acid, neutralized in 2 N KOH and centrifuged. The supernatant is transferred to columns containing 0.5 ml AG1-X8 anion exchange resin, washed with 6 ml borax buffer and eluted with 0.3 ml formic acid (0.1 M). The eluted samples are mixed with scintillation cocktail and counted.
  • AML14.3D10 expressing eotaxin receptor cells are subject to serum starvation for 16 hrs.
  • the cells are then mixed at a 3: 1 (v/v) ratio with low melting temperature agarose.
  • a 10 ul drop of the cell/agarose mixture is pipetted into a sterile Capsule Cup (Molecular Devices) at a cell density of approximately 200,000 cells/cup.
  • the cell/agarose drop forms a gel after about 5 min, and is assembled into the cup between two 3 um porosity polycarbonate membranes with running medium.
  • the assembled capsule cups is placed into the sensor chambers and then placed on the Cytosensor Microphysiometer (Molecular Devices) containing 1 ml of running medium.
  • the chambers are allowed to equilibrate for 1 hr at 37°C with a flow rate of 100 ul/min.
  • the experiment is initiated with an 8 min exposure of eotaxin, RANTES, MCP-3 and test compound at various concentrations and the acidification rate over baseline will be measured in the medium (McConnell, H.M., et al. (1992) Science 257, 1906-1912) until the cells return to the unstimulated level.
  • AML14.3D10 expressing eotaxin receptor cells are diluted in APA buffer (HBSS; 25mM Hepes; 0.2% BSA, pH7.2) at a concentration of 4 x lOfyml.
  • APA buffer HBSS; 25mM Hepes; 0.2% BSA, pH7.2
  • test compound and eotaxin, RANTES, or MCP-3 added into a 96-well plate and incubated at 37°C. 100 ml of cells are added to the plate and incubated for 20 sec to which 100 ml of APA cocktail (2 mis 8% formaldehyde; 460uLs 0.33uM Rhodamine- phalloidin; 1.85 mg 200ug/ml lysolecithin; 7.25 mis HBSS) is added.
  • characterization data observed may vary slightly according to and depending upon the particular assay or characterization method employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. It is intended, therefore, that the invention be defined by the scope of the claims which follow and that such claims be inte ⁇ reted as broadly as is reasonable.
  • CTGCTCTTCC TCGTCACCCT TCCATTCTGG ATCCACTATG TCAGGGGGCA TAACTGGGTT 300
  • ATCTTTTTCA TAATCCTGCT GACAATCGAC AGGTACCTGG CCATTGTCCA TGCTGTGTTT 420
  • GAGGGCTCTC CATTCCAGCC CAAGGAAGAC TAAGAATGAA TACCTCATGA GTATATTAGC 360
  • CTTCTTTCTT ATTGTTCTTA CTTATTTACG ATTACCCTAT CGTTTTCCCA AAATGTAAAA 960
  • TATATTGTTA TCATTATCTA GCCTGTTTTT TCCTGTTGTG TATTTCTTCC TTTAAATGCT 3300

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

The eosinophil eotaxin receptor has been isolated, cloned and sequenced. This receptor is a human β-chemokine receptor and has been designated 'CC CKR3'. The eosinophil eotaxin receptor may be used to screen and identify compounds that bind to the eosinophil eotaxin receptor. Such compounds would be useful in the treatment and prevention of atopic conditions including allergic rhinitis, dermatis, conjunctivitis, and particularly bronchial asthma.

Description

TITLE OF THE INVENTION EOSINOPHIL EOTAXIN RECEPTOR
CROSS REFERENCE TO RELATED APPLICATIONS This application claims priorty under 35 U.S.C. §
119(e) from provisional application Case Number 19634PV, filed April 26, 1996 and from provisional application Case Number 19697PV, filed April 26, 1996 as USSN 60/016,158.
FIELD OF THE INVENTION
This invention relates to an eosinophil eotaxin receptor ("CC CKR3"), in particular, the human eosinophil eotaxin receptor and nucleic acids encoding this receptor. This invention further relates to assays which may be used to screen and identify compounds that bind to the eosinophil eotaxin receptor. Such compounds would be useful in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma.
BACKGROUND OF THE INVENTION
Eosinophils play prominant roles in a variety of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma (for a reviews see e.g. Gleich, G. J., et al., Eosinophils. J. I. Gallin, I. M. Goldstein, R. Snyderman, Eds., Inflammation: Basic Principles and Clinical Correlates (Raven Press, Ltd., New York, 1992) and Seminario, M. C, et al. (1994) Current Opinion in Immunology 6, 860-864). A pivotal event in the process is the accumulation of eosinophils at the involved sites. While a number of the classical chemoattractants, including C5a, LTB4, and PAF, are known to attract eosinophils (Gleich, G. J., et al., Eosinophils. J. I. Gallin, et al. Eds., Inflammation: Basic Principles and Clinical Correlates (Raven Press, Ltd., New York, 1992)), these mediators are promiscuous, acting on a variety of leukocytes including neutrophils, and are unlikely to be responsible for the selective accumulation of eosinophils. In contrast, the chemokines a family of 8-10 kDa proteins are more restricted in the leukocyte subtypes they target and are potential candidates for the recruitment of eosinophils in atopic diseases and asthma (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179). Although there is a mounting body of evidence that eosinophils are recruited to sites of allergic inflammation by a number of β-chemokines, particularly eotaxin and RANTES, the receptor which mediates these actions has not been identified.
The chemokines contain four conserved cysteines, and are divided into two sub-families based on the arrangement of the first cysteine pair (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179). In the α-chemokine family, which includes IL-8, MGS A, NAP-2 and IP- 10, these two cysteines are separated by a single amino acid, while in the β-chemokine family, which includes RANTES ("regulated on activation T expressed and secreted"), MCP-1 ("monocyte chemotactic protein"), MCP-2, MCP-3, MlP-lα ("macrophage inflammatory protein"), MlP-lβ and eotaxin, these two cysteines are adjacent. There is a functional correlate to this structural division. The α-chemokines act primarily on neutrophils, and the β-chemokines on monocytes, lymphocytes, basophils and eosinophils (Baggiolini, M., Dewald, B. and Moser, B. (1994) Advances in Immunology 55, 97-179). In particular, RANTES, MCP-2, MCP-3, and MlP-lα have been shown to activate eosinophils in vitro (Dahinden, C. A., et al. (1994) Journal of Experimental Medicine 179, 751-756; Ebisawa, M., et al. (1994) Journal of Immunology 153, 2153-2160; Weber, M., et al. (1995) Journal of Immunology 154, 4166-4172), and RANTES to selectively attract eosinophils in vivo (Meurer, R., et al. (1993) Journal of Experimental Medicine 178, 1913-1921; Beck, L., et al. (1995) FASEB Journal 9, A804). Similarly, eotaxin, a new member of the β-chemokine family, first described in guinea pigs (Griffiths- Johnson, D. A., et al. (1993) Bichemical and Biophysical Research Communications 197, 1167-1172; Jose, P. J., et al. (1994) Journal of Experimental Medicine 179, 881-887) and mice (Rothenberg, et al. (1995) Proceedings of the National Academy of Sciences 92, 8960-8964) is also a potent attractant and activator of eosinophils both in vitro and in vivo. Moreover, eotaxin is generated during antigenic challenge in the guinea pig model of allergic airway inflammation (Jose, et al. (1994) J. Exp. Med., 179, 881-887; Rothenberg, et al. (1995) J. Exp. Med., 181, 1211-1216. The cloning of guinea pig eotaxin has been disclosed (PCT Patent Publication No. WO 95/07985; March 23, 1995). The cloning of the human eosinophil chemoattractant eotaxin has recently been reported (Ponath, et al., J. Clin. Invest. (1996) 97(3) 604-612) and eotaxin has been suggested to be a very important agent in the mechanism of allergic inflammation (Baggiolini, et al., J. Clin. Invest. (1996) 97(3) 587).
Eosinophils are attracted by a number of β-chemokines, the most potent of which are eotaxin (Griffiths- Johnson, D. A., et al. (1993) Bichemical and Biophysical Research Communications 197, 1167- 1172; Jose, P. J., et al. (1994) Journal of Experimental Medicine 179, 881-887; Rothenberg, et al. (1995) Proceedings ofthe National Academy of Sciences 92, 8960-8964) and RANTES (Dahinden, C. A., et al. (1994) Journal of Experimental Medicine 179, 751-756; Ebisawa, M., et al. (1994) Journal of Immunology 153, 2153-2160; Weber, M., et al. (1995) Journal of Immunology 154, 4166-4172;
Meurer, R., et al. (1993) Journal of Experimental Medicine 178, 1913- 1921 ; Beck, L., et al. (1995) FASEB Journal 9, A804). Although several human β-chemokine receptors have been characterized in detail, none have the appropriate selectivity to account for the observed responses.
While elucidation of the actions of β-chemokines on eosinophils has contributed greatly to the understanding of eosinophil biology, information regarding the cell surface receptors which mediate these effects remain sparse. Furthermore, there are no reports describing binding studies of any of the β-chemokines to primary eosinophils. The known β-chemokine receptors are members of the G protein-coupled receptor superfamily. Two of these receptors, CC CKR1 (12, 13) and CC CKR2 (MCP-1R) (Charo, I. F., et al. (1994) Proceecing of the National Academy of Sciences 91, 2752-2756; Myers, S. J., et al. (1995) Journal of Biological Chemistry 270, 5786- 5792; Franci, C, et al. (1995) Journal of Immunology 154, 6511- 6517) found on monocytes, have been extensively studied and their selectivity for the different chemokines defined. However, neither of these receptors has the necessary ligand selectivity or the appropriate expression patterns required to mediate the effects of the β-chemokines on eosinophils. For example, CC CKR1 binds RANTES with high affinity, but binds eotaxin poorly, and while the effects of eotaxin on CC CKR2 have not been studied this receptor has no avidity for RANTES (Myers, S. J., et al. (1995) Journal of Biological Chemistry 270, 5786-5792).
A review of the role of chemokines in allergic inflammation is provided by Kita, H., et al., J. Exp. Med. 183, 2421- 2426 (June 1996). In particular, this review discusses the role which the receptor CKR-3 plays in the process of allergic inflammation. The cloning, expression and characterization of the human eosinophil eotaxin receptor has been reported by Daugherty, B.J., et al., J. Exp. Med. 183, 2349-2354 (May 1996). This publication discloses the cloning and functional expression of the chemokine receptor CC CKR3, as well as its characterization.
The cloning and expression of a human eosinophil receptor was allegedly achieved by Combadiere, C, et al., J. Biological Chem. 270 (27), 16491-16494 (July 14, 1995). However, in a subsequent retraction (J. Biological Chem. 270, 30235 (1995)) they confirmed that the receptor which was actually cloned and expressed was not CC CKR3, but was another CC chemokine receptor CC CKR5. This receptor was subsequently characterized by Kitaura, M., et al., J. Biological Chem. Ill (13), 7725-7730 (March 29, 1996).
A human eotaxin receptor has been reported by Ponath, P.D., et al. J. Exp. Med. 183, 2437-2448 (June 1996) and Gerard, C.J., et al., PCT Publication No. WO 96/22371 (July 25, 1996). However, the sequence disclosed in this publication possesses an error in the assignment of threonine rather than serine at position # 276 of the receptor. In addition, functionality of the receptor was not fully demonstrated.
A retrovirus designated human immunodeficiency virus (HIV-1) is the etiological agent of the complex disease that includes progressive destruction of the immune system (acquired immune deficiency syndrome; AIDS) and degeneration of the central and peripheral nervous system. This virus was previously known as LAV, HTLV-III, or ARV. Entry of HIV-1 into a target cell requires cell- surface CD4 and additional host cell cofactors. Fusin has been identified as a cofactor required for infection with virus adapted for growth in transformed T-cells, however, fusin does not promote entry of macrophagetropic viruses which are believed to be the key pathogenic strains of HIV in vivo. It has recently been recognized that for efficient entry into target cells, human immunodeficiency viruses require the chemokine receptors CCR-5 and CXCR-4, as well as the primary receptor CD4 (Levy, N. Engl. J. Med.. 335(20). 1528-1530 (Nov. 14 1996). The principal cofactor for entry mediated by the envelope glycoproteins of primary macrophage-trophic strains of HIV-1 is CCR5, a receptor for the β-chemokines RANTES, MlP-lα and MIP- lβ (Deng, et al., Nature. 381. 661-666 (1996)). HIV attaches to the CD4 molecule on cells through a region of its envelope protein, gpl20. It is believed that the CD-4 binding site on the gpl20 of HIV interacts with the CD4 molecule on the cell surface, and undergoes conformational changes which allow it to bind to another cell-surface receptor, such as CCR5 and/or CXCR-4. This brings the viral envelope closer to the cell surface and allows interaction between gp41 on the viral envelope and a fusion domain on the cell surface, fusion with the cell membrane, and entry of the viral core into the cell. It has been shown that β-chemokine ligands prevent HIV-1 from fusing with the cell (Dragic, et al., Nature. 381. 667-673 (1996)). It has further been demonstrated that a complex of gpl20 and soluble CD4 interacts specifically with CCR-5 and inhibits the binding of the natural CCR-5 ligands MlP-lα and MlP-lβ (Wu, et al., Nature. 384. 179-183 (1996); Trkola, et al., Nature. 384. 184-187 (1996)). Humans who are homozygous for mutant CCR-5 receptors which do not serve as co-receptors for HIV-1 in vitro apper to be unusually resistant to HIV-1 infection and are not immunocompromised by the presence of this genetic variant (Nature. 382. 722-725 (1996)). Absence of CCR-5 appears to confer protection from HIV-1 infection (Nature. 382. 668-669 (1996)). Other chemokine receptors may be used by some strains of HIV-1 or may be favored by non-sexual routes of transmission. Although most HIV-1 isolates studied to date utilize CCR-5 or fusin, some can use both as well as the related CCR-2B and CCR-3 as co-receptors (Nature Medicine. 2(11), 1240-1243 (1996)). The determination that chemokine receptors are critical co-receptors for the entry of HIV into cells was pronounced a "1996 Breakthrough of the Year" by Science Magazine (Science. 274. 1987-1991 (Dec. 20, 1996)). The use of orally-active agents which modulate the action of the eosinophil eotaxin receptor would be a significant advance in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma. Further, agents which could block the eosinophil eotaxin receptor in humans who possess normal chemokine receptors should prevent infection in healthy individuals and slow or halt viral progression in infected patients.
It would also be desirable to know the molecular structure of the eosinophil eotaxin receptor in order to analyze this new receptor family and understand its normal physiological role. This could lead to a better understanding of the in vivo processes which occur upon ligand-receptor binding. Further, it would be desirable to use cloneoVeosinophil eotaxin receptor as essential components of an assay system which can identify new agents for the treatment and prevention of atopic conditions.
SUMMARY OF THE INVENTION
The present invention relates to a novel receptor which is the eosinophil eotaxin receptor. This receptor is a human β-chemokine receptor and has been designated "CC CKR3". One aspect of the present invention is directed to the human eosinophil eotaxin receptor, free from receptor-associated proteins. A further aspect of this invention is the human eosinophil eotaxin receptor which is isolated or purified.
Another aspect of this invention are eosinophil eotaxin receptors which are encoded by substantially the same nucleic acid sequences, but which have undergone changes in splicing or other RNA processing-derived modifications or mutagenesis induced changes, so that the expressed protein has a homologous, but different amino acid sequence from the native forms. These variant forms may have different and/or additional functions in human and animal physiology or in vitro in cell based assays.
The present invention further provides the eosinophil eotaxin receptor, CC CKR3, which is a β-chemokine receptor and which was cloned from primary eosinophils, and expressed in AML14.3D10 cells. This receptor binds the potent eosinophil attractants, eotaxin, RANTES and MCP-3 with high affinity. In addition, eotaxin and RANTES, and to a lessor extent MCP-3, induce Ca2+-fluxes in cells expressing CC CKR3. Correlation with the binding properties of primary eosinophils provide conclusive evidence that CC CKR3 is the primary endogenous receptor which mediates the effects of β- chemokines on eosinophils.
The present invention further relates to assays which employ a novel receptor which is the eosinophil eotaxin receptor.
This receptor is a human β-chemokine receptor and has been designated "CC CKR3". One aspect of the present invention is directed to assays employing the the human eosinophil eotaxin receptor, free from receptor-associated proteins. A further aspect of this invention is directed to assays which employ the human eosinophil eotaxin receptor which is isolated or purified. In addition, the present invention provides assays in which the eosinophil eotaxin receptor is expressed in an AML14.3D10 cell line. DETAILED DESCRIPΗON OF THE INVENTION
The present invention is directed to an eosinophil eotaxin receptor "CC CKR3" which is a G protein-coupled receptor and has been cloned from human eosinophils and which when stably expressed in AML14.3D10 cells binds eotaxin, RANTES and MCP-3 with high affinity. Competition binding studies against 125τ_human eotaxin gives Kd values of 0.1, 2.7, and 3.1 nM, respectively for the three β- chemokines. CC CKR3 also binds MCP-1 with lower affinity, but does not bind MlP-lα or MIP-lβ. Eotaxin, RANTES, and to a lessor extent MCP-3, but not the other chemokines activate CC CKR3 as determined by the ability to stimulate a Ca2+-flux in clones expressing the receptor. Competition binding studies on primary eosinophils give binding affinities for the different chemokines which are indistinguishable from those measured with CC CKR3. Since CC CKR3 is prominently expressed in eosinophils it is concluded that CC CKR3 is the eosinophil eotaxin receptor. Eosinophils also express a much lower level of a second chemokine receptor, CC CKR1, which appears to be responsible for the effects of MlP-lα .
The eosinophil eotaxin receptor is a protein containing various functional domains, including one or more domains which anchor the receptor in the cell membrane, and at least one ligand binding domain. As with many receptor proteins, it is possible to modify many of the amino acids, particularly those which are not found in the ligand binding domain, and still retain at least a percentage of the biological activity of the original receptor. In accordance with this invention, it is suggested that certain portions of the eosinophil eotaxin receptor are not essential for its activation by β-chemokines. Thus this invention specifically includes modified functionally equivalent eosinophil eotaxin receptors which have deleted, truncated, or mutated portions. This invention also specifically includes modified functionally equivalent eosinophil eotaxin receptors which contain modified and/or deletions in other domains, which are not accompanied by a loss of functional activity. Additionally, it is possible to modify other functional domains such as those that interact with second messenger effector systems, by altering binding specificity and/or selectivity. Such functionally equivalent mutant receptors are also within the scope of this invention.
A further aspect of this invention are nucleic acids which encode an eosinophil eotaxin receptor or a functional equivalent from human or other species. These nucleic acids may be free from associated nucleic acids, or they may be isolated or purified. For most cloning purposes, cDNA is a preferred nucleic acid, but this invention specifically includes other forms of DNA as well as RNAs which encode an eosinophil eotaxin receptor or a functional equivalent.
Yet another aspect of this invention relates to vectors which comprise nucleic acids encoding an eosinophil eotaxin receptor or a functional equivalent. These vectors may be comprised of DNA or RNA; for most cloning purposes DNA vectors are preferred. Typical vectors include plasmids, modified viruses, bacteriophage and cosmids, yeast artificial chromosomes and other forms of episomal or integrated DNA that can encode an eosinophil eotaxin receptor. It is well witnin the skill of the ordinary artisan to determine an appropriate vector for a particular gene transfer or other use.
A further aspect of this invention are host cells which are transformed with a gene which encodes an eosinophil eotaxin receptor or a functional equivalent. The host cell may or may not naturally express an eosinophil eotaxin receptor on the cell membrane. Preferably, once transformed, the host cells are able to express the eosinophil eotaxin receptor or a functional equivalent on the cell membrane. Depending on the host cell, it may be desirable to adapt the DNA so that particular codons are used in order to optimize expression. Such adaptations are known in the art, and these nucleic acids are also included within the scope of this invention. The receptors of this invention were cloned from RNA isolated from eosinophils. Degenerate PCR was used with primers designed from both CCCKR1 and CCCKR2, and clones screened by expression in the AML14.3D10 cell line. The cloning was made difficult by several factors. First, prior to this invention there was very little information available about the biochemical characteristics and intracellular signalling/effector pathways used by these receptors making screening procedures uncertain. Second, this receptor could not be expressed and/or functionally coupled in the cell lines normally used for clomng receptors such, as COS, CHO, HEK293. After repeated failures using standard lines, an obscure eosinophilic- like cell line, AML14.3D10, was tried and found to suitable for expression of the receptors described in this invention.
The present invention further relates to assays which employ a novel receptor which is the eosinophil eotaxin receptor. This receptor is a human β-chemokine receptor and has been designated "CC CKR3". One aspect of the present invention is directed to assays employing the the human eosinophil eotaxin receptor, free from receptor-associated proteins. A further aspect of this invention is directed to assays which employ the human eosinophil eotaxin receptor which is isolated or purified. In addition, the present invention provides assays in which the eosinophil eotaxin receptor is expressed in an AML14.3D10 cell line. A particular embodiment of this invention is directed to an assay to determine the presence of a compound which binds to the eosinophil eotaxin receptor. Thus, this invention also comprises a method to determine the presence of a compound which binds to an eosinophil eotaxin receptor comprising: (a) introducing a nucleic acid which encodes an eosinophil eotaxin receptor into a cell under conditions so that eosinophil eotaxin receptor is expressed;
(b) introducing a detector molecule or a nucleic acid encoding a detector molecule into the cell, wherein the detector molecule is directly or indirectly responsive to a eosinophil eotaxin- ligand binding event;
(c) contacting the cell with a compound suspected of binding to the eosinophil eotaxin receptor; and (d) determining whether the compound binds to the eosinophil eotaxin receptor by monitoring the detector molecule.
In a preferred embodiment of the present invention, the eosinophil eotaxin receptor is expressed in AML14.3D10 cells.
In another preferred embodiment of the present invention, the binding of the compound suspected of binding to the eosinophil eotaxin receptor is compared to the binding or the influence of eotaxin, RANTES and MCP-3.
A further embodiment of this invention is directed to an assay to determine the presence of a compound which antagonizes the binding of a known ligand to the eosinophil eotaxin receptor. Thus, this invention further comprises a method to determine the presence of a compound which antagonizes the eosinophil eotaxin receptor comprising:
(a) introducing a nucleic acid which encodes the eosinophil eotaxin receptor into a cell under conditions so that eosinophil eotaxin receptor is expressed;
(b) introducing a detector molecule or a nucleic acid encoding a detector molecule into the cell, wherein the detector molecule is directly or indirectly responsive to an eosinophil eotaxin- ligand antagonism event;
(c) contacting the cell with a compound suspected of antagonizing the eosinophil eotaxin receptor;
(d) contacting the cell with a compound which is a known ligand of the eosinophil eotaxin receptor; and (e) determining whether the compound antagonizes the action of the known ligand to the eosinophil eotaxin receptor by monitoring the detector molecule. In a preferred embodiment of the present invention, the eosinophil eotaxin receptor is expressed in AML14.3D10 cells.
In another preferred embodiment of the present invention, the known ligand of the eosinophil eotaxin receptor is eotaxin, RANTES and MCP-3.
One aspect of this invention is the development of a sensitive, robust, reliable and high-throughput screening assay which may be used to detect ligands which bind to the eosinophil eotaxin receptor, in particular, antagonists of the action of chemokines on eosinophils.
In particular, a typical protocol of such an assay is as follows. Assay buffer (50 mM Hepes, pH 7.2 w/ 0.5% BSA, 5 mM MgCl2, 1 mM CaCl2, 100 uM PMSF and 10 ug/ml phosphoramidon, leupeptin, aprotinin and chymostatin), test compound (or equivalent volume of solvent), 20 pM 125ι_numan eotaxin (2000 Ci/mmol), 25 ng unlabeled human eotaxin (non-specific binding wells only), and AML14.3D10 cells expressing eotaxin receptor cells, or eosinophils, are added sequentially in 96-well, round-bottom, polystyrene plates to a final volume of 250 uL. Assay plates are then mixed and incubated for 60 minutes at 31°C. After incubation, assay plates are harvested onto Packard 96-well GF/C Unifilter plates treated with 0.33% polyethylenimine (PEI) using Packard Filtermate 196 cell harvester. Wells and filters are washed with 200 uL 50 mM Hepes, pH 7.2 with 0.5M NaCl and 0.02% NaN3- After filtration, GF/C plates are dried and sealed. 25 uL Packard Microscint-O scintillant are then added to each well and counted for 2 minutes on Packard Topcount (liquid 125j setting).
Ligands detected using assays described herein may be used in the treatment and prevention of conditions which would be benefited by the modification of the activity of the eosinophil eotaxin receptor, such as in the treatment and prevention of atopic conditions including allergic rhinitis, dermatitis, conjunctivitis, and particularly bronchial asthma. A further aspect of this invention is directed to novel ligands which are identified using the subject assays.
The eosinophil eotaxin receptor and fragments are immunogenic. Thus, another aspect of this invention is antibodies and antibody fragments which can bind to eosinophil eotaxin receptor or an eosinophil eotaxin receptor fragment. These antibodies may be monoclonal antibodies and produced using either hybridoma technology or recombinant methods. They may be used as part of assay systems or to deduce the function of an eosinophil eotaxin receptor present in a cell.
A further aspect of this invention are antisense oligonucleotides nucleotides which can bind to eosinophil eotaxin receptor nucleotides and modulate receptor function or expression. A further aspect of this invention is a method of increasing the amount of eosinophil eotaxin receptor in a cell comprising, introducing into the cell a nucleic acid encoding an eosinophil eotaxin receptor, and allowing expression of the eosinophil eotaxin receptor.
As used throughout the specification and claims, the following definitions shall apply:
Ligand- any molecule which binds to an eosinophil eotaxin receptor of this invention. These ligands can have either agonist, partial agonist, partial antagonist or antagonist activity.
Free from receptor-associated proteins— the receptor protein is not in a mixture or solution with other membrane receptor proteins.
Free from associated nucleic acids— the nucleic acid is not covalently linked to DNA which it is naturally covalently hnked in the organism's chromosome. Isolated receptor- the protein is not in a mixture or solution with any other proteins.
Isolated nucleic acid— the nucleic acid is not in a mixture or solution with any other nucleic acid. Functional equivalent— a receptor which does not have the exact same amino acid sequence of a naturally occurring eosinophil eotaxin receptor, due to alternative splicing, deletions, mutations, or additions, but retains at least 1%, preferably 10%, and more preferably 25% of the biological activity of the naturally occurring receptor. Such derivatives will have a significant homology with the natural eosinophil eotaxin receptor and can be detected by reduced stringency hybridization with a DNA sequence obtained from an eosinophil eotaxin receptor. The nucleic acid encoding a functional equivalent has at least about 50% homology at the nucleotide level to a naturally occurring receptor nucleic acid.
Purified receptor— the receptor is at least about 95% pure. Purified nucleic acid— the nucleic acid is at least about 95% pure.
Single-letter abbreviations for amino acid residues are as follows: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, He; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gin; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.
Orphan Cloning of an Eosinophil Chemokine Receptor RT/PCR conducted using oligonucleotide primers developed from the amino acid residues clustered within transmembrane helicies II (TMII) and VII (TMVII) of the β-chemokine receptors, CC CKR1 (Neote, K., et al. (1993) Cell 72, 415-425) and MCP-1R (Charo, I. F., et al. (1994) Proceecing of the National Academy of Sciences 91, 2752-2756) on total RNA isolated from eosinophils yielded DNA fragments of -700 bases, a size consistent with that expected for a G protein coupled receptor. Analysis of several TMII to TMVII clones provided a novel sequence which was 76% homologous with human CC CKR1 at the nucleic acid level. Completion of the cloning of the 3' and 5' ends gave a sequence for a protein of 355 residues in length, 63% identical to CC CKR1, and 51% identical to CC CKR2B, its closest homologues. The amino acid sequence of the human eosinophil eotaxin receptor CC CKR3 is depicted below (SEQ ID NO:l):
Met Thr Thr Ser Leu Asp Thr Val Glu Thr Phe Gly Thr Thr Ser Tyr Tyr Asp Asp Val Gly Leu Leu Cys Glu Lys Ala Asp Thr Arg Ala Leu Met Ala Gin Phe Val Pro Pro Leu Tyr Ser Leu Val Phe Thr Val Gly Leu Leu Gly Asn Val Val Val Val Met lie Leu He Lys Tyr Arg Arg Leu Arg He Met Thr Asn He Tyr Leu Leu Asn Leu Ala He Ser Asp Leu Leu Phe Leu Val Thr Leu Pro Phe Trp He His Tyr Val Arg Gly His Asn Trp Val Phe Gly His Gly Met Cys Lys Leu Leu Ser Gly Phe Tyr His Thr Gly Leu Tyr Ser Glu He Phe Phe He He Leu Leu Thr He Asp Arg Tyr Leu Ala He Val His Ala Val Phe Ala Leu Arg Ala Arg Thr Val Thr Phe Gly Val He Thr Ser He Val Thr Trp Gly Leu Ala Val Leu Ala Ala Leu Pro Glu Phe He Phe Tyr Glu Thr Glu Glu Leu Phe Glu Glu Thr Leu Cys Ser Ala Leu Tyr Pro Glu Asp Thr Val Tyr Ser Trp Arg His Phe His Thr Leu Arg Met Thr He Phe Cys Leu Val Leu Pro Leu Leu Val Met Ala He Cys Tyr Thr Gly He He Lys Thr Leu Leu Arg Cys Pro Ser Lys Lys Lys Tyr Lys Ala He Arg Leu He Phe Val He Met Ala Val Phe Phe He Phe Trp Thr Pro Tyr Asn Val Ala He Leu Leu Ser Ser Tyr Gin Ser He Leu Phe Gly Asn Asp Cys Glu Arg Ser Lys His Leu Asp Leu Val Met Leu Val Thr Glu Val He Ala Tyr Ser His Cys Cys Met Asn Pro Val He Tyr Ala Phe Val Gly Glu Arg Phe Arg Lys Tyr Leu Arg His Phe Phe His Arg His Leu Leu Met His Leu Gly Arg Tyr He Pro Phe Leu Pro Ser Glu Lys Leu Glu Arg Thr Ser Ser Val Ser Pro Ser Thr Ala Glu Pro Glu Leu Ser He Val Phe The sequence for the cDNA encoding the human eosinophil eotaxin receptor CC CKR3 beginning with nucleotide 3587 and ending with nucleotide 4651 is depicted below (SEQ ID NO:2):
ATGA CAACCTCACT
3601 AGATACAGTT GAGACCTTTG GTACCACATC CTACTATGAT GACGTGGGCC
3651 TGCTCTGTGA AAAAGCTGAT ACCAGAGCAC TGATGGCCCA GTTTGTGCCC
3701 CCGCTGTACT CCCTGGTGTT CACTGTGGGC CTCTTGGGCA ATGTGGTGGT
3751 GGTGATGATC CTCATAAAAT ACAGGAGGCT CCGAATTATG ACCAACATCT 3801 ACCTGCTCAA CCTGGCCATT TCGGACCTGC TCTTCCTCGT CACCCTTCCA
3851 TTCTGGATCC ACTATGTCAG GGGGCATAAC TGGGTTTTTG GCCATGGCAT
3901 GTGTAAGCTC CTCTCAGGGT TTTATCACAC AGGCTTGTAC AGCGAGATCT
3951 TTTTCATAAT CCTGCTGACA ATCGACAGGT ACCTGGCCAT TGTCCATGCT
4001 GTGTTTGCCC TTCGAGCCCG GACTGTCACT TTTGGTGTCA TCACCAGCAT 4051 CGTCACCTGG GGCCTGGCAG TGCTAGCAGC TCTTCCTGAA TTTATCTTCT
4101 ATGAGACTGA AGAGTTGTTT GAAGAGACTC TTTGCAGTGC TCTTTACCCA
4151 GAGGATACAG TATATAGCTG GAGGCATTTC CACACTCTGA GAATGACCAT
4201 CTTCTGTCTC GTTCTCCCTC TGCTCGTTAT GGCCATCTGC TACACAGGAA
4251 TCATCAAAAC GCTGCTGAGG TGCCCCAGTA AAAAAAAGTA CAAGGCCATC 4301 CGGCTCATTT TTGTCATCAT GGCGGTGTTT TTCATTTTCT GGACACCCTA
4351 CAATGTGGCT ATCCTTCTCT CTTCCTATCA ATCCATCTTA TTTGGAAATG
4401 ACTGTGAGCG GAGCAAGCAT CTGGACCTGG TCATGCTGGT GACAGAGGTG
4451 ATCGCCTACT CCCACTGCTG CATGAACCCG GTGATCTACG CCTTTGTTGG
4501 AGAGAGGTTC CGGAAGTACC TGCGCCACTT CTTCCACAGG CACTTGCTCA 4551 TGCACCTGGG CAGATACATC CCATTCCTTC CTAGTGAGAA GCTGGAAAGA
4601 ACCAGCTCTG TCTCTCCATC CACAGCAGAG CCGGAACTCT CTATTGTGTT
4651 T or a degenerate variation thereof. The 5' genomic DNA flanking sequence encoding the human eosinophil eotaxin receptor further comprises the region beginning with nucleotide 1 and ending with nucleotide 3586 as depicted below (SEQ ID NO:3):
1 GGATCCCTAC CTTCCCCATC AGAGCTAGGG GGCATGGAGC GCTCTCTGCT
51 AAGATGGGGA CCCCCAAGGA ATGTCTCCCT GTGGGGCACT TCCTTACCAG 101 ATGGGATGGC CAGTGCGGTT AAGTTGGTGG TCAGGCAGAA AAAAAAGATC
151 TAGTTTGTAC TCTTGAGAGT TCCTCGGTTT GTTCATGGCA TGGGCAGGGA
201 GTCAAGGAGC AGCAGCCTTG CCTCAGTGCC TACCAGTGCA GGAAAAGGTG
251 CATAGCCTGG GCCAGGGCCA GGGCCCTGGT GGAGGCGTAG TGGTAACAGA
301 GAGGGCTCTC CATTCCAGCC CAAGGAAGAC TAAGAATGAA TACCTCATGA 351 GTATATTAGC TACAAACCAC CACAGCAGGT TCCAGAAAAA GGCTCAGCGT
401 TGGAACCAGG TCACCCCCAC TCAGCAGACA CCAGTCATAT AAATCAAGGA
451 CCAACAGGAG ACAGGAACAC CCCCTTCCCA CTCTGCCCCA TGTCTCAAGT
501 TGTAGTGGCC CTTCCTCCAG ATCTCTGCCA CCATCTTAGA AAGGAACACT
551 GAAAGAAGAA ACTGAAATTA TAAGCTGACA GCATAAAGAG GATGAGTAAA 601 ACCTAAAATC ATTGTTCACA TGAATGAATC AAGAGAAGTT TAAACCACTT
651 TGGACTAAAA TGTGTGAATC CTTTTTCCTG CTATCCAGCA GATGAGAAGC
701 TGGTAACAGA GACCACAATA GTTTGGAGAC TAAAGAATCA TTGCACATTT
751 CACTGCTGAG TTGTATTGTG AGTAATTTTA GTTGACCTCA CTTTGTAAAT
801 CTTGCACACG GGGCAATCCA ATATCTGCAC AAGAGATATG TTAACCAGTG 851 GTAAATGCTG CATGAGGAGA TTGGGTGATT TTTACTTTCG TTTTTGTGCT
901 CTTCTTTCTT ATTGTTCTTA CTTATTTACG ATTACCCTAT CGTTTTCCCA
951 AAATGTAAAA GGCCATTTTG AAAGCCTAAT TCAAACCTCT TCACTATTTT
1001 GTATCTAAGT ATTCACCTTG ATTGAGACTG GGTAGACAGG TGAAAACCAT
1051 ATCAGGTTTT TAATTTTTTA ATTTTTAATT ATTTATTTAT TTATTTATTT 1101 TTTGAGATGG AGTCTGGCTG TCGCCCAGGC TGGAGTGCAG CGGCGTGATC
1151 ACAGTTCACT GCAGCCTCAA CCTTCTAGGC TCAAGGGATT CTCCCACCTC 1201 AGCCCCCCAA GTAGTTGGGA CCACACGTAT GCGCCACCAT GCCTGGCTAA
1251 TTTCTTATTT TTTTGTAGAG ATAGGATCTC ACTATATTGT CCAGGCTGGT
1301 CTTGAATTCC TGGGCTCAGG TGAGCCTCCC ACCTGGGCCT CCCAAAGTAC
1351 TGGGATTACA GGCATGAGCC AAGGTCCCCT GCCCATATGA GATTTTCTGT
1401 CTCTGATCCC ATGCAGCTAG TAATCAAGGA CTTGGCTGCT GACTCTGGAG
1451 GACCTGCATG CTTTCTTGAG CTGTGAACTT CAGTGCTAAA AGCTCATAGG
1501 CAGCCCTGAA ACCCAAACCA AAAGGTTCTA TGGTTTATCA TCCTGATCAT 1551 GTTGATTTTA TAGAAATAAC ACATGAATTA AAGACACTAC CCTCAAACTG
1601 AGCAAAACTT AAGTAATTTT TTTAAAGTTT GACCTGTTTT TAAATCACTC
1651 TTGGAGAAAA AGGAAAATAA ATACAAATAA TTAACGGTGA ATACAGGCTA
1701 CTATACCTTT GTTCTCCAGA ATTAGCAGTT CTGTTCTTTT CTTGCTTTAG
1751 ATGCTGAAGT GCAGAAGGAC ACTCTGTGAT TGTACGTGTG TAACTGACAA 1801 AATGTGTATT TTTTTTCTCA GCTGCTATGG ATTGGATTAT GCTATTATGA
1851 ATAAGAATGC TGATGGGAGC ACACACAAAC CATTTGTTCC TCAGTCCATT
1901 TTCCTCCTCA AAAGCCTGGA ATGTGCCATT GATCAGTGGG AGATGTACCT
1951 GGACAGACCC ATGAAAAGAG ATCAACAAGT TCCACCCAAG GGACCCTATT
2001 TTTCCTAATT TCATTTGAAA TGGCTTCTAA TTGTCCTTCT TTCATTCCTG 2051 CTTCCTACCA GTTTTACAGC TTTTTCTGGT TTCAAATGTG AACTCACATA
2101 CACTCTCATT TTTCCTCATC ACAACCCCAA GTGACCCAAT GGTCCTCACT
2151 TTCGATATAA GTAAAGGAGG CTCTGCATTA AGGGCTTGTC CAAGGCACGC
2201 AGCTGAGAGG CGCTAGGACT GGCTCCATTT CCATCTCTAT TCTCACTGAC
2251 TTTGACTACC CAGAACCCCA ACATGTGGGG CCTCAGTATT CGATCAATTA 2301 TTCTATTAAG AAGCAAAAAC AATTCCCCGC ATTGGCCCCA GTTATTAAGC
2351 ATTTCTCAGA TTTACCTTGA GAAATGCCCA TCGGCCTGTA TATTCACATC
2401 TTCACCCTTG TCCCTTCCTC CTAGAAAGGA GAAAGTCAGT TGGATGCCCT
2451 CTGAGGAACT AGTGCATGGC TTAACTGTCC TTCCATGACT CCTGCCTTAT
2501 CTGTTTTCTA TTTTCCTCCT TTTCCACCGA AGTCTATAAT CTCAAGAAAA 2551 GCAGGCACTG GCCTTAGGGC TCCTGGCCTA AGAAATATCA AGTCCAGTGA 2601 GAAATCCCAT TGACTGACCC CTCCTGCTTA CCCCTTTGTG ATGGAGAAGC
2651 TCCCAGGGGT TTGCTTTTTG CATGTTACCA GGCCTAACTC AGCATCACCA
2701 GGGGCAAGAA AAGGAAAGTA ACCTAAACTA ATGCTGCTTA TAATTGTAAT
2751 TATTGTAATA GTTAATTACT GTGATTGTAC ATGTGTAACA GACAAAATGT
2801 GTATTTTTTT CACAGCTGCT GTGGATTGGA TTATGCCATT TGGAATAAGA
2851 ATGCTGTTAA GAGCACACAA GCCAGGTTCC TCAAGTCCGT AGCAAATTTT
2901 TCAAAAGTTA AATTTAAAAA TCACTACATT TGAATCTAGT GACAGGAGAA 2951 ATGGACATGG ATAGAGACTA AAGATCTAGC CCAAATTTTA TATTTACTTG
3001 TTAGAGGATT TTGAACAAAT TACTAAATTT CTTCAAGGTT CAATTTCCCC
3051 ATTAACTATA ATGAATGTCT CATCATTATG GGGCCCTGGA GAAGCATAAT
3101 TACTTGTAAT TGTAATAATC ATTGTTATTA TTATTATACA TATTTTGCTT
3151 TTAAATGGAT AAGGATTTTT AAGGTATATG TAAACTGTAA AACATAAAAT 3201 GCAAAATGCC GTAAGAGACA GTAGTAATAA TAATGATTAT TATATTGTTA
3251 TCATTATCTA GCCTGTTTTT TCCTGTTGTG TATTTCTTCC TTTAAATGCT
3301 TACAGAAATC TGTATCCCCA TTCTTCACCA CCACCCCACA ACATTTCTGC
3351 TTCTTTTCCC ATGCCGGTCA TGCTAACTTT GAAAGCTTCA GCTCTTTCCT
3401 TCCTCAATCC TTCTCCTGGC ACCTCTGATA TGCCTTTTGA AATTCATGTT 3451 AAAGAATCCC TAGGCTGCTA TCACATGTGG CATCTTTGTT GAGTACATGA
3501 ATAAATCAAC TGGTGTGTTT TACGAAGGAT GATTATGCTT CATTGTGGGA
3551 TTGTATTTTT CTTCTTCTAT CACAGGGAGA AGTGAA
degenerate variation thereof.
The sequence for the cDNA encoding human eosinophil eotaxin receptor further comprises the terminator region beginning with nucleotide 4652 and ending with nucleotide 5099 as depicted below (SEQ ID NO:4):
4652 TAGGTCAGA TGCAGAAAAT TGCCTAAAGA GGAAGGACCA AGGAGATGAA
4701 GCAAACACAT TAAGCCTTCC ACACTCACCT CTAAAACAGT CCTTCAAACT 4751 TCCAGTGCAA CACTGAAGCT CTTGAAGACA CTGAAATATA CACACAGCAG
4801 TAGCAGTAGA TGCATGTACC CTAAGGTCAT TACCACAGGC CAGGGGCTGG
4851 GCAGCGTACT CATCATCAAC CCTAAAAAGC AGAGCTTTGC TTCTCTCTCT
4901 AAAATGAGTT ACCTACATTT TAATGCACCT GAATGTTAGA TAGTTACTAT
4951 ATGCCGCTAC AAAAAGGTAA AACTTTTTAT ATTTTATACA TTAACTTCAG 5001 CCAGCTATTG ATATAAATAA AACATTTTCA CACAATACAA TAAGTTAACT
5051 ATTTTATTTT CTAATGTGCC TAGTTCTTTC CCTGCTTAAT GAAAAGCTT
or a degenerate variation thereof.
As will be appreciated by one skilled in the art, there is a substantial amount of redundancy in the set of codons which translate specific amino acids. Accordingly, this invention also includes alternative base sequences wherein a codon (or codons) are replaced with another codon such that the amino acid sequence translated by the DNA sequences remains unchanged. For purposes of this specification, a sequence bearing one or more such replaced codons will be defined as a degenerate variation. Also included are mutations (exchanges of individual amino acids) which one skilled in the art would expect to have no effect on functionality, such as valine for leucine, arginine for lysine, and asparagine for glutamine. The amino acid sequence of CC CKR3 shares some sequence homology with CC CKR1 (Neote, K., et al. (1993) Cell 72, 415-425), CC CKR2B (Charo, I. F., et al. (1994) Proceecing ofthe National Academy of Sciences 91, 2752-2756), CC CKR4 (Power, C. A., et al. (1995) Journal of Biological Chemistry 270, 19495-19500) and V28 (Raport, C. J., et al. (1995) Gene 163, 295-299). The sequence of this protein, designated CC CKR3, is comparable to that previously reported by Combadiere et al. (Combadiere, C, et al. (1995) Journal of Biological Chemistry 270, 16491-16494) except that it contains a lysine in place of asparagine at position 107. Genomic cloning provided confirmation of the subject sequence, including lysine at position 107. The sequence discrepency, which results from a substitution of G to T at the third position of the codon for residue 107, could represent a genetic polymorphism. This is highly unlikely, however, because all α- and β-chemokine receptors analyzed to date contain lysine in that position including the recently described basophilic β-chemokine receptor (Power, C. A., et al. (1995) Journal of Biological Chemistry 270, 19495-19500), CC CKR1 (Neote, K., et al. (1993) Cell 72, 415-425), MCP-1R (Charo, I. F., et al. (1994) Proceecing of the National Academy of Sciences 91, 2752-2756), IL-8RA and IL-8RB (Holmes, W. E., et al. (1991) Science 253, 1278-1280; Murphy, P. M., et al. (1991) Science 253, 1280-1283), the three murine β-chemokine receptors (Post, T. W., et al. (1995) Journal of Immunology 155, 5299-5305; Gao, J. L., et al. (1995) Journal of Biological Chemistry 270, 17494-17501) as well as three human chemokine-like receptors (Loetscher, M., et al. (1994) Journal of Biological Chemistry 269, 232-237; Raport, C. J., et al. (1995) Gene 163, 295-299; Combadiere, C, et al. (1995) DNA and Cell Biology 14, 673-680; Federsppiel, B., et al. (1993) Genomics 16, 707-712). An unusual feature of CC CKR3 is the cluster of negatively charged amino acids (ETEELFEET) distal to TMIV in the second extracellular loop. Expression of the Human CC CKR3 in AML14.3D10 Cells
Once a full length cDNA encoding CC CKR3 was isolated and cloned into the expression vector pBJ/NEO the resulting plasmid designated pBJ/NEO/CCCKR3, was transfected into the AML14.3D10 line.
The CC CKR3 transfected AML14.3D10 cell line has been placed on restricted deposit with American Type Culture Collection in Rockville, Maryland as ATCC No. CRL- 12079, on April 5, 1996.
Stable clones were selected for neomycin resistance, and a number were chosen for further analysis. To demonstrate expression of receptor protein, a western blot was performed using antisera generated against a peptide derived from the predicted C-terminus of CC CKR3. Immunoreactive bands migrating at approximately 45-55 kd are present in primary eosinophils and the 3.16 clone, indicating that CC CKR3 is indeed expressed in these cells. There was no immunoreactive bands present in neutrophils indicating that the antisera was indeed identifying an eosinophil-specific protein. A nonspecific pattern of immuno- reactivity was detected in untransfected AML14.3D10 cells, and furthermore, this pattern was identical in clone 3.49 indicating that this neomycin-resistant clone is a non-expressor of CC CKR3. Of the 27 neomycin resistant clones studied, 19 failed to express CC CKR3. The other 8 did express the receptor as judged both by Western analysis, and by the ability of eotaxin and RANTES to induce Ca2+-fluxes. The non- expressing clones were used as negative controls in subsequent experiments.
Binding to CC CKR3 on Intact AML14/CCCKR3.16 Cells
Because preliminary experiments with three different CC CKR3 expressing clones indicated that they bound 125τ_eotaxin, competition studies using this labeled ligand were performed to characterize the binding properties of the receptor. As shown in Table 1, unlabeled human eotaxin competed with an Kd of 0.1 nM. Results with murine eotaxin were essentially identical. Scatchard analysis demonstrated that eotaxin binds with a single affinity and that the different clones expressed 2-4 xlO^ receptors/cell. The ability to bind eotaxin is due to CC CKR3 since neither immunoreactive negative clones, such as 3.49, nor untransfected cells displayed any specific binding. Clearly, CC CKR3 is a high affinity receptor for eotaxin. Cross-competition studies with the two other β-chemokines known to be eosinophil chemoattractants, RANTES and MCP-3, demonstrated that they too have considerable affinity for CC CKR3, with Kd's of about 3 nM (See Table 1). In contrast, MCP-1 competed with much lower affinity (Kd=60 nM), and MlP-lα, and MIP-lβ failed to compete at all (See Table 1). Similarly, the α-chemokine IL-8 did not inhibit eotaxin binding.
Competition studies were also carried out against 125ι_ MCP-3. Again, human and murine eotaxin competed strongly with Kd's of 0.2 and 0.3 nM (Table 1). RANTES and MCP-3 also demonstrated high affinity with Kd's of 0.5 and 0.7 nM, values about 4-fold lower than observed against eotaxin. As in the studies with eotaxin, MCP-1 competed weakly (Kd = 16 nM), and MlP-lα, and MlP-lβ failed to competete at all. Thus despite some small quantitative differences the overall ligand selectivity of the receptor is the same whether measured by competition against eotaxin or MCP-3, and the order of potency, eotaxin>MCP-3=RANTES»MCP-l, is identical.
CC CKR3 is functionally coupled in AML14.3D10 cells
In order to determine whether human CC CKR3 was functionally coupled when expressed in the AML14.3D10 line, intracellular Ca2+ levels were measured in response to various β- chemokines. Both 100 nM eotaxin and RANTES induced Ca2+-fluxes in cells expressing the receptor. Suφrisingly, lμM of MCP-3 was required to induce a response, and that response was smaller than those observed for eotaxin or RANTES. No response at all was generated by addition of MlP-lα, MlP-lβ, MCP-1 or IL-8 at concentrations as high as lμM. The responses to eotaxin, RANTES, and MCP-3 are due to the specific expression of CC CKR3 since none of these mediators induced fluxes in untransfected cells or in clone 3.49. While the preliminary functional characterization by Combadiere et. al. differs greatly from the present invention, they were not able to demonstrate any specific binding to cells putatively expressing the receptor, and such functional data have now been retracted (Combadiere, C, et al. (1995) Journal of Biological Chemistry 270, 30235).
Binding properties of primary eosinophils
The selectivity of CC CKR3 for the various β-chemokines mirrors the effectiveness of these ligands as eosinophil chemoattractants suggesting that CC CKR3 is the primary mediator of chemokine induced eosinophil chemotaxis. To provide additional pharmacological evidence, binding studies were conducted on primary eosinophils. When measured by competition against 125τ_eotaχin, unlabeled human eotaxin gave an Kd = 0.1 nM, a value identical to that obtained on cloned CC CKR3 (see Table 1). Scatchard analysis showed a single binding affinity, and 4 x 10$ sites/cell. The number of binding sites varied by less than 2-fold for the 3 donors used in the studies. The affinites for RANTES and MCP-3 were also identical to those measured on CC CKR3, and as with CC CKR3, neither MlP-lα , or MIP-lβ, showed any ability to compete with radiolabeled eotaxin (see Table 1). Similarly, the Kd's obtained by competition against 125ι_MCP-3 on eosinophils are effectively indistinguishable to those measured against cloned CC CKR3 (see Table 1). All of the observations and measurements, taken together with the Western blot showing expression of CC CKR3, verify that CC CKR3 is the eosinophil eotaxin receptor, and appears to be largely responsible for mediating the effects of most β-chemokines on eosinophils.
Stably expressed in the eosinophilic line AML14.3D10, CC CKR3 binds eotaxin, RANTES and MCP-3, with high affinity, with a rank order of potency of eotaxin>RANTES=MCP-3. MCP-1 binds with much lower affinity, while MlP-lα and MlP-lβ fail to bind at all. The selectivity of CC CKR3 mirrors most of the binding activity of primary eosinophils. In fact, when measured by competition against 125τ_ eotaxin, the binding affinities on eosinophils for all of these β- chemokines are indistinguishable from those obtained with cloned CC CKR3. Moreover, CC CKR3 was cloned from eosinophils, and as shown by Western blotting is heavily expressed in these cells. The abilities of the different chemokines to activate CC CKR3 are consistent with the binding data as eotaxin, RANTES, and to a lessor extent MCP- 3 all stimulate Ca^+ fluxes in clones which express the receptor, while MCP-1, MlP-lα and MIP-lβ do not, even at concentrations as high as 1 μM. Thus, based on its properties, and expression, CC CKR3, is the eosinophil eotaxin receptor.
TABLE 1
Binding affinities of various chemokines comparing CC CKR3 expressed in AML14.3D10 with primary eosinophils
Figure imgf000028_0001
competitor CC CKR3 eosinophils
125τ_numan eotaxin human-eotaxin 0.1 ±0.04 (4) 0.1±0.03 (3) murine-eotaxin 0.1 ±0.04 (3) O. l±O.Ol (2)
MCP-3 2.7±1.7 (5) 3.0±0.2 (2)
RANTES 3.1±0.6 (5) 2.6±0.3 (2)
MCP-1 60±9 (3) 41±2 (2)
MlP-la N.B. (4) N.B. (2)
MlP-lβ N.B. (4) N.B. (2)
Figure imgf000028_0002
human-eotaxin 0.2±0.1 (4) 0.2±0.1 (2) murine-eotaxin 0.3±0.1 (2) 0.2±0.1 (3)
MCP-3 0.7±0.4 (4) l.l±O.ό (10)
RANTES 0.5±0.3 (4) 0.9±0.4 (8)
MCP-1 16±2 (3) 61±13 (2)
MlP-la N.B. (4) see text
MlP-lβ N.B. (4) N.B. (2)
Competition binding experiments were carried out against the indicated iodinated ligand as follows and as described herein. Equilibrium binding of β-chemokines to AML14.3D10 cells expressing CC CKR3 and to primary eosinophils was examined with increasing concentrations of unlabelled human eotaxin, murine eotaxin, RANTES, MCP-3, or MCP-1 to compete against fixed concentrations of either 125ι_human eotaxin, or 125i-MCP-3. Also the competition with 100 nM concentrations of MlP-lα, and MIP-lβ was examined. The experiments were carried out either with AML14.3D10 cells expressing CC CKR3, or with eosinophils. All values are the averages of triplicate determinations. Typically, 4000-6000 cpm of iodinated ligand was bound in the absence of compeititor with S/N ratios exceeding 15. Human and murine eotaxin are the human and murine chemokines, respectively. "N.B." means that no competition was observed. All results are the averages of the number of experiments shown in parenthesis.
Various changes and modifications may be made in the products and processes of the present invention without departing from the spirit and scope thereof. The various embodiments and the examples which have been set forth herein are given for the puφose of illustrating the present invention and shall not be construed as being limitations on the scope or spirit of the instant invention.
EXAMPLE 1
mRNA Isolation and cDNA Cloning
Total RNA was isolated from purified eosinophils with TRIzol reagent (BRL) and used in a RT/PCR reaction (Daugherty, B. L., et al. (1991) Nucleic Acids Research 19, 2471-2476) using oligonucleotide primers designed from the human CC CKR1 and MCP- 1RB cDNA sequences (Neote, K., et al. (1993) Cell 72, 415-425; Charo, I. F., et al. (1994) Proceecing of the National Academy of Sciences 91, 2752-2756). The primers used for PCR corresponded to a consensus sequence encoded in transmembrane domains (TM) II and VII: 5*-PCR primer (TMII) (SEQ ID NO:5):
5'-AACCTGGCCAT(C,T)TCTGA(C,T)CTGC-3' 3'-RT/PCR primer (TMVII) (SEQ ID NO:6):
5'-GAAC(C,T)TCTC(C,A)CCAACGAAGGC.
The resultant PCR product of -700 bp was subcloned into plasmid pNoTA (Five Prime, Three Prime, Ine) and sequenced using Sequenase (USB). The remaining 5' and 3' sequence encoding CC CKR3 was cloned by rapid amplification of cDNA ends (RACE) using both the 5'-RACE and 3'-RACE kits (Clontech) with the following primer sequences: (5 -RACE) (SEQ ID NO:7): 5'-TCTCGCTGTACAAGCCTGTGTG-3';
(3'-RACE) (SEQ ID NO:8):
5 -CCTTCTCTCTTCCTATCAATCC-3*. The resultant PCR products (5'-RACE, -450 bp; 3*-RACE, -700 bp) were subcloned into pCRII (Invitrogen) and sequenced. Upon identification of the 5'-end of the cDNA containing the initiator ATG codon and the 3'-end containing the termination codon TAG, a new set of PCR primers were designed to reamplify the entire coding region from eosinophil total RNA for expression of CC CKR3. The primer sequences used for RT/PCR were: 5'-PCR primer (SEQ ID NO:9):
5 -ATATATTAAGCTTCCACCATGACAACCTCACTAGATACAG- 3*; 3'-RT/PCR primer (SEQ ID NO: 10):
5 -ATATATTCTAGAGCGGCCGCTAAAACACAATAGAGAGTTCC- 3'. The resultant PCR product of 1105 bp was digested with
Hindill and Notl and subcloned into plasmid pBJ/NEO to yield pBJ/NEO/CCCKR3. The plasmid pBJ/NEO was prepared essentially as follows. Plasmid pD5/Igh/Neo (Daugherty, B.L., et al. (1991) Nucleic Acids Research 19, 2471-2476) was digested with the restriction enzyme Sail, filled in with E. coli DNA polymerase I Klenow fragment to create a blunt end and subsequently digested with the restriction enzyme Notl. The CM VIE intron A fragment from plasmid p89-l 1 was digested with Clal, filled in to create a blunt end and subsequently digested with Hindill. These fragments were used in a three-way ligation with a Hindill and Notl fragment of the human C5a receptor cDNA. The C5a receptor fragment was excised with Hindill and Notl and replaced with the eotaxin receptor cDNA of 1105 bp obtained by RT/PCR with oligonucleotides SEQ ID NO:9 and SEQ ID NO: 10 after digestion with Hindill and Notl. Several clones were sequenced and one clone comprising the consensus sequence was chosen for expression of CC CKR3 in heterologous cells.
EXAMPLE 2
Transfection into AML14.3D10 Human Eosinophilic Cell Line
AML14.3D10 cells (Paul, C. C, et al. (1995) Blood 86, 3737-3744) were cultured in RPMI-1640, 10% FBS, 1 mM sodium pyruvate, 0.5 μM β-mercaptoethanol and 2 mM L-glutamine (complete medium). Cells were harvested at a density of 0.3 x 10°7mL, washed once in PBS, resuspended in RPMI at 107/mL, and 25 μg of plasmid was added. Electroporation was carried out at 300 V, 960 μF using a Gene Pulser (BioRad). Following electroporation, cells were chilled at 0°C for 10 min and then plated in complete medium at 10^/T75 flask and cultured at 37°C, 5% C02- After 16-24 hr, cells were pelleted and resuspended in complete medium containing 2 mg/mL Geneticin (GIBCO). Cells were maintained in selection medium for 8-10 days until individual surviving clusters appeared. Invididual cells were then transferred to 96-well plates and expanded. AML14/CCCKR3 sublines were assayed for the ability to generate a Ca^+ flux in response to either RANTES or eotaxin. Positive sublines were then probed by western blotting with an antibody raised against the predicted C- terminus of CC CKR3. Cell lines positive in both sets of assays were then characterized for their ability to bind to a variety of CC chemokines, including eotaxin, RANTES, MCP-3, MlP-lα, MlP-lβ and MIP-1. EXAMPLE 3
Purification of Eosinophils
Primary eosinophils were isolated from granulophoresis preparations obtained from allergic and asthmatic donors (Bach, M. K., et al. (1990) Journal of Immunological Methods 130, 277-281). The leukocytes were mixed with equal volumes of HBSS and layered over LSM (Organon Teknika) as described (Rollins, T. E., et al. (1988) Journal of Biological Chemistry 263, 520-526). After lysis of erythrocytes with NH4CI, the granulocytes were subsequently treated with anti-CD 16 microbeads followed by MACS separation (Miltenyl Biotech) (Hansel, T. T., et al. (1991) Journal of Immunological Methods 145, 105-110). Typically the eosinophil preparations were >99% pure as determined using the LeukoStat staining kit (Fisher).
EXAMPLE 4
Generation of α-CC CKR3 antisera and immunoblotting
Polyclonal rabbit antisera was generated to CC CKR3 using the C-terminal decapeptide sequence TAEPELSIVF. Peptide synthesis, coupling to thyroglobulin and production of antisera was performed (Miller, D. K., et al. (1993) Journal of Biological Chemistry 268, 18062-18069). Whole cells were boiled and sonicated in Laemli sample buffer (Laemmli, U. K. (1970) Nature 227, 680-685), electrophoresed on 4-20% SDS gels (Novex), transferred to polyvinylidene difluoride membranes (BioRad), and blocked with 5% nonfat dry milk in TBST (20 mM Tris, 200 mM NaCl, 0.1% Tween-20) for 16 hr at 4°C. The membrane was incubated with antisera at 1: 1000 in TBST for 1 hr at room temperature, washed, and subsequently incubated with goat anti- rabbit HRP (Zymed) at 1:4000 in TBST for 30 min also at room temperature. After washing, the membrane was treated with ECL western blotting reagents (Amersham) for 1 min, covered in plastic wrap and exposed to film for 2 min. EXAMPLE 5
Chemokine binding assays
Recombinant MCP-3, MCP-1, RANTES, murine and human eotaxin were obtained from Peprotech (Princeton, NJ). 125ι_ MCP-3 and 125τ_MIP-lα were obtained from New England Nuclear (Boston, MA), and ^i-human-eotaxin was obtained from Amersham. Binding of 125τ_ιabeled ligands (typically a total of 2 x 104 cpm) in the presence of varying concentrations of unlabeled ligands to intact cells (typically 1.5 x 104 , 10^, or 10^ for experiments with labeled eotaxin, MCP-3, or MlP-lα, respectively) were performed at 32°C (Van Riper, G., et al. (1993) Journal of Experimantal Medicine 111, 851-856).
EXAMPLE 6
Ligand-induced Ca2+ fluxes
Human CC CKR3 expressing AML14 clones or purified eosinophils were incubated with 1.25 μg/ml Indo-I (Molecular Probes, Eugene, OR) in RPMI 1640, 10 mM HEPES, 5% FBS, for 60 min at 37°C (Van Riper, G., et al. (1993) Journal of Experimantal Medicine 111, 851-856). Loaded cells were washed and incubated at 37°C before the addition of ligands. Calcium fluxes were performed on a FACS analyzer (Becton Dickinson & Co., Mountain View, CA) with an excitation wavelenth of 365 nm and dual emission wavelength of 405 and 488 nm.
EXAMPLE 7
CC CKR3 Binding Assay Assay buffer (50 mM Hepes, pH 7.2 w/ 0.5% BSA, 5 mM
MgCl2, 1 mM CaCl2, 100 uM PMSF and 10 ug/ml phosphoramidon, leupeptin, aprotinin and chymostatin), test compound (or equivalent volume of solvent), 20 pM 125i-human eotaxin (2000 Ci/mmol), 25 ng unlabeled human eotaxin (non-specific binding wells only), and AML14.3D10 cells expressing eotaxin receptor cells, or eosinophils, are added sequentially in 96-well, round-bottom, polystyrene plates to a final volume of 250 uL. Assay plates are then mixed and incubated for 60 minutes at 31°C. After incubation, assay plates are harvested onto Packard 96-well GF/C Unifilter plates treated with 0.33% polyethylenimine (PEI) using Packard Filtermate 196 cell harvester. Wells and filters are washed with 200 uL 50 mM Hepes, pH 7.2 with 0.5M NaCl and 0.02% NaN3. After filtration, GF/C plates are dried and sealed. 25 uL Packard Microscint-O scintillant are then added to each well and counted for 2 minutes on Packard Topcount (liquid 125j setting).
EXAMPLE 8
Phosphoinositide 3-kinase (PI-3K) Assay
AML14.3D10 expressing eotaxin receptor (CCCKR3) cells are incubated with test compound and stimulated with eotaxin, RANTES, or MCP-3, pelleted and lysed in 1 mL lysis buffer (1% Nonidet P-40, 100 mM NaCl, 20 mM Tris, pH 7.4, 10 mM iodoacetamide, 46 mM b- glyceraphosphate, 10 mM NaF, 1 mM PMSF, 1 ug/mL leupeptin, 1 ug/mL chymostatin, 1 ug/mL antipain, 1 ug/mL pepstatin A, and 1 mM sodium orthovanadate). Ly sates are then pre-cleared for 1 hr with uncoupled protein A Affi-Gel beads. Immunoprecipitation is then performed with p85 polyclonal antiserum (1 ul/mL lysate; Upstate Biologies, New York, NY), coupled to protein A Affi-Gel beads (Bio- Rad) at 4°C for 2 hr. Immunoprecipitates are washed and subjected to in vitro lipid kinase assays by using a lipid mixture, 100 ul 0.1 mg/ml Ptdlns and 0.1 mg/ml phosphatidylserine dispersed by sonication into solution in 20 mM HEPES, pH 7.0, and 1 mM EDTA. The reaction is initiated by the addition of 100 mM ATP and 20 uCi [gamma-32P]ATP (3000 Ci/mmol) in 20 ul kinase buffer. The reaction is then terminated after 15 min and the phosphoinositide lipids are separated by thin layer chromatography (TLC) and visualized by exposure to iodine vapor autoradiography. EXAMPLE 9
Chemotaxis Assay
AML14.3D10 expressing eotaxin receptor cells are isolated by centrifugation (van Riper, G., et al. (1994) J. Immunol. 152, 4055- 4061) for 15 min at 150 X g, washed and resuspended at 10' cells/ml in HBSS (pH 7.4) containing 1 mM CaCl2 and 1 mM MgCl2 (chemotaxis buffer). The chemotaxis experiments is then performed in Transwell dishes (6.5 mm, Costar, Cambridge, MA). The lower chamber contains 0.6 ml of chemotaxis buffer and is separated from the upper chamber containing 10" cells by a 5-mm pore Nucleopore polycarbonate membrane (Nucleopore Coφoration, Pleasanton, CA). After a 15 min preincubation at 37°C, test compound and eotaxin, RANTES, or MCP-3 are added to the lower chamber to a final concentration of 300 nM. After 2 hrs at 37°C, the upper chamber inserts are removed, and the cells that migrate to the lower chamber are enumerated by a Coulter Counter (Coulter Electronics, Hialeah, FL).
EXAMPLE 10
Ligand-Dependent Inositol Phosphate Release Assay
AML14.3D10 expressing eotaxin receptor cells are labeled with [3H] inositol (10 uCi/ml) for 24 hrs as described (Wu, D., et al. (1993) Science 261, 101-103). Test compound and arious concentrations of eotaxin, MCP-3, or RANTES are then added to the cells for 30 min. The cells are lysed in 10% perchloric acid, neutralized in 2 N KOH and centrifuged. The supernatant is transferred to columns containing 0.5 ml AG1-X8 anion exchange resin, washed with 6 ml borax buffer and eluted with 0.3 ml formic acid (0.1 M). The eluted samples are mixed with scintillation cocktail and counted. EXAMPLE 1 1
Acidification Rate Assay
AML14.3D10 expressing eotaxin receptor cells are subject to serum starvation for 16 hrs. The cells are then mixed at a 3: 1 (v/v) ratio with low melting temperature agarose. A 10 ul drop of the cell/agarose mixture is pipetted into a sterile Capsule Cup (Molecular Devices) at a cell density of approximately 200,000 cells/cup. The cell/agarose drop forms a gel after about 5 min, and is assembled into the cup between two 3 um porosity polycarbonate membranes with running medium. The assembled capsule cups is placed into the sensor chambers and then placed on the Cytosensor Microphysiometer (Molecular Devices) containing 1 ml of running medium. The chambers are allowed to equilibrate for 1 hr at 37°C with a flow rate of 100 ul/min. The experiment is initiated with an 8 min exposure of eotaxin, RANTES, MCP-3 and test compound at various concentrations and the acidification rate over baseline will be measured in the medium (McConnell, H.M., et al. (1992) Science 257, 1906-1912) until the cells return to the unstimulated level.
EXAMPLE 12
Actin Polymerization Assay
AML14.3D10 expressing eotaxin receptor cells are diluted in APA buffer (HBSS; 25mM Hepes; 0.2% BSA, pH7.2) at a concentration of 4 x lOfyml. One ul of test compound and eotaxin, RANTES, or MCP-3 added into a 96-well plate and incubated at 37°C. 100 ml of cells are added to the plate and incubated for 20 sec to which 100 ml of APA cocktail (2 mis 8% formaldehyde; 460uLs 0.33uM Rhodamine- phalloidin; 1.85 mg 200ug/ml lysolecithin; 7.25 mis HBSS) is added. Plates are then centrifuged at 2000 RPM for 5 min, cleared and then 100 ul of HBSS is added to all wells which are read in a Fluoroskan II fluorometer. While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention. For example, procedures other than the particular experimental procedures as set forth herein above may be applicable as a consequence of degeneracy and variations in the sequences of the proteins and DNA of the invention indicated above. Likewise, the characterization data observed may vary slightly according to and depending upon the particular assay or characterization method employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. It is intended, therefore, that the invention be defined by the scope of the claims which follow and that such claims be inteφreted as broadly as is reasonable.
SEQUENCE LISTING
(1) GENERAL INFORMATION
(i) APPLICANT: DAUGHERTY, BRUCE L. DEMARTINO, JULIE A. SICILIANO, SALVATORE J. SPRINGER, MARTIN S.
(ii) TITLE OF THE INVENTION: EOSINOPHIL EOTAXIN RECEPTOR
(iii) NUMBER OF SEQUENCES: 4
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Merck & Co. Inc.
(B) STREET: P.O. Box 2000, 126 E. Lincoln Ave.
(C) CITY: Rahway
(D) STATE: NJ
(E) COUNTRY: USA
(F) ZIP: 07065-0900
(V) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Diskette
(B) COMPUTER: IBM Compatible
(C) OPERATING SYSTEM: DOS
(D) SOFTWARE: FastSEQ for Windows Version 2.
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: 60/016,15E
(B) FILING DATE: 26-APR-1996
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Eric Thies, J.
(B) REGISTRATION NUMBER: 35,382
(C) REFERENCE/DOCKET NUMBER: 19634Y
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 908-594-3904
(B) TELEFAX: 908-594-4720
(C) TELEX: (2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 355 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
Met Thr Thr Ser Leu Asp Thr Val Glu Thr Phe Gly Thr Thr Ser Tyr
1 5 10 15
Tyr Asp Asp Val Gly Leu Leu Cys Glu Lys Ala Asp Thr Arg Ala Leu
20 25 30
Met Ala Gin Phe Val Pro Pro Leu Tyr Ser Leu Val Phe Thr Val Gly
35 40 45
Leu Leu Gly Asn Val Val Val Val Met He Leu He Lys Tyr Arg Arg
50 55 60
Leu Arg He Met Thr Asn He Tyr Leu Leu Asn Leu Ala He Ser Asp 65 70 75 80
Leu Leu Phe Leu Val Thr Leu Pro Phe Trp He His Tyr Val Arg Gly
85 90 95
His Asn Trp Val Phe Gly His Gly Met Cys Lys Leu Leu Ser Gly Phe
100 105 110
Tyr His Thr Gly Leu Tyr Ser Glu He Phe Phe He He Leu Leu Thr
115 120 125
He Asp Arg Tyr Leu Ala He Val His Ala Val Phe Ala Leu Arg Ala
130 135 140
Arg Thr Val Thr Phe Gly Val He Thr Ser He Val Thr Trp Gly Leu 145 150 155 160
Ala Val Leu Ala Ala Leu Pro Glu Phe He Phe Tyr Glu Thr Glu Glu
165 170 175
Leu Phe Glu Glu Thr Leu Cys Ser Ala Leu Tyr Pro Glu Asp Thr Val
180 185 190
Tyr Ser Trp Arg His Phe His Thr Leu Arg Met Thr He Phe Cys Leu
195 200 205
Val Leu Pro Leu Leu Val Met Ala He Cys Tyr Thr Gly He He Lys
210 215 220
Thr Leu Leu Arg Cys Pro Ser Lys Lys Lys Tyr Lys Ala He Arg Leu 225 230 235 240
He Phe Val He Met Ala Val Phe Phe He Phe Trp Thr Pro Tyr Asn
245 250 255
Val Ala He Leu Leu Ser Ser Tyr Gin Ser He Leu Phe Gly Asn Asp
260 265 270
Cys Glu Arg Ser Lys His Leu Asp Leu Val Met Leu Val Thr Glu Val
275 280 285
He Ala Tyr Ser His Cys Cys Met Asn Pro Val He Tyr Ala Phe Val
290 295 300
Gly Glu Arg Phe Arg Lys Tyr Leu Arg His Phe Phe His Arg His Leu 305 310 315 320
Leu Met His Leu Gly Arg Tyr He Pro Phe Leu Pro Ser Glu Lys Leu
325 330 335
Glu Arg Thr Ser Ser Val Ser Pro Ser Thr Ala Glu Pro Glu Leu Ser 340 345 350 He Val Phe 355
(2) INFORMATION FOR SEQ ID NO:2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1065 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
ATGACAACCT CACTAGATAC AGTTGAGACC TTTGGTACCA CATCCTACTA TGATGACGTG 60
GGCCTGCTCT GTGAAAAAGC TGATACCAGA GCACTGATGG CCCAGTTTGT GCCCCCGCTG 120
TACTCCCTGG TGTTCACTGT GGGCCTCTTG GGCAATGTGG TGGTGGTGAT GATCCTCATA 180
AAATACAGGA GGCTCCGAAT TATGACCAAC ATCTACCTGC TCAACCTGGC CATTTCGGAC 240
CTGCTCTTCC TCGTCACCCT TCCATTCTGG ATCCACTATG TCAGGGGGCA TAACTGGGTT 300
TTTGGCCATG GCATGTGTAA GCTCCTCTCA GGGTTTTATC ACACAGGCTT GTACAGCGAG 360
ATCTTTTTCA TAATCCTGCT GACAATCGAC AGGTACCTGG CCATTGTCCA TGCTGTGTTT 420
GCCCTTCGAG CCCGGACTGT CACTTTTGGT GTCATCACCA GCATCGTCAC CTGGGGCCTG 480
GCAGTGCTAG CAGCTCTTCC TGAATTTATC TTCTATGAGA CTGAAGAGTT GTTTGAAGAG 540
ACTCTTTGCA GTGCTCTTTA CCCAGAGGAT ACAGTATATA GCTGGAGGCA TTTCCACACT 600
CTGAGAATGA CCATCTTCTG TCTCGTTCTC CCTCTGCTCG TTATGGCCAT CTGCTACACA 660
GGAATCATCA AAACGCTGCT GAGGTGCCCC AGTAAAAAAA AGTACAAGGC CATCCGGCTC 720
ATTTTTGTCA TCATGGCGGT GTTTTTCATT TTCTGGACAC CCTACAATGT GGCTATCCTT 780
CTCTCTTCCT ATCAATCCAT CTTATTTGGA AATGACTGTG AGCGGAGCAA GCATCTGGAC 840
CTGGTCATGC TGGTGACAGA GGTGATCGCC TACTCCCACT GCTGCATGAA CCCGGTGATC 900
TACGCCTTTG TTGGAGAGAG GTTCCGGAAG TACCTGCGCC ACTTCTTCCA CAGGCACTTG 960
CTCATGCACC TGGGCAGATA CATCCCATTC CTTCCTAGTG AGAAGCTGGA AAGAACCAGC 1020
TCTGTCTCTC CATCCACAGC AGAGCCGGAA CTCTCTATTG TGTTT 1065 (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3586 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
GGATCCCTAC CTTCCCCATC AGAGCTAGGG GGCATGGAGC GCTCTCTGCT AAGATGGGGA 60
CCCCCAAGGA ATGTCTCCCT GTGGGGCACT TCCTTACCAG ATGGGATGGC CAGTGCGGTT 120
AAGTTGGTGG TCAGGCAGAA AAAAAAGATC TAGTTTGTAC TCTTGAGAGT TCCTCGGTTT 180
GTTCATGGCA TGGGCAGGGA GTCAAGGAGC AGCAGCCTTG CCTCAGTGCC TACCAGTGCA 240
GGAAAAGGTG CATAGCCTGG GCCAGGGCCA GGGCCCTGGT GGAGGCGTAG TGGTAACAGA 300
GAGGGCTCTC CATTCCAGCC CAAGGAAGAC TAAGAATGAA TACCTCATGA GTATATTAGC 360
TACAAACCAC CACAGCAGGT TCCAGAAAAA GGCTCAGCGT TGGAACCAGG TCACCCCCAC 420
TCAGCAGACA CCAGTCATAT AAATCAAGGA CCAACAGGAG ACAGGAACAC CCCCTTCCCA 480
CTCTGCCCCA TGTCTCAAGT TGTAGTGGCC CTTCCTCCAG ATCTCTGCCA CCATCTTAGA 540
AAGGAACACT GAAAGAAGAA ACTGAAATTA TAAGCTGACA GCATAAAGAG GATGAGTAAA 600
ACCTAAAATC ATTGTTCACA TGAATGAATC AAGAGAAGTT TAAACCACTT TGGACTAAAA 660
TGTGTGAATC CTTTTTCCTG CTATCCAGCA GATGAGAAGC TGGTAACAGA GACCACAATA 720
GTTTGGAGAC TAAAGAATCA TTGCACATTT CACTGCTGAG TTGTATTGTG AGTAATTTTA 780
GTTGACCTCA CTTTGTAAAT CTTGCACACG GGGCAATCCA ATATCTGCAC AAGAGATATG 840
TTAACCAGTG GTAAATGCTG CATGAGGAGA TTGGGTGATT TTTACTTTCG TTTTTGTGCT 900
CTTCTTTCTT ATTGTTCTTA CTTATTTACG ATTACCCTAT CGTTTTCCCA AAATGTAAAA 960
GGCCATTTTG AAAGCCTAAT TCAAACCTCT TCACTATTTT GTATCTAAGT ATTCACCTTG 1020
ATTGAGACTG GGTAGACAGG TGAAAACCAT ATCAGGTTTT TAATTTTTTA ATTTTTAATT 1080
ATTTATTTAT TTATTTATTT TTTGAGATGG AGTCTGGCTG TCGCCCAGGC TGGAGTGCAG 1140
CGGCGTGATC ACAGTTCACT GCAGCCTCAA CCTTCTAGGC TCAAGGGATT CTCCCACCTC 1200
AGCCCCCCAA GTAGTTGGGA CCACACGTAT GCGCCACCAT GCCTGGCTAA TTTCTTATTT 1260
TTTTGTAGAG ATAGGATCTC ACTATATTGT CCAGGCTGGT CTTGAATTCC TGGGCTCAGG 1320 TGAGCCTCCC ACCTGGGCCT CCCAAAGTAC TGGGATTACA GGCATGAGCC AAGGTCCCCT 1380
GCCCATATGA GATTTTCTGT CTCTGATCCC ATGCAGCTAG TAATCAAGGA CTTGGCTGCT 1440
GACTCTGGAG GACCTGCATG CTTTCTTGAG CTGTGAACTT CAGTGCTAAA AGCTCATAGG 1500
CAGCCCTGAA ACCCAAACCA AAAGGTTCTA TGGTTTATCA TCCTGATCAT GTTGATTTTA 1560
TAGAAATAAC ACATGAATTA AAGACACTAC CCTCAAACTG AGCAAAACTT AAGTAATTTT 1620
TTTAAAGTTT GACCTGTTTT TAAATCACTC TTGGAGAAAA AGGAAAATAA ATACAAATAA 1680
TTAACGGTGA ATACAGGCTA CTATACCTTT GTTCTCCAGA ATTAGCAGTT CTGTTCTTTT 1740
CTTGCTTTAG ATGCTGAAGT GCAGAAGGAC ACTCTGTGAT TGTACGTGTG TAACTGACAA 1800
AATGTGTATT TTTTTTCTCA GCTGCTATGG ATTGGATTAT GCTATTATGA ATAAGAATGC 1860
TGATGGGAGC ACACACAAAC CATTTGTTCC TCAGTCCATT TTCCTCCTCA AAAGCCTGGA 1920
ATGTGCCATT GATCAGTGGG AGATGTACCT GGACAGACCC ATGAAAAGAG ATCAACAAGT 1980
TCCACCCAAG GGACCCTATT TTTCCTAATT TCATTTGAAA TGGCTTCTAA TTGTCCTTCT 2040
TTCATTCCTG CTTCCTACCA GTTTTACAGC TTTTTCTGGT TTCAAATGTG AACTCACATA 2100
CACTCTCATT TTTCCTCATC ACAACCCCAA GTGACCCAAT GGTCCTCACT TTCGATATAA 2160
GTAAAGGAGG CTCTGCATTA AGGGCTTGTC CAAGGCACGC AGCTGAGAGG CGCTAGGACT 2220
GGCTCCATTT CCATCTCTAT TCTCACTGAC TTTGACTACC CAGAACCCCA ACATGTGGGG 2280
CCTCAGTATT CGATCAATTA TTCTATTAAG AAGCAAAAAC AATTCCCCGC ATTGGCCCCA 2340
GTTATTAAGC ATTTCTCAGA TTTACCTTGA GAAATGCCCA TCGGCCTGTA TATTCACATC 2400
TTCACCCTTG TCCCTTCCTC CTAGAAAGGA GAAAGTCAGT TGGATGCCCT CTGAGGAACT 2460
AGTGCATGGC TTAACTGTCC TTCCATGACT CCTGCCTTAT CTGTTTTCTA TTTTCCTCCT 2520
TTTCCACCGA AGTCTATAAT CTCAAGAAAA GCAGGCACTG GCCTTAGGGC TCCTGGCCTA 2580
AGAAATATCA AGTCCAGTGA GAAATCCCAT TGACTGACCC CTCCTGCTTA CCCCTTTGTG 2640
ATGGAGAAGC TCCCAGGGGT TTGCTTTTTG CATGTTACCA GGCCTAACTC AGCATCACCA 2700
GGGGCAAGAA AAGGAAAGTA ACCTAAACTA ATGCTGCTTA TAATTGTAAT TATTGTAATA 2760
GTTAATTACT GTGATTGTAC ATGTGTAACA GACAAAATGT GTATTTTTTT CACAGCTGCT 2820
GTGGATTGGA TTATGCCATT TGGAATAAGA ATGCTGTTAA GAGCACACAA GCCAGGTTCC 2880
TCAAGTCCGT AGCAAATTTT TCAAAAGTTA AATTTAAAAA TCACTACATT TGAATCTAGT 2940
GACAGGAGAA ATGGACATGG ATAGAGACTA AAGATCTAGC CCAAATTTTA TATTTACTTG 3000 TTAGAGGATT TTGAACAAAT TACTAAATTT CTTCAAGGTT CAATTTCCCC ATTAACTATA 3060
ATGAATGTCT CATCATTATG GGGCCCTGGA GAAGCATAAT TACTTGTAAT TGTAATAATC 3120
ATTGTTATTA TTATTATACA TATTTTGCTT TTAAATGGAT AAGGATTTTT AAGGTATATG 3180
TAAACTGTAA AACATAAAAT GCAAAATGCC GTAAGAGACA GTAGTAATAA TAATGATTAT 3240
TATATTGTTA TCATTATCTA GCCTGTTTTT TCCTGTTGTG TATTTCTTCC TTTAAATGCT 3300
TACAGAAATC TGTATCCCCA TTCTTCACCA CCACCCCACA ACATTTCTGC TTCTTTTCCC 3360
ATGCCGGTCA TGCTAACTTT GAAAGCTTCA GCTCTTTCCT TCCTCAATCC TTCTCCTGGC 3420
ACCTCTGATA TGCCTTTTGA AATTCATGTT AAAGAATCCC TAGGCTGCTA TCACATGTGG 3480
CATCTTTGTT GAGTACATGA ATAAATCAAC TGGTGTGTTT TACGAAGGAT GATTATGCTT 3540
CATTGTGGGA TTGTATTTTT CTTCTTCTAT CACAGGGAGA AGTGAA 3586
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 448 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4 :
TAGGTCAGAT GCAGAAAATT GCCTAAAGAG GAAGGACCAA GGAGATGAAG CAAACACATT 60
AAGCCTTCCA CACTCACCTC TAAAACAGTC CTTCAAACTT CCAGTGCAAC ACTGAAGCTC 120
TTGAAGACAC TGAAATATAC ACACAGCAGT AGCAGTAGAT GCATGTACCC TAAGGTCATT 180
ACCACAGGCC AGGGGCTGGG CAGCGTACTC ATCATCAACC CTAAAAAGCA GAGCTTTGCT 240
TCTCTCTCTA AAATGAGTTA CCTACATTTT AATGCACCTG AATGTTAGAT AGTTACTATA 300
TGCCGCTACA AAAAGGTAAA ACTTTTTATA TTTTATACAT TAACTTCAGC CAGCTATTGA 360
TATAAATAAA ACATTTTCAC ACAATACAAT AAGTTAACTA TTTTATTTTC TAATGTGCCT 420
AGTTCTTTCC CTGCTTAATG AAAAGCTT 448

Claims

WHAT IS CLAIMED IS:
1. An eosinophil eotaxin receptor, free from receptor-associated proteins.
2. The eosinophil eotaxin receptor of Claim 1 which is human.
3. An isolated eosinophil eotaxin receptor.
4. The eosinophil eotaxin receptor of Claim 3 which is human.
5. The receptor of Claim 2 which comprises a full length receptor or which comprises the amino acid sequence (SEQ ID
NO: l).
6. The receptor of Claim 4 which comprises a full length receptor or which comprises the amino acid sequence (SEQ ID NO: l).
8. A functional equivalent of the eosinophil eotaxin receptor of Claim 1.
9. A nucleic acid which encodes an eosinophil eotaxin receptor receptor or a functional equivalent, said nucleic acid being free from associated nucleic acids.
10. A nucleic acid according to Claim 9 which encodes a human eosinophil eotaxin receptor, or a functional equivalent. 11. The nucleic acid of Claim 10 which is a DNA.
12. The nucleic acid of Claim 11 which comprises the nucleotide sequence (SEQ ID NO:2).
13. The nucleic acid of Claim 12 which further comprises the nucleotide sequence (SEQ ID NO:3).
14. The nucleic acid of Claim 13 which further comprises the nucleotide sequence (SEQ ID NO:4).
15. A vector comprising a nucleic acid which encodes an eosinophil eotaxin receptor receptor, or a functional equivalent.
16. A vector according to Claim 15 which is selected from the group consisting of: plasmids, and modified viruses, yeast artificial chromosomes, bacteriophages and cosmids or transposable elements
17. A vector according to Claim 16 wherein the nucleic acid encodes human eosinophil eotaxin receptor receptor or a functional equivalent.
18. A host cell comprising a vector according to Claim 17.
19. The host cell of Claim 18 wherein the nucleic acid encodes human eosinophil eotaxin receptor receptor, or a functional equivalent.
20. The host cell of Claim 19 which is from the AML14.3D10 cell line. 21. A nucleic acid encoding an eosinophil eotaxin receptor receptor clone that belongs to the β-chemokine receptor family and that hybridizes with a nucleotide which a human eosinophil eotaxin receptor under reduced stringency of hybridization.
22. A method to determine the presence of a compound which binds to an eosinophil eotaxin receptor comprising:
(a) introducing a nucleic acid which encodes an eosinophil eotaxin receptor into a cell under conditions so that eosinophil eotaxin receptor is expressed;
(b) introducing a detector molecule or a nucleic acid encoding a detector molecule into the cell, wherein the detector molecule is directly or indirectly responsive to a eosinophil eotaxin- ligand binding event; (c) contacting the cell with a compound suspected of binding to the eosinophil eotaxin receptor; and
(d) determining whether the compound binds to the eosinophil eotaxin receptor by monitoring the detector molecule.
23. The method of Claim 22 wherein the eosinophil eotaxin receptor is human.
24. The method of Claim 22 wherein the nucleic acid which encodes an eosinophil eotaxin receptor comprises the nucleotide sequence (SEQ ID NO:2).
25. The method of Claim 22 wherein the result of step (d) is compared to that obtained using a known ligand of the eosinophil eotaxin receptor.
26. The method of Claim 25 wherein the known ligand of the eosinophil eotaxin receptor is eotaxin. 27. The method of Claim 25 wherein the known ligand of the eosinophil eotaxin receptor is RANTES.
28. The method of Claim 25 wherein the known ligand of the eosinophil eotaxin receptor is MCP-3.
29. The method of Claim 22 wherein the eosinophil eotaxin receptor is expressed in a host cell which does not naturally express the human eosinophil eotaxin receptor
30. The method of Claim 29 wherein the host cell is from the AML14.3D10 cell line.
31. A ligand identified by the method of Claim 22.
32. A ligand identified by the method of Claim 23.
33. A ligand identified by the method of Claim 24.
34. A ligand identified by the method of Claim 25.
35. A ligand identified by the method of Claim 30.
PCT/US1997/006568 1996-04-26 1997-04-24 Eosinophil eotaxin receptor WO1997041154A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP97925399A EP1012190A4 (en) 1996-04-26 1997-04-24 Eosinophil eotaxin receptor
JP53897097A JP3689118B2 (en) 1996-04-26 1997-04-24 Eosinophil eotaxin receptor
CA002252850A CA2252850A1 (en) 1996-04-26 1997-04-24 Eosinophil eotaxin receptor

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US1615896P 1996-04-26 1996-04-26
US1711396P 1996-04-26 1996-04-26
US60/017,113 1996-04-26
US60/016,158 1996-04-26
GB9700894.0 1997-01-17
GBGB9700894.0A GB9700894D0 (en) 1997-01-17 1997-01-17 Eosinophil eotain receptor

Publications (1)

Publication Number Publication Date
WO1997041154A1 true WO1997041154A1 (en) 1997-11-06

Family

ID=27268678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/006568 WO1997041154A1 (en) 1996-04-26 1997-04-24 Eosinophil eotaxin receptor

Country Status (4)

Country Link
EP (1) EP1012190A4 (en)
JP (1) JP3689118B2 (en)
CA (1) CA2252850A1 (en)
WO (1) WO1997041154A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1998050434A1 (en) * 1997-05-07 1998-11-12 Schering Corporation Methods for eosinophil depletion and compositions therefor
WO1999066037A2 (en) * 1998-06-17 1999-12-23 Recherches Expertises Et Developpement Medicaux Parenz Inc. Antisense oligonucleotides for treating or preventing atopic diseases and neoplastic cell proliferation
WO2000062814A2 (en) * 1999-04-21 2000-10-26 Children's Hospital Medical Center Intracellular pharmaceutical targeting
US6207155B1 (en) 1997-05-07 2001-03-27 Schering Corporation Method of eosinophil depletion with antibody to CCR 3 receptor
WO2003014153A2 (en) * 2001-08-10 2003-02-20 Topigen Pharmaceutique Inc. Cellular virus receptors and methods of use.
US6994977B2 (en) 1995-01-19 2006-02-07 Children's Medical Center Corporation Method of identifying inhibitors of C—C chemokine receptor 3
WO2009137912A1 (en) * 2008-05-15 2009-11-19 Topigen Pharmaceuticals Inc. Oligonucleotides for treating inflammation and neoplastic cell proliferation
US8119790B2 (en) 2004-10-29 2012-02-21 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5401629A (en) * 1990-08-07 1995-03-28 The Salk Institute Biotechnology/Industrial Associates, Inc. Assay methods and compositions useful for measuring the transduction of an intracellular signal

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6806061B1 (en) * 1995-01-19 2004-10-19 Children's Medical Center Corporation G protein-coupled receptor gene and methods of use therefor
US7265201B1 (en) * 1995-06-23 2007-09-04 Millennium Pharmaceuticals, Inc. Human chemotactic cytokine
US6265184B1 (en) * 1995-12-20 2001-07-24 Icos Corporation Polynucleotides encoding chemokine receptor 88C

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5401629A (en) * 1990-08-07 1995-03-28 The Salk Institute Biotechnology/Industrial Associates, Inc. Assay methods and compositions useful for measuring the transduction of an intracellular signal

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
DAMM VAN J., ET AL.: "STRUCTURAL AND FUNCTIONAL IDENTIFICATION OF TWO HUMAN, TUMOR- DERIVED MONOCYTE CHEMOTACTIC PROTEINS (MCP-2 AND MCP-3) BELONGING TO THE CHEMOKINE FAMILY.", THE JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 176., 1 January 1992 (1992-01-01), US, pages 59 - 65., XP000673185, ISSN: 0022-1007, DOI: 10.1084/jem.176.1.59 *
DAUGHERTY B. L., ET AL.: "CLONING, EXPRESSION, AND CHARACTERIZATION OF THE HUMAN EOSINOPHIL EOTAXIN RECEPTOR.", THE JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 183., no. 05., 1 May 1996 (1996-05-01), US, pages 2349 - 2354., XP000600043, ISSN: 0022-1007, DOI: 10.1084/jem.183.5.2349 *
PONATH P. D., ET AL.: "CLONING OF THE HUMAN EOSINOPHIL CHEMOATTRACTANT, EOTAXIN. ÖEXPRESSION, RECEPTOR BINDING, AND FUNCTIONAL SUGGEST A MECHANISM FOR THE SELECTIVE RECRUITMENT OF EOSINOPHILS.", JOURNAL OF CLINICAL INVESTIGATION, AMERICAN SOCIETY FOR CLINICAL INVESTIGATION, US, vol. 97., no. 03., 1 February 1996 (1996-02-01), US, pages 604 - 612., XP000571506, ISSN: 0021-9738, DOI: 10.1172/JCI118456 *
PONATH P. D., ET AL.: "MOLECULAR CLONING AND CHARACTERIZATION OF A HUMAN EOTAXIN RECEPTOR EXPRESSED SELECTIVELY ON EOSINOPHILS.", THE JOURNAL OF EXPERIMENTAL MEDICINE, ROCKEFELLER UNIVERSITY PRESS, US, vol. 183., no. 06., 1 June 1996 (1996-06-01), US, pages 2437 - 2448., XP002915743, ISSN: 0022-1007, DOI: 10.1084/jem.183.6.2437 *
SCHALL T. J., ET AL.: "A HUMAN T CELL-SPECIFIC MOLECULE IS A MEMBER OF A NEW GENE FAMILY.", THE JOURNAL OF IMMUNOLOGY, THE AMERICAN ASSOCIATION OF IMMUNOLOGISTS, US, vol. 141., no. 03., 1 August 1988 (1988-08-01), US, pages 1018 - 1025., XP002040096, ISSN: 0022-1767 *
See also references of EP1012190A4 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7402657B2 (en) 1995-01-19 2008-07-22 Millennium Pharmaceuticals, Inc. C-C chemokine receptor 3 proteins
US7012133B1 (en) 1995-01-19 2006-03-14 Children's Medical Center Corp. Antibodies to C-C chemokine Receptor 3 Protein
US6994977B2 (en) 1995-01-19 2006-02-07 Children's Medical Center Corporation Method of identifying inhibitors of C—C chemokine receptor 3
WO1998050434A1 (en) * 1997-05-07 1998-11-12 Schering Corporation Methods for eosinophil depletion and compositions therefor
US6207155B1 (en) 1997-05-07 2001-03-27 Schering Corporation Method of eosinophil depletion with antibody to CCR 3 receptor
US6822087B1 (en) 1998-06-17 2004-11-23 Topigen Pharmaceutiques Inc. Antisense oligonucleotides for treating or preventing atopic diseases and neoplastic cell proliferation
WO1999066037A2 (en) * 1998-06-17 1999-12-23 Recherches Expertises Et Developpement Medicaux Parenz Inc. Antisense oligonucleotides for treating or preventing atopic diseases and neoplastic cell proliferation
WO1999066037A3 (en) * 1998-06-17 2000-04-20 Rech Expertises Et Dev Medicau Antisense oligonucleotides for treating or preventing atopic diseases and neoplastic cell proliferation
US7629324B2 (en) 1998-06-17 2009-12-08 Topigen Pharmaceutiques Inc. Antisense oligonucleotides for treating atopic diseases and neoplastic cell proliferation
WO2000062814A3 (en) * 1999-04-21 2001-03-15 Childrens Hosp Medical Center Intracellular pharmaceutical targeting
US6358697B2 (en) 1999-04-21 2002-03-19 Children's Hospital Medical Center Intracellular pharmaceutical targeting
WO2000062814A2 (en) * 1999-04-21 2000-10-26 Children's Hospital Medical Center Intracellular pharmaceutical targeting
WO2003014153A3 (en) * 2001-08-10 2003-08-28 Topigen Pharma Inc Cellular virus receptors and methods of use.
WO2003014153A2 (en) * 2001-08-10 2003-02-20 Topigen Pharmaceutique Inc. Cellular virus receptors and methods of use.
US8119790B2 (en) 2004-10-29 2012-02-21 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
US8202851B2 (en) 2004-10-29 2012-06-19 Topigen Pharmaceuticals Inc. Antisense oligonucleotides for treating allergy and neoplastic cell proliferation
WO2009137912A1 (en) * 2008-05-15 2009-11-19 Topigen Pharmaceuticals Inc. Oligonucleotides for treating inflammation and neoplastic cell proliferation
US8470999B2 (en) 2008-05-15 2013-06-25 Luc Paquet Oligonucleotides for treating inflammation and neoplastic cell proliferation

Also Published As

Publication number Publication date
JP3689118B2 (en) 2005-08-31
EP1012190A4 (en) 2004-04-28
JP2002503950A (en) 2002-02-05
CA2252850A1 (en) 1997-11-06
EP1012190A1 (en) 2000-06-28

Similar Documents

Publication Publication Date Title
JP4117904B2 (en) CC-KRKR-5, CC-chemokine receptor, derivatives thereof and use thereof
Horuk Chemokine receptors
CA2265915C (en) Cxcr3 chemokine receptor, antibodies, nucleic acids, and methods of use
US6730301B1 (en) MCP-1 receptor antibodies
JP3415162B2 (en) IL-13 receptor polypeptide
Zhao et al. Chemokine receptor CCR5 functionally couples to inhibitory G proteins and undergoes desensitization
AU3402697A (en) Uses of a chemokine receptor for inhibiting hiv-1 infection
JPH11178588A (en) Cdna clone my1 encoding new human 7-trans membrane receptor
JPH10510719A (en) Human G protein chemokine receptor HDGNR10
CZ20011026A3 (en) Isolated polynucleotide, expression vector, cultivated cell, DNA construct, production process of protein, isolated polypeptide, production process of antibody, antibody per se and detection method
WO1997041154A1 (en) Eosinophil eotaxin receptor
JP2000083669A (en) Human splicing variant cxcr4b of cxcr4 kemokine receptor
US20070065849A1 (en) Nucleic acid encoding eosinophil eotaxin receptor
WO1997041225A2 (en) Mammalian mixed lymphocyte receptors, chemokine receptors [mmlr-ccr]
AU2009200390A1 (en) Type 2 cytokine receptor and nucleic acids encoding same
US20020086359A1 (en) Novel guanosine triphosphate (GTP) binding protein-coupled receptor proteins
Youn et al. Molecular cloning and characterization of a cDNA, CHEMR1, encoding a chemokine receptor with a homology to the human CC chemokine receptor, CCR-4
KR100462431B1 (en) Novel mouse cxc chemokine receptor
JPH10327879A (en) Cdna clone hnfdy20 encoding new human 7-transmemberane receptor
AU2002343671A1 (en) Type 2 cytokine receptor and nucleic acids encoding same
Youn et al. The Seventh Transmembrane Domain of CC Chemokine Receptor 5 Is Critical for MIP-1β Binding and Receptor Activation: Role of Met 287
JPH1118782A (en) Novel human neurotensin receptor type 2 and its spliced variant
US20050118675A1 (en) Heptahelix receptor and its use as leukotriene B4 receptor
WO2001088130A1 (en) Molecular cloning and identification of a human leukotriene-like receptor ltxr
CA2562162A1 (en) Human g-protein chemokine receptor hdgnr10

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

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: PAT.BUL.47/97, UNDER INID(30)"PRIORITY DATA",REPLACE"08/640991"BY"60/017113"

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

Ref document number: 2252850

Country of ref document: CA

Ref country code: CA

Ref document number: 2252850

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1997925399

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1997925399

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1997925399

Country of ref document: EP