EP0907734A2 - GALANIN RECEPTOR GalR2 - Google Patents

GALANIN RECEPTOR GalR2

Info

Publication number
EP0907734A2
EP0907734A2 EP97929814A EP97929814A EP0907734A2 EP 0907734 A2 EP0907734 A2 EP 0907734A2 EP 97929814 A EP97929814 A EP 97929814A EP 97929814 A EP97929814 A EP 97929814A EP 0907734 A2 EP0907734 A2 EP 0907734A2
Authority
EP
European Patent Office
Prior art keywords
leu
galr2
galanin
ala
ctg
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP97929814A
Other languages
German (de)
French (fr)
Inventor
Brian T. Bloomquist
Michael L. Mccaleb
Linda J. Cornfield
Heeja Yoo-Warren
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer AG
Bayer Corp
Original Assignee
Bayer AG
Bayer Corp
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 US08/665,034 external-priority patent/US6410686B1/en
Application filed by Bayer AG, Bayer Corp filed Critical Bayer AG
Publication of EP0907734A2 publication Critical patent/EP0907734A2/en
Withdrawn legal-status Critical Current

Links

Definitions

  • This invention relates to a novel neuropeptide galanin receptor and its nucleic acid sequence.
  • Galanin is a 29 amino acid peptide hormone (30 amino acids in human) which is present in a wide range of central and peripheral tissues. Skofitsch, Peptides 6, 509 (1985); Merchenthaler, Prog. Neurobiol. 40, 711 (1993). Galanin is involved in many diverse physiological functions. Galanin is known to regulate the secretion of both endocrine and exocrine hormones. Galanin inhibits insulin secretion from pancreatic beta cells, and can inhibit pancreatic amylase secretion; in the stomach, galanin inhibits gastrin and somatostatin secretion.
  • Galanin stimulates VIP (vasoactive intestinal protein) release from the hypothalamus, prolactin and growth hormone release from the pituitary; and inhibits the secretion of ACTH in the hypothalamus. Furthermore, the secretion of neurotransmitters can be modulated. For example, galanin can inhibit the release of histamine and norepinephrine in the hypothalamus. Other secondary messenger systems are also regulated: galanin can either stimulate or inhibit intracellular cAMP accumulation; is involved in the closure of N- and L-type voltage-sensitive calcium channels, and in the opening of ATP-sensitive and - insensitive potassium channels; and has been shown to stimulate the release of calcium from intracellular stores. Moreover, galanin is involved in the inhibition of acetylcholine release and the inhibition of muscarinic receptor-mediated phosphoinositide turnover. Bartfai, Crit. Rev. Neurobiol. 7, 229 (1993)
  • Galanin is implicated in the modulation of many cognitive and sensory functions. Galanin has potent antinociceptive effects, and can impair performance in one-trial learning, t-maze, and swim maze learning and memory paradigms. Its inhibition of the anoxic release of glutamate, as well as its inhibitory actions on cholinergic function suggest a role in neuroprotection, and in the development of Alzheimer's Disease. Crawley, Regulatory Peptides 59, 1 (1995). Galanin is known to induce feeding in rodents and, in contrast with the effects of Neuropeptide Y on feeding, galanin increases preference for fat intake. Akabayashi, Proc. Natl. Acad. Set. USA 91, 10375 (1994).
  • Galanin is also involved in the regulation of gastrointestinal smooth muscle contraction. Because of the important role of galanin in these many physiological processes, there is a strong need to further develop materials and methods for investigating the mechanistic behavior of the receptors and for treating diseased and other abnormal states associated with these physiological processes.
  • GalRl receptor has been obtained from rat. Burgevin, J. Mol. Neurosci. 6, 33 (1995). The in vivo functions mediated through this cloned GalRl receptor have not yet been elucidated.
  • EP-0711830-A2 disclose a closely-related GalRl sequence, differing in that Cysl5 ⁇ Trp is varied.
  • the present invention provides a novel galanin receptor protein, the GalR2 receptor. Also provided are the nucleic acid sequences encoding this novel receptor protein as well as methods for using this protein and its nucleic acid sequence, and methods useful for developing and identifying compounds for the treatment of diseases and disorders in which galanin is implicated. The importance of this discovery is manifested in the effects of galanin, which include antinociceptive activity, smooth muscle contraction, cardiovascular activity, pituitary hormone release, cognition, and increased food intake. Thus, this receptor protein is useful for screening for galanin agonist and antagonist activity for controlling these conditions.
  • nucleic acid sequences encoding the complete rat receptor and partial sequences of the human receptor.
  • These nucleic acid sequences have a variety of uses. For example, they are useful for making vectors and for transforming cells, both of which are ultimately useful for production of the GalR2 receptor protein. They are also useful as scientific research tools for developing nucleic acid probes for determining receptor expression levels, e.g., to identify diseased or otherwise abnormal states. They are useful for developing analytical tools such as antisense oligonucleotides for selectively inhibiting expression of the receptor gene to determine physiological responses.
  • the present invention can also be used to isolate the homologous nucleic acid sequence of other species, such as human, primate, dog, mouse, etc.
  • a homogeneous composition comprising the receptor GalR2 protein.
  • the protein is useful for screening drugs for agonist and antagonist activity, and, therefore, for screening for drugs useful in regulating physiological responses associated with the GalR2 receptor.
  • antagonists to the GalR2 receptor could be used to treat obesity and diabetes by reducing appetite and food consumption, whereas agonists could be used for the treatment of anorexic conditions.
  • drugs could be used to treat Alzheimer's disease, stroke, neuropathic pain, and/or endocrine disorders.
  • the proteins are also useful for developing antibodies for detection of the protein.
  • vectors such as plasmids, comprising the receptor GalR2 nucleic acid sequence that may further comprise additional regulatory elements, e.g., promotors,
  • additional regulatory elements e.g., promotors
  • transformed cells that express the GalR2 receptor
  • nucleic acid probes e.g., nucleic acid probes
  • antisense oligonucleotides e.g., agonists, (f) antagonists, and (g) transgenic mammals.
  • transgenic mammals e.g., transgenic mammals.
  • Further aspects of the invention comprise methods for making and using the foregoing compounds and compositions.
  • the invention includes polynucleotide molecules coding for a rat or human GalR2, or a galanin binding fragment thereof.
  • a purified and isolated rat or human GalR2 protein The GalR2 protein comprising SEQ ID NO:2.
  • the full-length GalR2 protein the partial sequence of which is indicated in SEQ ID NO:6.
  • SEQ ID NO:3 or a galanin binding fragment thereof.
  • a polynucleotide molecule comprising SEQ ID NO: 3 or a degenerate variant thereof.
  • a purified and isolated variant of rat GalR2 protein comprising SEQ ID NO:4.
  • Figure 1 is the polynucleotide sequence of rat GalR2 of the invention.
  • Figure 2 is the amino acid sequence of rat GalR2.
  • Figures 3-4 are the polynucleotide sequence of the Y107 variant of rat GalR2.
  • Figure 5 is the amino acid sequence of Yl 07(omitting putative intron).
  • Figure 6 is the partial polynucleotide sequence of human GalR2.
  • Figure 7 is the partial amino acid sequence of human GalR2.
  • Figure 8 is the representative saturation isotherm of [ 125 I]hGalanin binding to rat GalR2
  • the present invention comprises, in part, a novel galanin receptor protein, the GalR2 receptor.
  • Particularly preferred embodiments of the GalR2 receptor are those having an amino acid sequence substantially the same as SEQ ID NO: 2, 4 or 6.
  • reference to the GalR2 receptor is meant as a reference to any protein having an amino acid sequence substantially the same as SEQ ID NO: 2, 4 or 6.
  • the present invention also comprises the nucleic acid sequence encoding the GalR2 protein, which nucleic acid sequence is substantially the same as SEQ ID NO: 1, 3 or 5.
  • Receptors SEQ ID NO: 1 and SEQ ID NO: 3 are nucleic acid sequences of rat GalR2 receptors but SEQ ID NO: 3 appears to contain an intronic region; therefore, SEQ ID NO: 1 is the preferred embodiment of the rat GalR2 receptor of this invention.
  • Receptor SEQ ID NO: 5 is the partial nucleic acid sequence of human GalR2.
  • Receptors SEQ ID NO: 2 and 4 are rat GalR2 receptors and appear to be allelic variants.
  • Receptor SEQ ID NO: 6 is the partial amino acid sequence of human GalR2.
  • a protein "having an amino acid sequence substantially the same as SEQ ID NO: x" means a protein whose amino acid sequence is the same as SEQ ID NO: x or differs only in a way such that IC 50 [galanin] as determined according to the method detailed in Example 2, infra, are less than or equal to 1 nM.
  • a molecule having a nucleotide sequence substantially the same as SEQ ID NO: y means a nucleic acid encoding a protein "having an amino acid sequence substantially the same as SEQ ID NO: y*" (wherein “y*” is the number of the amino acid sequence for which nucleotide sequence "y” codes) as defined above.
  • This definition is intended to encompass natural allelic variations in the GalR2 sequence.
  • Cloned nucleic acid provided by the present invention may encode GalR2 protein of any species of origin, including (but not limited to), for example, mouse, rat, rabbbit, cat, dog, primate, and human.
  • the nucleic acid provided by the invention encodes GaIR2 receptors of mammalian, and most preferably, rat or human origin.
  • the invention also includes nucleotide sequences encoding chimeric proteins comprised of parts of the GalR2 receptor and parts of other related seven-transmembrane receptors.
  • the BMB77 clone (SEQ ID NO: 1) (see Example 1, infra) has a 1.7-kb cDNA insert with a open reading frame from nucleotides 279 to 1394 that encodes a 372 amino acid protein (SEQ ID NO: 2).
  • Hydrophobicity plot analysis using the PEPPLOT function of GCG shows that the GalR2 receptor has seven transmembrane-like domains, indicating it might be a G-protein-coupIed receptor.
  • GalR2 is 26 amino acids longer in length than GalRl, the only other published galanin receptor. This extra length of amino acids within GalR2 is due to an extended C-terminal tail sequence. However, the putative N-terminal extracellular domain of GalR2 is about 7 amino acids shorter than the corresponding region in GalRl. It is also important to note that the GaIR2 sequence shows only 38% amino acid sequence identity to the GalRl receptor.
  • the Y107 clone (SEQ ID NO: 3) ⁇ see Example 1, infra) has a 1.9-kb cDNA insert with an open reading frame from nucleotides 17-384, and from nucleotides 874-1621.
  • the sequence between nucleotides 384 and 874 contains multiple STOP codons in all three reading frames.
  • the dinucleotides GT and AG at positions 385-386 and 872- 873, respectively, fulfill the criteria for being a splice donor and acceptor site, respectively.
  • the partial human GalR2 nucleic acid sequence (SEQ ID NO: 5) contains a 337 amino acid opening reading frame.
  • the initial leucine residue of this partial human GalR2 protein (SEQ ID NO: 6) corresponds to amino acid Leu 51 in rat GalR2 (SEQ ID NO: 2 and 4).
  • Nucleic acid hybridization probes provided by the invention are DNAs consisting essentially of the nucleotide sequences complementary to any sequence depicted in SEQ ID NO:s 1 and 3 that is effective in nucleic acid hybridization.
  • Nucleic acid probes are useful for detecting GalR2 gene expression in cells and tissues using techniques well-known in the art, including, but not limited to, Northern blot hybridization, in situ hybridization, and Southern hybridization to reverse transcriptase - polymerase chain reaction product DNAs.
  • the probes provided by the present invention including oligonucleotide probes derived therefrom, are also useful for Southern hybridization of mammalian, preferably human, genomic DNA for screening for restriction fragment length polymorphism (RFLP) associated with certain genetic disorders.
  • RFLP restriction fragment length polymorphism
  • the term complementary means a nucleic acid having a sequence that is sufficiently complementary in the Watson-Crick sense to a target nucleic acid to bind to the target under physiological conditions or experimental conditions which those skilled in the art routinely use when employing probes.
  • a protein having substantially the same affinity profile as the GalR2 receptor means a protein in which the IC 50 of each of the peptides listed in Table 1 , infra, is no more than an order of magnitude greater than those listed in Table 1 for each of the respective peptides as measured according to the methods described in
  • DNA which encodes receptor GalR2 may be obtained, in view of the instant disclosure, by chemical synthesis, by screening reverse transcripts of mRNA from appropriate cells or cell line cultures, by screening genomic libraries from appropriate cells, or by combinations of these procedures, as illustrated below. Screening of mRNA or genomic DNA may be carried out with oligonucleotide probes generated from the GalR2 gene sequence information provided herein.
  • Probes may be labeled with a detectable group such as a fluorescent group, a radioactive atom or a chemiluminescent group in accordance with known procedures and used in conventional hybridization assays, as described in greater detail in the Examples below.
  • the GalR2 gene sequence may be obtained by use of the polymerase chain reaction (PCR) procedure, with the PCR oligonucleotide primers being produced from the GalR2 gene sequence provided herein. See U.S. Patent Nos.
  • Receptor GalR2 may be synthesized in host cells transformed with a recombinant expression construct comprising a nucleic acid encoding the receptor GalR2.
  • a recombinant expression construct can also be comprised of a vector that is a replicable DNA construct.
  • Vectors are used herein either to amplify DNA encoding GalR2 and/or to express DNA which encodes GalR2.
  • a recombinant expression construct is a replicable DNA construct in which a DNA sequence encoding GaIR2 is operably linked to suitable control sequences capable of effecting the expression of GalR2 in a suitable host. The need for such control sequences will vary depending upon the host selected and the transformation method chosen.
  • control sequences include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation.
  • Amplification vectors do not require expression control domains. All that is needed is the ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants. See, Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Edition, Cold Spring Harbor Press, New York, 1989).
  • Vectors useful for practicing the present invention include plasmids, viruses
  • the vector replicates and functions independently of the host genome, or may, in some instances, integrate into the genome itself.
  • Suitable vectors will contain replicon and control sequences which are derived from species compatible with the intended expression host.
  • the vectors may be self-replicating.
  • Suitable vectors for the purposes of the present invention include pBluescript, pcDNA3, pSV-SPORT, and, for insect cells, baculovirus.
  • a preferred vector is the plasmid pcDNA3 (Invitrogen, San Diego, CA).
  • Isolated plasmids, DNA sequences, or synthesized oligonucleotides are cleaved, tailored, and relegated in the form desired.
  • Site-specific DNA cleavage is performed by treating with the suitable restriction enzyme (or enzymes) under conditions that are generally understood in the art, and the particulars of which are specified by the manufacturer of these commercially available restriction enzymes. See, e.g., New England Biolabs, Product Catalog.
  • suitable restriction enzyme or enzymes
  • about 1 ⁇ g of plasmid or DNA sequence is cleaved by one unit of enzyme in about 20 ⁇ l of buffer solution. Often excess of restriction enzyme is used to ensure complete digestion of the DNA substrate. Incubation times of about one hour to two hours at about 37°C are workable, although variations are tolerable. After each incubation, protein is removed by extraction with phenol/chloroform, and may be followed by ether extraction.
  • the nucleic acid may be recovered from aqueous fractions by precipitation with ethanol. If desired, size separation of the cleaved fragments may be performed by polyacrylamide gel or agarose gel electrophoresis using standard techniques. A general description of size separations is found in Methods in Enzymology 65, 499-560 (1980).
  • Transformed host cells are cells which have been transformed or transfected with recombinant expression constructs made using recombinant DNA techniques and comprising mammalian GalR2-encoding sequences.
  • Preferred host cells for transient transfection are
  • Transformed host cells may ordinarily express GalR2, but host cells transformed for purposes of cloning or amplifying nucleic acid hybridization probe DNA need not express the receptor.
  • the mammalian GalR2 protein When expressed, the mammalian GalR2 protein will typically be located in the host cell membrane. See, Sambrook et al., ibid. Cultures of cells derived from multicellular organisms are desirable hosts for recombinant GalR2 protein synthesis. In principal, any higher eukaryotic cell culture is workable, whether from vertebrate or invertebrate culture. However, mammalian cells are preferred, as illustrated in the Examples. Propagation of such cells in cell culture has become a routine procedure. See Tissue Culture (Academic Press, Kruse & Patterson, Eds., 1973). Examples of useful host cell lines are bacteria cells, insect cells, yeast cells, human 293 cells,
  • VERO and HeLa cells LM ⁇ C cells, and WI 138, BHK, CHO, COS-7, CV, and MDCK cell lines (American Type Culture Collection, Rockville, MD). CHO cells are preferred.
  • the invention provides homogeneous compositions of mammalian GalR2 produced by transformed eukaryotic cells as provided herein. Such homogeneous compositions are intended to be comprised of mammalian GalR2 protein that comprises at least 90% of the protein in such homogenous composition.
  • the invention also provides membrane preparation from cells expressing GalR2 as the result of transformation with a recombinant expression construct, as described here.
  • Mammalian GalR2 protein made from cloned genes in accordance with the present invention may be used for screening compounds for GalR2 agonist or antagonist activity, or for determining the amount of a GalR2 agonist or antagonist drug in a solution (e.g., blood plasma or serum).
  • host cells may be transformed with a recombinant expression construct of the present invention, GalR2 protein expressed in those host cells, the cells lysed, and the membranes from those cells used to screen compounds for GaIR2 binding activity.
  • Competitive binding assays in which such procedures may be carried out are well known in the art. By selection of host cells which do not ordinarily express GalR2, pure or crude preparations of membranes containing GalR2 can be obtained.
  • GaIR2 agonists and antagonists can be identified by transforming host cells with a recombinant expression construct as provided by the present invention. Membranes obtained from such cells (and membranes of intact cells) can be used in binding studies wherein the drug dissociation activity is monitored.
  • the neurotransmitter galanin is a regulator of appetite, cognition, endocrine function, pain, and smooth muscle control.
  • the various galanin analogs/fragments that induce these physiological responses bind with a high affinity to the GalR2 receptor. It is therefore evident that by modulating the activity of the GalR2 receptor, various physiological activities can be regulated.
  • antagonists to the GalR2 receptor identified by the methods described herein, could be used to treat obesity, diabetes, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, and/or endocrine disorders.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a drug identified by the method described herein and a pharmaceutically acceptable carrier.
  • Such drugs and carrier can be administered by various routes, for example oral, subcutaneous, intramuscular, intravenous or intracerebral.
  • the preferred route of administration would be oral at daily doses of about 0.01-100 mg/kg.
  • This invention provides a method of treating obesity, diabetes, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, or endocrine disorders wherein the abnormality is improved by reducing the activity of GalR2 receptor or blocking the binding of ligands to a GalR2 receptor which comprises administering an effective amount of the pharmaceutical composition described above.
  • the recombinant expression constructs of the present invention are useful in molecular biology to transform cells which do not ordinarily express GalR2 to thereafter express this receptor. Such cells are useful as intermediates for making cell membrane preparations useful for receptor binding assays, which are in turn useful for drug screening. Drugs identified from such receptor assays can be used for the treatment of obesity, diabetes, anorexia, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, or endocrine disorders.
  • the recombinant expression constructs of the present invention are also useful in gene therapy.
  • Cloned genes of the present invention, or fragments thereof, may also be used in gene therapy carried out by homologous recombination or site-directed mutagenesis. See generally Thomas & Capecchi, Cell 51, 503-512 (1987); Bertling, Bioscience Reports 7, 107- 112 (1987); Smithies et al., Nature 317, 230-234 (1985).
  • Oligonucleotides of the present invention are useful as diagnostic tools for probing GalR2 gene expression in tissues.
  • tissues are probed in situ with oligonucleotide probes carrying detectable groups by conventional autoradiographic techniques, as explained in greater detail in the Examples below, to investigate native expression of this receptor or pathological conditions relating thereto.
  • chromosomes can be probed to investigate the presence or absence of the GalR2 gene, and potential pathological conditions related thereto, as also illustrated by the Examples below.
  • Probes according to the invention should generally be at least about 15 nucleotides in length to prevent binding to random sequences, but, under the appropriate circumstances may be smaller.
  • the invention also provides antibodies that are immunologically reactive to a mammalian GalR2, preferably rat or human GalR2.
  • the antibodies provided by the invention are raised in animals by inoculation with cells that express a mammalian GalR2 or epitopes thereof, using methods well known in the art.
  • Animals that are used for such inoculations include individuals from species comprising cows, sheep, pigs, mice, rats, rabbits, hamsters, goats and primates.
  • Preferred animals for inoculation are rodents (including mice, rats, hamsters) and rabbits. The most preferred animal is the mouse.
  • Cells that can be used for such inoculations, or for any of the other means used in the invention include any cell line which naturally expresses a mammalian GalR2, or any cell or cell line that expresses a mammalian GalR2 or any epitope thereof as a result of molecular or genetic engineering, or that has been treated to increase the expression of a mammalian
  • GaIR2 by physical, biochemical or genetic means.
  • Preferred cells are human cells, most preferably HEK 293 cells that have been transformed with a recombinant expression construct comprising a nucleic acid encoding a mammalian GalR2, preferably a rat or human GalR2, and that express the mammalian GalR2 gene product.
  • the present invention provides monoclonal antibodies that are immunologically reactive with an epitope of mammalian GalR2 or fragment thereof and that is present on the surface of mammalian cells, preferably human or mouse cells. These antibodies are made using methods and techniques well known to those of skill in the art.
  • Monoclonal antibodies provided by the present invention are produced by hybridoma cell lines, that are also provided by the invention and that are made by methods well known in the art.
  • Hybridoma cell lines are made by fusing individual cells of a myeloma cell line with spleen cells derived from animals immunized with cells expressing the GalR2 receptor, preferably rat or human cells, as described above.
  • the myeloma cell lines used in the invention include lines derived from myelomas of mice, rats, hamsters, primates and humans.
  • Preferred myeloma cell lines are from mouse.
  • the animals from whom spleens are obtained after immunization are rats, mice and hamsters, preferably mice, most preferably Balb/c mice.
  • Spleen cells and myeloma cells are fused using a number of methods well known in the art, including but not limited to incubation with inactivated Sendai virus and incubation in the presence of polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the most preferred method for cell fusion is incubation in the presence of a solution of 45% (w/v) PEG- 1450.
  • Monoclonal antibodies produced by hybridoma cell lines can be harvested from cell culture supernatant fluids from in vitro cell growth; alternatively, hybridoma cells can be injected subcutaneously and/or into the peritoneal cavity of an animal, most preferably a mouse, and the monoclonal antibodies obtained from blood and/or ascites fluid.
  • Monoclonal antibodies provided by the present invention are also produced by recombinant genetic methods well known to those of skill in the art, and the present invention encompasses antibodies made by such methods that are immunologically reactive with an epitope of a mammalian GalR2.
  • the present invention encompasses fragments of the antibody that are immunologically reactive with an epitope of a mammalian GalR2. Such fragments are produced by any number of methods, including but not limited to proteolytic cleavage, chemical synthesis or preparation of such fragments by means of genetic engineering technology.
  • the present invention also encompasses single-chain antibodies that are immunologically reactive with an epitope of a mammalian GalR2 made by methods known to those of skill in the art.
  • the present invention also encompasses an epitope of a mammalian GalR2 that is comprised of sequences and/or a conformation of sequences present in the mammalian GalR2 molecule.
  • This epitope may be naturally occurring, or may be the result of proteolytic cleavage of the mammalian GalR2 molecule and isolation of an epitope-containing peptide or may be obtained by synthesis of an epitope-containing peptide using methods well known to those skilled in the art.
  • the present invention also encompasses epitope peptides produced as a result of genetic engineering technology and synthesized by genetically engineered prokaryotic or eukaryotic cells.
  • the invention also includes chimeric antibodies, comprised of light chain and heavy chain peptides immunologically reactive to an epitope that is a mammalian GaIR2.
  • the chimeric antibodies embodied in the present invention include those that are derived from 13 naturally occurring antibodies as well as chimeric antibodies made by means of genetic engineering technology well known to those of skill in the art.
  • non-human transgenic animals grown from germ cells transformed with the GalR2 nucleic acid sequence according to the invention and that express the GalR2 receptor according to the invention and offspring and descendants thereof.
  • transgenic non-human mammals comprising a homologous recombination knockout of the native GalR2 receptor, as well as transgenic non-human mammals grown from germ cells transformed with nucleic acid antisense to the GaIR2 nucleic acid of the invention and offspring and descendants thereof.
  • transgenic animals which the native GalR2 receptor has been replaced with the human homolog.
  • offspring and descendants of all of the foregoing transgenic animals are also encompassed by the invention.
  • Transgenic animals according to the invention can be made using well known techniques with the nucleic acids disclosed herein.
  • transgenic animals are useful for screening for and determining the physiological effects of GalR2 receptor agonists and antagonist. Consequently, such transgenic animals are useful for developing drugs to regulate physiological activities in which galanin participates.
  • oligo(dT) anchor primer was used for reverse transcription, and the library was cloned unidirectionally into pcDNA3 vector which contains a CMV (cytomegalovirus) promoter for eukaryotic expression.
  • the cDNA library had 5.3 x 10 s primary recombinants with an average insert size of 2.59 kb.
  • the probe was prepared by digesting the parent GalRl plasmid with Sad and AccI, separating the fragments by agarose gel electrophoresis, and purifying the 1-kb SacI-AccI fragment from the gel.
  • the probe was labeled with digoxigenin dUTP according to the manufacturer's instructions (GENIUS Kit, Boehringer Mannheim, Indianapolis, IN, PN 1093
  • the filter lifts, denaturation, neutralization, hybridization, and washing were done according to the manufacturer's instructions except that hybridization was done at 30°C and the washes were performed twice for 40 minutes each: once at room temperature; the second 15 at 37°C.
  • Plasmid DNA homologous to the probe was purified and subcloned into pBluescript vector (Stratagene (La Jolla, California) 212206) by standard molecular biological techniques.
  • This clone designated SI2112, was subjected to sequence analysis and was found to contain a sequence which had characteristics of a novel, but truncated member of the G-protein-coupled 7TMD receptor family.
  • This clone was later used as probe to determine the identity of a novel galanin-binding clone (Y107) found in the expression cloning strategy (see below.)
  • the partial human GalR2 cDNA was obtained by screening approximately two million phage of a human small intestine library (Clontech (Palo Alto, CA) HL 1133a) were screened with a human GalR2 probe obtained by PCR amplification from human genomic DNA. The conditions used were essentially as described for the rat GalR2 isolation.
  • the [ l25 IJgalanin binding assay was performed in the fiaskette chamber.
  • the cells were washed once with 25 mM Tris-HCl, 10 mM MgCl 2 , pH 7.4, and blocked for 15 minutes with 1 ml total binding buffer (25 mM Tris-HCl, 10 mM MgCl 2 , 1% bovine serum albumin, pH 7.4) at room temperature.
  • the darkbox was brought to room temperature for 1 hour after which the slides were developed in D- 19 developer (Kodak (Rochester, NY) 1464593) for three minutes at 15°C, placed in fixer (Kodak (Rochester, NY) 197 1746) for three minutes at 15°C, washed in water, and air dried.
  • Cells were stained with Diff-Quik stain set (Baxter (McGaw Park, IL) B4132-1) and air dried. Slides were dipped into xylenes and mounted with DPX mountant (Electron Microscopy Sciences (Fort Washington, PA) 13510). Positive cells were identified using dark field microscopy.
  • SI2112 probe and, 3) DNA from pool Y107, when used to transiently transfect COS-7 cells, conferred the ability upon the cells to bind galanin, it was deduced that the clone within pool Y107, which conferred galanin-binding ability when expressed, was likely to be an expressible version of the SI2112 clone. Therefore, clones from pool Y107 were probed with radiolabeled DNA prepared from SI2112, and a single clone hybridizing to the SI2112 probe was purified from non-homologous clones. This clone was called Y107.
  • the first intronless version of the Y107 was in the pSV-SPORT vector (GIBCO (Gaithersburg, MD) 15386-014); the complete cDNA insert of this clone was subcloned into the pcDNA3 vector (Invitrogen (San Diego, CA) V790-20) in order to maintain, for subsequent pharamacological analyses, common vector and promoter (CMV) backgrounds amongst our clones.
  • the intronless clone in pSV-SPORT was designated BMB77.sv40; the intronless clone contained within pcDNA3 is named BMB77.
  • Clones BMB77 and Y107 differ by one amino acid in sequence. Pharmacological analyses have been performed with both the Y107 (SEQ ID NO: 3 and 4) and BMB77 (SEQ ID NO: 1 and 2) clones.
  • Plasmid DNA was sequenced by Lark Technologies Inc. (Houston, Texas) and Biotechnology Resource Laboratory of Yale University (New Haven, CT) using Sequenase Kit (US Biochemical (Cleveland, OH) 70770) or Applied Biosystems' automatic sequencer system (Model 373 A). The peptide sequence was deduced from the long open-reading-frame of the nucleotide sequence. DNA and peptide sequences were analyzed using the GCG program (Genetics Computer Group, Madison, WI).
  • SEQ ID NO: 1 the nucleic acid sequence of clone BMB77
  • SEQ ID NO: 2 the amino acid sequence of clone BMB77
  • SEQ ID NO: 3 the nucleic acid sequence of clone Y107
  • SEQ ID NO: 4 the amino acid sequence, omitting the putative intronic region, of clone Y107
  • SEQ ID NO: 5 the partial nucleic acid sequence of human GalR2
  • SEQ ID NO: 6 the partial amino acid sequence of human GaIR2
  • Monkey kidney cells were maintained in T-l 75 cm 2 flasks (Nunc, Inc. (Naperville, IL) 171226) at 37°C with 5% CO 2 in a humidified atmosphere.
  • Cells were grown in Dulbecco's Modified Eagle Medium (DMEM) (GIBCO (Gaithersburg, MD) 11965- 092) supplemented with 2 mM glutamine, 10% fetal bovine serum, 1 mM sodium pyruvate and a antibiotic/antimycotic comprised of pennicillin streptomycin amphotericinB (GIBCO, Gaithersburg, MD, PN 15240-013).
  • DMEM Dulbecco's Modified Eagle Medium
  • GEBCO Gibco's Modified Eagle Medium
  • a antibiotic/antimycotic comprised of pennicillin streptomycin amphotericinB
  • Cells at 70% confluency were transfected with rat GalR2 DNA using the Lipofectamine reagent (GIBCO (G
  • DNA and 90 ⁇ l of lipofectamine were added to each flask. Media was replaced 24 hours post transfection, and membranes were harvested 24 hours later.
  • Rat GalRl Receptor (clone BMB77) 293 cells (human embryonic kidney, ATCC) were plated onto a T-25 flask one day prior to transfection, such that they were 50-70% confluent at the time of transfection.
  • 15 ug of rat GalR2 (BMB77) DNA were added to 0.3 ml of Optimem I culture media (GIBCO Life Sciences), and 25 ul of lipofectamine were added to 0.3 ml of Optimem I.
  • the DNA and lipofectamine solutions were combined and incubated at room temperature for 20 minutes. An additional 2.4 ml of Optimem I was added to the DNA/lipofectamine mixture.
  • the media was aspirated from T-25 flask containing 293 cells, washed with Optimem I, and the DNA/lipofectamine mixture was added to the 293 cells. After a 5-6 hour incubation in a 37°C incubator (5% CO 2 ) for 5-6 hours, the DNA/lipofectamine mixture was replaced with culture media (DMEM, 10% fetal bovine serum, 2 mM glutamine and antibiotic/antimycotic.)
  • culture media DMEM, 10% fetal bovine serum, 2 mM glutamine and antibiotic/antimycotic.
  • the media was removed from each flask of transfected cells, and the cells were washed twice with 20 ml ice-cold phosphate buffered saline.
  • the cells were scraped from the flask in 5 ml of Tris buffer (20 mM Tris-HCl, 5 mM EDTA, pH 7.7), and then transferred to a centrifuge tube. Each flask was rinsed with an additional 5 ml of Tris buffer, and combined in the centrifuge tube.
  • the cells were homogenized in a Polytron PT-3000 (Brinkman Instruments, Mill Valley, New York) for 2 x 10 seconds (12 mM probe, 7000-8000 rpm) and 19 centrifuged at 20,000 x g for 30 minutes at 4°C. The pellet was resuspended in fresh Tris buffer, and centrifuged again at 20,000 x g for 30 minutes at 4°C. Protein concentration was measured using the Bio-Rad kit according to the standard manufacturer's protocol (Bio-Rad Laboratories (Hercules, CA) 500-0001) using bovine IgG as the standard.
  • the binding assays were performed on 96-well plates (GF/C Millipore Corp., Bedford, MA PN MAFC NOB 50) pretreated with 0.3% polyethylenimine (PEI) for at least 3 hours prior to use.
  • PEI polyethylenimine
  • the PEI was aspirated from the plates on a vacuum manifold, and the wells were rinsed with 200 ⁇ l of ice-cold binding buffer (25 mM Tris, 10 mM MgCl 2 , 0.1%
  • Table 1 summarizes the IC J0 values (50% inhibition of specific binding, as determined using nonlinear regression analysis) of various standard peptides, fragments and chimeras for 20
  • Transiently expressed rat GalRl is included in Table 1 for comparison of its pharmacological profile to this novel rat GalR2 receptor.
  • the preliminary pharmacological binding profile for rat GalR2 differs from rat GalRl such that rat galanin itself has about 10-fold lower affinity for GalR2, no matter how it is expressed.
  • the chimeric peptide C7 also has about 10-fold lower affinity for GalR2; this difference, however, is not observed with rat GalR2 stably expressed in 293 cells.
  • Rat(l-16)galanin has about 5- to 10-fold lower affinity for GalR2 than GalRl (Table 1).
  • the ratio of affinities of (1-12) and (l-15)galanin are markedly different for GalRl (22-fold difference), while these two peptides have more equivalent binding affinities for GalR2.
  • Table 1 summarizes the IC 30 values for various standard peptides for [ l25 I]hGalanin binding to rat GalRl and GalR2 clones.
  • the averages ⁇ standard error of the mean (SEM) represent values from at least three independent experiments. Two independent experiments are represented by the average, followed by the individual values in parentheses. Remaining values without SEM are from a single experiment.
  • SEQ ID NO: 1 Clone BMB77 nucleic acid sequence
  • SEQ ID NO: 2 Clone BMB77 amino acid sequence
  • SEQ ID NO: 3 Clone Y107 nucleic acid sequence
  • SEQ ID NO: 4 Clone Y107 amino acid sequence (omitting putative intron)
  • SEQ ID NO: 5 Human GaIR2 partial nucleic acid sequence
  • SEQ ID NO: 6 Human GalR2 partial amino acid sequence

Landscapes

  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention provides a novel galanin receptor protein, the GalR2 receptor. Also provided are the nucleic acid sequences encoding this novel receptor protein as well as methods for using this protein and its nucleic acid sequence, and methods useful for developing and identifying compounds for the treatment of diseases and disorders in which galanin is implicated. The importance of this discovery is manifested in the effects of galanin, which include antinociceptive activity, smooth muscle contraction, cardiovascular activity, pituitary hormone release, cognition, and increased food intake. Thus, this receptor protein is useful for screening for galanin agonist and antagonist activity for controlling these conditions.

Description

GALANIN RECEPTOR GalR2
This application claims priority from U.S. Provisional Application No. 60/019946, filed June 5, 1996.
BACKGROUND OF THE INVENTION 1. Field of the Invention
This invention relates to a novel neuropeptide galanin receptor and its nucleic acid sequence. 2. Description of the Related Art
Galanin is a 29 amino acid peptide hormone (30 amino acids in human) which is present in a wide range of central and peripheral tissues. Skofitsch, Peptides 6, 509 (1985); Merchenthaler, Prog. Neurobiol. 40, 711 (1993). Galanin is involved in many diverse physiological functions. Galanin is known to regulate the secretion of both endocrine and exocrine hormones. Galanin inhibits insulin secretion from pancreatic beta cells, and can inhibit pancreatic amylase secretion; in the stomach, galanin inhibits gastrin and somatostatin secretion. Galanin stimulates VIP (vasoactive intestinal protein) release from the hypothalamus, prolactin and growth hormone release from the pituitary; and inhibits the secretion of ACTH in the hypothalamus. Furthermore, the secretion of neurotransmitters can be modulated. For example, galanin can inhibit the release of histamine and norepinephrine in the hypothalamus. Other secondary messenger systems are also regulated: galanin can either stimulate or inhibit intracellular cAMP accumulation; is involved in the closure of N- and L-type voltage-sensitive calcium channels, and in the opening of ATP-sensitive and - insensitive potassium channels; and has been shown to stimulate the release of calcium from intracellular stores. Moreover, galanin is involved in the inhibition of acetylcholine release and the inhibition of muscarinic receptor-mediated phosphoinositide turnover. Bartfai, Crit. Rev. Neurobiol. 7, 229 (1993)
Galanin is implicated in the modulation of many cognitive and sensory functions. Galanin has potent antinociceptive effects, and can impair performance in one-trial learning, t-maze, and swim maze learning and memory paradigms. Its inhibition of the anoxic release of glutamate, as well as its inhibitory actions on cholinergic function suggest a role in neuroprotection, and in the development of Alzheimer's Disease. Crawley, Regulatory Peptides 59, 1 (1995). Galanin is known to induce feeding in rodents and, in contrast with the effects of Neuropeptide Y on feeding, galanin increases preference for fat intake. Akabayashi, Proc. Natl. Acad. Set. USA 91, 10375 (1994). Galanin is also involved in the regulation of gastrointestinal smooth muscle contraction. Because of the important role of galanin in these many physiological processes, there is a strong need to further develop materials and methods for investigating the mechanistic behavior of the receptors and for treating diseased and other abnormal states associated with these physiological processes.
Pharmacological data suggest the existence of several galanin receptor subtypes. Wynick, Proc. Natl. Acad Sci. USA 90, 4231 (1990); Zen-Fa, J. Pharmacol. Exp. Ther. 272, 371 (1995). Galanin receptors are known to be linked to the Gj proteins, and there is some evidence that certain galanin receptor subtypes may be linked to cholera toxin-sensitive Gs proteins. Gillison, Diabetes 43, 24 (1994); Chen, Am. J. Physiol. 266, Gl 13 (1994). One galanin receptor has been cloned and it is a member of the seven transmembrane (7TMD) class of G protein-linked receptors. It has been designated as GalRl. Habert-Ortoli, Proc. Natl. Acad. Sci. USA 91, 9780; WO95/22608. In addition to this human GalRl receptor, the GalRl receptor has been obtained from rat. Burgevin, J. Mol. Neurosci. 6, 33 (1995). The in vivo functions mediated through this cloned GalRl receptor have not yet been elucidated. EP-0711830-A2 disclose a closely-related GalRl sequence, differing in that Cysl5→Trp is varied.
SUMMARY OF THE INVENTION
The present invention provides a novel galanin receptor protein, the GalR2 receptor. Also provided are the nucleic acid sequences encoding this novel receptor protein as well as methods for using this protein and its nucleic acid sequence, and methods useful for developing and identifying compounds for the treatment of diseases and disorders in which galanin is implicated. The importance of this discovery is manifested in the effects of galanin, which include antinociceptive activity, smooth muscle contraction, cardiovascular activity, pituitary hormone release, cognition, and increased food intake. Thus, this receptor protein is useful for screening for galanin agonist and antagonist activity for controlling these conditions. In one aspect of the present invention, we provide isolated nucleic acid sequences for a novel galanin receptor, the GalR2 receptor. In particular, we provide the cDNA sequences encoding the complete rat receptor and partial sequences of the human receptor. These nucleic acid sequences have a variety of uses. For example, they are useful for making vectors and for transforming cells, both of which are ultimately useful for production of the GalR2 receptor protein. They are also useful as scientific research tools for developing nucleic acid probes for determining receptor expression levels, e.g., to identify diseased or otherwise abnormal states. They are useful for developing analytical tools such as antisense oligonucleotides for selectively inhibiting expression of the receptor gene to determine physiological responses. The present invention can also be used to isolate the homologous nucleic acid sequence of other species, such as human, primate, dog, mouse, etc.
In another aspect of the present invention, we provide a homogeneous composition comprising the receptor GalR2 protein. The protein is useful for screening drugs for agonist and antagonist activity, and, therefore, for screening for drugs useful in regulating physiological responses associated with the GalR2 receptor. Specifically, antagonists to the GalR2 receptor could be used to treat obesity and diabetes by reducing appetite and food consumption, whereas agonists could be used for the treatment of anorexic conditions. Furthermore, drugs could be used to treat Alzheimer's disease, stroke, neuropathic pain, and/or endocrine disorders. The proteins are also useful for developing antibodies for detection of the protein.
Flowing from the foregoing are a number of other aspects of the invention, including (a) vectors, such as plasmids, comprising the receptor GalR2 nucleic acid sequence that may further comprise additional regulatory elements, e.g., promotors, (b) transformed cells that express the GalR2 receptor, (c) nucleic acid probes, (d) antisense oligonucleotides, (e) agonists, (f) antagonists, and (g) transgenic mammals. Further aspects of the invention comprise methods for making and using the foregoing compounds and compositions.
The invention includes polynucleotide molecules coding for a rat or human GalR2, or a galanin binding fragment thereof. A polynucleotide molecule comprising SEQ ID NO.l or a degenerate variant thereof. A polynucleotide molecule comprising the full-length cDNA, or a degenerate variant thereof, corresponding to the partial sequence shown in SEQ ID NO:5. A purified and isolated rat or human GalR2 protein. The GalR2 protein comprising SEQ ID NO:2. The full-length GalR2 protein, the partial sequence of which is indicated in SEQ ID NO:6. A purified and isolated rat or human GalR2 protein or fragment thereof having galanin binding activity. A polynucleotide molecule coding for a variant of rat GalR2 comprising
SEQ ID NO:3, or a galanin binding fragment thereof. A polynucleotide molecule comprising SEQ ID NO: 3 or a degenerate variant thereof. A purified and isolated variant of a rat or human GalR2 protein or fragment thereof having galanin binding activity. A purified and isolated variant of rat GalR2 protein comprising SEQ ID NO:4.
The foregoing merely summarize certain aspects of the present invention and is not intended, nor should it be construed, to limit the invention in any manner. All patents and other publications recited herein are hereby incorporated by reference in their entirety.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is the polynucleotide sequence of rat GalR2 of the invention.
Figure 2 is the amino acid sequence of rat GalR2.
Figures 3-4 are the polynucleotide sequence of the Y107 variant of rat GalR2. Figure 5 is the amino acid sequence of Yl 07(omitting putative intron).
Figure 6 is the partial polynucleotide sequence of human GalR2.
Figure 7 is the partial amino acid sequence of human GalR2.
Figure 8 is the representative saturation isotherm of [125I]hGalanin binding to rat GalR2
(clone BMB77) transiently expressed in COS-7 cells. The inset shows the corresponding linear Rosenthal plot. B/F axis on the Rosenthal plot indicates the ratio of Bound to Free radioligand.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
The present invention comprises, in part, a novel galanin receptor protein, the GalR2 receptor. Particularly preferred embodiments of the GalR2 receptor are those having an amino acid sequence substantially the same as SEQ ID NO: 2, 4 or 6. As used herein, reference to the GalR2 receptor is meant as a reference to any protein having an amino acid sequence substantially the same as SEQ ID NO: 2, 4 or 6. The present invention also comprises the nucleic acid sequence encoding the GalR2 protein, which nucleic acid sequence is substantially the same as SEQ ID NO: 1, 3 or 5. Receptors SEQ ID NO: 1 and SEQ ID NO: 3 are nucleic acid sequences of rat GalR2 receptors but SEQ ID NO: 3 appears to contain an intronic region; therefore, SEQ ID NO: 1 is the preferred embodiment of the rat GalR2 receptor of this invention. Receptor SEQ ID NO: 5 is the partial nucleic acid sequence of human GalR2. Receptors SEQ ID NO: 2 and 4 are rat GalR2 receptors and appear to be allelic variants. Receptor SEQ ID NO: 6 is the partial amino acid sequence of human GalR2.
As used herein, a protein "having an amino acid sequence substantially the same as SEQ ID NO: x" (where "x" is the number of one of the protein sequences recited herein) means a protein whose amino acid sequence is the same as SEQ ID NO: x or differs only in a way such that IC50[galanin] as determined according to the method detailed in Example 2, infra, are less than or equal to 1 nM. Those skilled in the art will appreciate that conservative substitutions of amino acids can be made without significantly diminishing the protein's affinity for galanin and fragments and analogs thereof. Other substitutions may be made that increase the protein's affinity for these compounds. Making and identifying such proteins is a routine matter given the teachings herein, and can be accomplished, for example, by altering the nucleic acid sequence encoding the protein (as disclosed herein), inserting it into a vector, transforming a cell, expressing the nucleic acid sequence, and measuring the binding affinity of the resulting protein, all as taught herein. As used herein the term "a molecule having a nucleotide sequence substantially the same as SEQ ID NO: y" (wherein "y" is the number of one of the protein-encoding nucleotide sequences listed in the Sequence Listing) means a nucleic acid encoding a protein "having an amino acid sequence substantially the same as SEQ ID NO: y*" (wherein "y*" is the number of the amino acid sequence for which nucleotide sequence "y" codes) as defined above. This definition is intended to encompass natural allelic variations in the GalR2 sequence. Cloned nucleic acid provided by the present invention may encode GalR2 protein of any species of origin, including (but not limited to), for example, mouse, rat, rabbbit, cat, dog, primate, and human. Preferably the nucleic acid provided by the invention encodes GaIR2 receptors of mammalian, and most preferably, rat or human origin. The invention also includes nucleotide sequences encoding chimeric proteins comprised of parts of the GalR2 receptor and parts of other related seven-transmembrane receptors.
The BMB77 clone (SEQ ID NO: 1) (see Example 1, infra) has a 1.7-kb cDNA insert with a open reading frame from nucleotides 279 to 1394 that encodes a 372 amino acid protein (SEQ ID NO: 2). Hydrophobicity plot analysis using the PEPPLOT function of GCG (Genetics Computer Group, Madison, WI) shows that the GalR2 receptor has seven transmembrane-like domains, indicating it might be a G-protein-coupIed receptor. GalR2 is 26 amino acids longer in length than GalRl, the only other published galanin receptor. This extra length of amino acids within GalR2 is due to an extended C-terminal tail sequence. However, the putative N-terminal extracellular domain of GalR2 is about 7 amino acids shorter than the corresponding region in GalRl. It is also important to note that the GaIR2 sequence shows only 38% amino acid sequence identity to the GalRl receptor.
The Y107 clone (SEQ ID NO: 3) {see Example 1, infra) has a 1.9-kb cDNA insert with an open reading frame from nucleotides 17-384, and from nucleotides 874-1621. The sequence between nucleotides 384 and 874 contains multiple STOP codons in all three reading frames. Furthermore, the dinucleotides GT and AG at positions 385-386 and 872- 873, respectively, fulfill the criteria for being a splice donor and acceptor site, respectively. Moreover, when the nucleotide sequences 17-384 and 874-1621 are joined together, an open reading frame is formed which has a cognate translation product nearly identical to that of clone BMB77 (SEQ ID NO: 4). Therefore, it is likely that the region between these two open reading frames is an intron.
There is a single nucleotide change between positions 963 in clone BMB77 (SEQ ID NO: 1) and 1 190 in clone Y107 (SEQ ID NO: 3). This adenine to cytosine transversion results in a change from Ser229 in clone BMB77 (SEQ ID NO: 2) to Arg229 in clone Y107 (SEQ ID NO: 4). Amino acid 229 is located in the third intracellular loop of GalR2. The third intracellular loop of other seven transmembrane domain G protein-coupled receptors is an important domain for effecting coupling of the receptor to its G protein. Bockaert, Curr. Op. Neurobiol. 1, 32-42 (1991). This polymorphic amino acid position could change the G protein binding characterisitics of the GalR2 receptor variants. Gillison, Diabetes 43, 24 (1994); Chen, Λm. J. Physiol. 266, Gl 13 (1994).
The partial human GalR2 nucleic acid sequence (SEQ ID NO: 5) contains a 337 amino acid opening reading frame. The initial leucine residue of this partial human GalR2 protein (SEQ ID NO: 6) corresponds to amino acid Leu51 in rat GalR2 (SEQ ID NO: 2 and 4).
Nucleic acid hybridization probes provided by the invention are DNAs consisting essentially of the nucleotide sequences complementary to any sequence depicted in SEQ ID NO:s 1 and 3 that is effective in nucleic acid hybridization. Nucleic acid probes are useful for detecting GalR2 gene expression in cells and tissues using techniques well-known in the art, including, but not limited to, Northern blot hybridization, in situ hybridization, and Southern hybridization to reverse transcriptase - polymerase chain reaction product DNAs. The probes provided by the present invention, including oligonucleotide probes derived therefrom, are also useful for Southern hybridization of mammalian, preferably human, genomic DNA for screening for restriction fragment length polymorphism (RFLP) associated with certain genetic disorders. As used herein, the term complementary means a nucleic acid having a sequence that is sufficiently complementary in the Watson-Crick sense to a target nucleic acid to bind to the target under physiological conditions or experimental conditions which those skilled in the art routinely use when employing probes. Receptor GalR2 binds various fragments and analogs of galanin with affinities different from that of the known receptors. The rank order of binding affinity of receptor GalR2 was found to be: galanin = (2-29)galanin > (l-15)galanin » (3-29)galanin
Table 1, infra, presents a more detailed affinity profile of the GalR2 receptor for galanin and various fragments thereof. As used herein, a protein having substantially the same affinity profile as the GalR2 receptor means a protein in which the IC50 of each of the peptides listed in Table 1 , infra, is no more than an order of magnitude greater than those listed in Table 1 for each of the respective peptides as measured according to the methods described in
Example 2.
The production of proteins such as receptor GalR2 from cloned genes by genetic engineering means is well known in this art. The discussion which follows is accordingly intended as an overview of this field, and is not intended to reflect the full state of the art. DNA which encodes receptor GalR2 may be obtained, in view of the instant disclosure, by chemical synthesis, by screening reverse transcripts of mRNA from appropriate cells or cell line cultures, by screening genomic libraries from appropriate cells, or by combinations of these procedures, as illustrated below. Screening of mRNA or genomic DNA may be carried out with oligonucleotide probes generated from the GalR2 gene sequence information provided herein. Probes may be labeled with a detectable group such as a fluorescent group, a radioactive atom or a chemiluminescent group in accordance with known procedures and used in conventional hybridization assays, as described in greater detail in the Examples below. In the alternative, the GalR2 gene sequence may be obtained by use of the polymerase chain reaction (PCR) procedure, with the PCR oligonucleotide primers being produced from the GalR2 gene sequence provided herein. See U.S. Patent Nos.
4,683,195 to Mullis et al. and 4,683,202 to Mullis.
Receptor GalR2 may be synthesized in host cells transformed with a recombinant expression construct comprising a nucleic acid encoding the receptor GalR2. Such a recombinant expression construct can also be comprised of a vector that is a replicable DNA construct. Vectors are used herein either to amplify DNA encoding GalR2 and/or to express DNA which encodes GalR2. For the purposes of this invention, a recombinant expression construct is a replicable DNA construct in which a DNA sequence encoding GaIR2 is operably linked to suitable control sequences capable of effecting the expression of GalR2 in a suitable host. The need for such control sequences will vary depending upon the host selected and the transformation method chosen. Generally, control sequences include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation. Amplification vectors do not require expression control domains. All that is needed is the ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants. See, Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Edition, Cold Spring Harbor Press, New York, 1989). Vectors useful for practicing the present invention include plasmids, viruses
(including phage), retroviruses, and integratable DNA fragments ( . e. , fragments integratable into the host genome by homologous recombination). The vector replicates and functions independently of the host genome, or may, in some instances, integrate into the genome itself. Suitable vectors will contain replicon and control sequences which are derived from species compatible with the intended expression host. The vectors may be self-replicating. Suitable vectors for the purposes of the present invention include pBluescript, pcDNA3, pSV-SPORT, and, for insect cells, baculovirus. A preferred vector is the plasmid pcDNA3 (Invitrogen, San Diego, CA).
Construction of suitable vectors containing the desired coding and control sequences employs standard ligation and restriction techniques that are well understood in the art.
Isolated plasmids, DNA sequences, or synthesized oligonucleotides are cleaved, tailored, and relegated in the form desired.
Site-specific DNA cleavage is performed by treating with the suitable restriction enzyme (or enzymes) under conditions that are generally understood in the art, and the particulars of which are specified by the manufacturer of these commercially available restriction enzymes. See, e.g., New England Biolabs, Product Catalog. In general, about 1 μg of plasmid or DNA sequence is cleaved by one unit of enzyme in about 20 μl of buffer solution. Often excess of restriction enzyme is used to ensure complete digestion of the DNA substrate. Incubation times of about one hour to two hours at about 37°C are workable, although variations are tolerable. After each incubation, protein is removed by extraction with phenol/chloroform, and may be followed by ether extraction. The nucleic acid may be recovered from aqueous fractions by precipitation with ethanol. If desired, size separation of the cleaved fragments may be performed by polyacrylamide gel or agarose gel electrophoresis using standard techniques. A general description of size separations is found in Methods in Enzymology 65, 499-560 (1980).
Transformed host cells are cells which have been transformed or transfected with recombinant expression constructs made using recombinant DNA techniques and comprising mammalian GalR2-encoding sequences. Preferred host cells for transient transfection are
COS-7 cells. Transformed host cells may ordinarily express GalR2, but host cells transformed for purposes of cloning or amplifying nucleic acid hybridization probe DNA need not express the receptor. When expressed, the mammalian GalR2 protein will typically be located in the host cell membrane. See, Sambrook et al., ibid. Cultures of cells derived from multicellular organisms are desirable hosts for recombinant GalR2 protein synthesis. In principal, any higher eukaryotic cell culture is workable, whether from vertebrate or invertebrate culture. However, mammalian cells are preferred, as illustrated in the Examples. Propagation of such cells in cell culture has become a routine procedure. See Tissue Culture (Academic Press, Kruse & Patterson, Eds., 1973). Examples of useful host cell lines are bacteria cells, insect cells, yeast cells, human 293 cells,
VERO and HeLa cells, LMΗC cells, and WI 138, BHK, CHO, COS-7, CV, and MDCK cell lines (American Type Culture Collection, Rockville, MD). CHO cells are preferred.
The invention provides homogeneous compositions of mammalian GalR2 produced by transformed eukaryotic cells as provided herein. Such homogeneous compositions are intended to be comprised of mammalian GalR2 protein that comprises at least 90% of the protein in such homogenous composition. The invention also provides membrane preparation from cells expressing GalR2 as the result of transformation with a recombinant expression construct, as described here.
Mammalian GalR2 protein made from cloned genes in accordance with the present invention may be used for screening compounds for GalR2 agonist or antagonist activity, or for determining the amount of a GalR2 agonist or antagonist drug in a solution (e.g., blood plasma or serum). For example, host cells may be transformed with a recombinant expression construct of the present invention, GalR2 protein expressed in those host cells, the cells lysed, and the membranes from those cells used to screen compounds for GaIR2 binding activity. Competitive binding assays in which such procedures may be carried out are well known in the art. By selection of host cells which do not ordinarily express GalR2, pure or crude preparations of membranes containing GalR2 can be obtained. Further, GaIR2 agonists and antagonists can be identified by transforming host cells with a recombinant expression construct as provided by the present invention. Membranes obtained from such cells (and membranes of intact cells) can be used in binding studies wherein the drug dissociation activity is monitored.
It is known that the neurotransmitter galanin is a regulator of appetite, cognition, endocrine function, pain, and smooth muscle control. As shown herein, the various galanin analogs/fragments that induce these physiological responses bind with a high affinity to the GalR2 receptor. It is therefore evident that by modulating the activity of the GalR2 receptor, various physiological activities can be regulated. Specifically, antagonists to the GalR2 receptor, identified by the methods described herein, could be used to treat obesity, diabetes, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, and/or endocrine disorders.
This invention provides a pharmaceutical composition comprising an effective amount of a drug identified by the method described herein and a pharmaceutically acceptable carrier. Such drugs and carrier can be administered by various routes, for example oral, subcutaneous, intramuscular, intravenous or intracerebral. The preferred route of administration would be oral at daily doses of about 0.01-100 mg/kg.
This invention provides a method of treating obesity, diabetes, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, or endocrine disorders wherein the abnormality is improved by reducing the activity of GalR2 receptor or blocking the binding of ligands to a GalR2 receptor which comprises administering an effective amount of the pharmaceutical composition described above.
The recombinant expression constructs of the present invention are useful in molecular biology to transform cells which do not ordinarily express GalR2 to thereafter express this receptor. Such cells are useful as intermediates for making cell membrane preparations useful for receptor binding assays, which are in turn useful for drug screening. Drugs identified from such receptor assays can be used for the treatment of obesity, diabetes, anorexia, hyperlipidemia, stroke, neuropathic pain, Alzheimer's disease, or endocrine disorders.
The recombinant expression constructs of the present invention are also useful in gene therapy. Cloned genes of the present invention, or fragments thereof, may also be used in gene therapy carried out by homologous recombination or site-directed mutagenesis. See generally Thomas & Capecchi, Cell 51, 503-512 (1987); Bertling, Bioscience Reports 7, 107- 112 (1987); Smithies et al., Nature 317, 230-234 (1985).
. Oligonucleotides of the present invention are useful as diagnostic tools for probing GalR2 gene expression in tissues. For example, tissues are probed in situ with oligonucleotide probes carrying detectable groups by conventional autoradiographic techniques, as explained in greater detail in the Examples below, to investigate native expression of this receptor or pathological conditions relating thereto. Further, chromosomes can be probed to investigate the presence or absence of the GalR2 gene, and potential pathological conditions related thereto, as also illustrated by the Examples below. Probes according to the invention should generally be at least about 15 nucleotides in length to prevent binding to random sequences, but, under the appropriate circumstances may be smaller. The invention also provides antibodies that are immunologically reactive to a mammalian GalR2, preferably rat or human GalR2. The antibodies provided by the invention are raised in animals by inoculation with cells that express a mammalian GalR2 or epitopes thereof, using methods well known in the art. Animals that are used for such inoculations include individuals from species comprising cows, sheep, pigs, mice, rats, rabbits, hamsters, goats and primates. Preferred animals for inoculation are rodents (including mice, rats, hamsters) and rabbits. The most preferred animal is the mouse.
Cells that can be used for such inoculations, or for any of the other means used in the invention, include any cell line which naturally expresses a mammalian GalR2, or any cell or cell line that expresses a mammalian GalR2 or any epitope thereof as a result of molecular or genetic engineering, or that has been treated to increase the expression of a mammalian
GaIR2 by physical, biochemical or genetic means. Preferred cells are human cells, most preferably HEK 293 cells that have been transformed with a recombinant expression construct comprising a nucleic acid encoding a mammalian GalR2, preferably a rat or human GalR2, and that express the mammalian GalR2 gene product. The present invention provides monoclonal antibodies that are immunologically reactive with an epitope of mammalian GalR2 or fragment thereof and that is present on the surface of mammalian cells, preferably human or mouse cells. These antibodies are made using methods and techniques well known to those of skill in the art.
Monoclonal antibodies provided by the present invention are produced by hybridoma cell lines, that are also provided by the invention and that are made by methods well known in the art. Hybridoma cell lines are made by fusing individual cells of a myeloma cell line with spleen cells derived from animals immunized with cells expressing the GalR2 receptor, preferably rat or human cells, as described above. The myeloma cell lines used in the invention include lines derived from myelomas of mice, rats, hamsters, primates and humans. Preferred myeloma cell lines are from mouse. The animals from whom spleens are obtained after immunization are rats, mice and hamsters, preferably mice, most preferably Balb/c mice. Spleen cells and myeloma cells are fused using a number of methods well known in the art, including but not limited to incubation with inactivated Sendai virus and incubation in the presence of polyethylene glycol (PEG). The most preferred method for cell fusion is incubation in the presence of a solution of 45% (w/v) PEG- 1450. Monoclonal antibodies produced by hybridoma cell lines can be harvested from cell culture supernatant fluids from in vitro cell growth; alternatively, hybridoma cells can be injected subcutaneously and/or into the peritoneal cavity of an animal, most preferably a mouse, and the monoclonal antibodies obtained from blood and/or ascites fluid.
Monoclonal antibodies provided by the present invention are also produced by recombinant genetic methods well known to those of skill in the art, and the present invention encompasses antibodies made by such methods that are immunologically reactive with an epitope of a mammalian GalR2.
The present invention encompasses fragments of the antibody that are immunologically reactive with an epitope of a mammalian GalR2. Such fragments are produced by any number of methods, including but not limited to proteolytic cleavage, chemical synthesis or preparation of such fragments by means of genetic engineering technology. The present invention also encompasses single-chain antibodies that are immunologically reactive with an epitope of a mammalian GalR2 made by methods known to those of skill in the art.
The present invention also encompasses an epitope of a mammalian GalR2 that is comprised of sequences and/or a conformation of sequences present in the mammalian GalR2 molecule. This epitope may be naturally occurring, or may be the result of proteolytic cleavage of the mammalian GalR2 molecule and isolation of an epitope-containing peptide or may be obtained by synthesis of an epitope-containing peptide using methods well known to those skilled in the art. The present invention also encompasses epitope peptides produced as a result of genetic engineering technology and synthesized by genetically engineered prokaryotic or eukaryotic cells.
The invention also includes chimeric antibodies, comprised of light chain and heavy chain peptides immunologically reactive to an epitope that is a mammalian GaIR2. The chimeric antibodies embodied in the present invention include those that are derived from 13 naturally occurring antibodies as well as chimeric antibodies made by means of genetic engineering technology well known to those of skill in the art.
Also provided by the present invention are non-human transgenic animals grown from germ cells transformed with the GalR2 nucleic acid sequence according to the invention and that express the GalR2 receptor according to the invention and offspring and descendants thereof. Also provided are transgenic non-human mammals comprising a homologous recombination knockout of the native GalR2 receptor, as well as transgenic non-human mammals grown from germ cells transformed with nucleic acid antisense to the GaIR2 nucleic acid of the invention and offspring and descendants thereof. Further included as part of the present invention are transgenic animals which the native GalR2 receptor has been replaced with the human homolog. Of course, offspring and descendants of all of the foregoing transgenic animals are also encompassed by the invention.
Transgenic animals according to the invention can be made using well known techniques with the nucleic acids disclosed herein. E.g., Leder et al., U.S. Patent Nos. 4,736,866 and 5, 175,383; Hogan et al., Manipulating the Mouse Embryo, A Laboratory
Manual (Cold Spring Harbor Laboratory (1986)); Capecchi, Science 244, 1288 (1989); and Zimmer and Gruss, Nature 338, 150 (1989). Such transgenic animals are useful for screening for and determining the physiological effects of GalR2 receptor agonists and antagonist. Consequently, such transgenic animals are useful for developing drugs to regulate physiological activities in which galanin participates.
The following Examples are provided for illustrative purposes only and are not intended, nor should they be construed, as limiting the invention in any manner.
EXAMPLES Example 1: Isolation and Sequencing of Rat GalR2 Receptor
Isolation of rat hypothalamus mRNA and construction ofcDNA library
An expression cloning strategy was used to clone the novel galanin receptor from a rat hypothalamus cDNA library. RNA was obtained from 9 frozen rat hypothalami weighing a total of 0.87 grams. Poly(A) RNA was isolated directly from the tissue using the Promega PolyATtract System 1000 kit (Promega (Madison, WI) Z5420). The hypothalami were homogenized in 4 mL of 4M guanidine thiocyanate-25mM sodium citrate, pH 7.1-2% β- mercaptoethanol using a Polytron at full-speed for approximately 1 minute. To the homogenized tissue 8 mL of 4M guanidine thiocyanate-25mM sodium citrate, pH 7.1-1% β- mercaptoethanol which had been preheated to 70°C was added. After mixing thoroughly, 870 pmol biotinylated oligo(dT) was added; the mixture was incubated at 70 °C for 5 minutes. The homogenate was subjected to centrifugation at 12000 x g for 10 minutes at room temperature; the homogenate was transferred to a clean tube and 10.44 mL Streptavidin MAGNESPHERE* Paramagnetic Particles (SA-PMPs) which had been prepared as per the published protocol was added. (Promega Corporation (Madison, WI) published protocol TM 228). The homogenate and SA-PMPs were incubated together for 2 minutes at room temperature after which the homogenate was decanted while the SA-PMP-biotinylated oligo(dT)-hypothalamic poly(A) RNA complex was retained in the tube by a magnetic stand. The complex was washed as per the protocol, after which the RNA was precipitated and resuspended in water. 25 micrograms of this poly(A) RNA was used by Invitrogen (Invitrogen Corporation, San Diego, CA) to prepare a cDNA expression library. The protocols used by Invitrogen to prepare the cDNA library are essentially based upon the procedures of Okayama and Berg (Molec. Cell. Biol. 2, 161 (1982)) and Gubler and Hoffman (Gene 25, 263 ( 1983)) (Invitrogen Corporation (San Diego, CA) publications 130813sa and
130928sa). An oligo(dT) anchor primer was used for reverse transcription, and the library was cloned unidirectionally into pcDNA3 vector which contains a CMV (cytomegalovirus) promoter for eukaryotic expression. The cDNA library had 5.3 x 10s primary recombinants with an average insert size of 2.59 kb.
Isolation of a novel galanin receptor cDNA clone 1. Homology cloning strategy
In order to isolate novel receptor(s) for galanin, approximately 2 million phage plaques of a rat small intestine library (Stratagene (La Jolla, CA) 936508) were screened with rat GalRl coding sequence DNA as probe under low stringency conditions (30% formamide,
6X SSC (0.9M NaCl/0.09M NaCitrate), 0.1% N-lauroyl sarcosine, 0.2% __ JS, 3% blocking reagent.) The probe was prepared by digesting the parent GalRl plasmid with Sad and AccI, separating the fragments by agarose gel electrophoresis, and purifying the 1-kb SacI-AccI fragment from the gel. The probe was labeled with digoxigenin dUTP according to the manufacturer's instructions (GENIUS Kit, Boehringer Mannheim, Indianapolis, IN, PN 1093
657). The filter lifts, denaturation, neutralization, hybridization, and washing were done according to the manufacturer's instructions except that hybridization was done at 30°C and the washes were performed twice for 40 minutes each: once at room temperature; the second 15 at 37°C.
One plaque containing DNA homologous to the probe was purified and subcloned into pBluescript vector (Stratagene (La Jolla, California) 212206) by standard molecular biological techniques. This clone, designated SI2112, was subjected to sequence analysis and was found to contain a sequence which had characteristics of a novel, but truncated member of the G-protein-coupled 7TMD receptor family. This clone was later used as probe to determine the identity of a novel galanin-binding clone (Y107) found in the expression cloning strategy (see below.)
The partial human GalR2 cDNA was obtained by screening approximately two million phage of a human small intestine library (Clontech (Palo Alto, CA) HL 1133a) were screened with a human GalR2 probe obtained by PCR amplification from human genomic DNA. The conditions used were essentially as described for the rat GalR2 isolation.
2. Expression cloning strategy The rat hypothalamus cDNA library was plated on the Luria Broth/ Ampicillin
(GIBCO) plates in pools of 1,000 independent colonies. The plates were incubated at 37°C for about 20 hours and the bacteria from each plate were scraped in 4-5 ml LB/Ampicillin media. Two ml of the bacteria samples were used for plasmid preparation and one ml of each pool was stored at -80°C in 15% glycerol. COS-7 cells were grown in Dulbecco's Modified Eagle Medium (DMEM, GIBCO
(Gaithersburg, MD) 11965-092), 10% fetal bovine serum (GIBCO (Gaithersburg, MD) 16000-028), and IX antibiotic/antimycotic solution (GIBCO (Gaithersburg, MD) 15240- 039). Cells were maintained by trypsinizing and splitting at 50 to 70% confluency.
Twenty-four hours before transfection, cells were plated into fiaskette chambers (Nunc, Inc. (Naperville, IL) 177453) at 3x 105 cells flaskette (equivalent to 3x 104 cells/cm2).
Two μg of plasmid DNA from each pool was transfected into the cells using 10 μl of Lipofectamine (GIBCO (Gaithersburg, MD) 18324-012) according to the manufacturer's protocol.
Forty-eight hours after transfection, the [l25IJgalanin binding assay was performed in the fiaskette chamber. The cells were washed once with 25 mM Tris-HCl, 10 mM MgCl2, pH 7.4, and blocked for 15 minutes with 1 ml total binding buffer (25 mM Tris-HCl, 10 mM MgCl2, 1% bovine serum albumin, pH 7.4) at room temperature. After aspirating off the blocking solution, 1 ml of binding buffer containing 100 pM , 5I-hGalanin (NEN DuPont _
16
(Boston, MA) NEX-333) was added and flasks were incubated at room temperature for 90 minutes. Following the incubation, the labeling buffer was removed and the flaskettes were rinsed (approximately 2 mL per fiaskette) four times with ice-cold binding buffer. After a final rinse with ice-cold phosphate buffered saline (PBS)(GIBCO (Gaithersburg, MD) 14190- 136), the cells were fixed with ice-cold PBS/1 % glutaraldehyde (Sigma (St. Louis, MO)
G5882). The solution was removed and residual glutaraldehyde rinsed away with one wash of ice-cold PBS/0.5 M Tris (pH 7.5) followed by one wash of ice-cold PBS. After separating the slide bases from their tops, the slides were dipped in 0.5% gelatin at 42 °C and dried under vacuum. The dried slides were dipped in photographic emulsion (NTB-2) (Kodak (Rochester, NY) 165 4433) diluted 1:1 in 0.02% Aerosol-OT (Sigma (St. Louis, MO) A6627) at 42 °C, dried at room temperature for 1 hour, and exposed in the darkbox for four or five days at 4°C. After sufficient exposure time, the darkbox was brought to room temperature for 1 hour after which the slides were developed in D- 19 developer (Kodak (Rochester, NY) 1464593) for three minutes at 15°C, placed in fixer (Kodak (Rochester, NY) 197 1746) for three minutes at 15°C, washed in water, and air dried. Cells were stained with Diff-Quik stain set (Baxter (McGaw Park, IL) B4132-1) and air dried. Slides were dipped into xylenes and mounted with DPX mountant (Electron Microscopy Sciences (Fort Washington, PA) 13510). Positive cells were identified using dark field microscopy.
Two positive pools were identified. Since the hypothalamus could express different subtypes of galanin receptor, we analyzed the positive pools for GalRl receptors by PCR and homology. Of the 2 positive pools tested as described above, 1 contained GalRl as determined by PCR and homology analyses. However, the other pool (Y107) was negative for GalRl by PCR, but homologous to SI2112 probe (see Homology strategy, above). Because: 1) SI2112 sequence indicated it was likely a novel, albeit truncated and unexpressible, G-protein-coupled receptor; 2) pool Y107 DNA showed homology to the
SI2112 probe; and, 3) DNA from pool Y107, when used to transiently transfect COS-7 cells, conferred the ability upon the cells to bind galanin, it was deduced that the clone within pool Y107, which conferred galanin-binding ability when expressed, was likely to be an expressible version of the SI2112 clone. Therefore, clones from pool Y107 were probed with radiolabeled DNA prepared from SI2112, and a single clone hybridizing to the SI2112 probe was purified from non-homologous clones. This clone was called Y107.
Sequence analysis of clone Y107 (SEQ ID NO: 3) revealed the presence of one intron of 489-bp length, beginning after nucleotide 384 (the second nucleotide of the codon for 17 amino acid 133, an arginine residue). Using Y107 DNA as probe, an intronless version of the Y107 cDNA was obtained from a PC 12 cell cDNA library by homology cloning. The first intronless version of the Y107 was in the pSV-SPORT vector (GIBCO (Gaithersburg, MD) 15386-014); the complete cDNA insert of this clone was subcloned into the pcDNA3 vector (Invitrogen (San Diego, CA) V790-20) in order to maintain, for subsequent pharamacological analyses, common vector and promoter (CMV) backgrounds amongst our clones. The intronless clone in pSV-SPORT was designated BMB77.sv40; the intronless clone contained within pcDNA3 is named BMB77. Clones BMB77 and Y107 differ by one amino acid in sequence. Pharmacological analyses have been performed with both the Y107 (SEQ ID NO: 3 and 4) and BMB77 (SEQ ID NO: 1 and 2) clones.
DNA and peptide sequences analysis
Plasmid DNA was sequenced by Lark Technologies Inc. (Houston, Texas) and Biotechnology Resource Laboratory of Yale University (New Haven, CT) using Sequenase Kit (US Biochemical (Cleveland, OH) 70770) or Applied Biosystems' automatic sequencer system (Model 373 A). The peptide sequence was deduced from the long open-reading-frame of the nucleotide sequence. DNA and peptide sequences were analyzed using the GCG program (Genetics Computer Group, Madison, WI). The results are embodied in SEQ ID NO: 1 (the nucleic acid sequence of clone BMB77), SEQ ID NO: 2 (the amino acid sequence of clone BMB77), SEQ ID NO: 3 (the nucleic acid sequence of clone Y107), SEQ ID NO: 4 (the amino acid sequence, omitting the putative intronic region, of clone Y107), SEQ ID NO: 5 (the partial nucleic acid sequence of human GalR2), and SEQ ID NO: 6 (the partial amino acid sequence of human GaIR2).
Example 2: Pharmacological Characterization of the Novel Rat Galanin Receptor
Transient Transfection
Monkey kidney cells (COS-7) were maintained in T-l 75 cm2 flasks (Nunc, Inc. (Naperville, IL) 171226) at 37°C with 5% CO2 in a humidified atmosphere. Cells were grown in Dulbecco's Modified Eagle Medium (DMEM) (GIBCO (Gaithersburg, MD) 11965- 092) supplemented with 2 mM glutamine, 10% fetal bovine serum, 1 mM sodium pyruvate and a antibiotic/antimycotic comprised of pennicillin streptomycin amphotericinB (GIBCO, Gaithersburg, MD, PN 15240-013). Cells at 70% confluency were transfected with rat GalR2 DNA using the Lipofectamine reagent (GIBCO (Gaithersburg, MD) 18324-012). 15 μg of 18
DNA and 90 μl of lipofectamine were added to each flask. Media was replaced 24 hours post transfection, and membranes were harvested 24 hours later.
Stable Expression of the Rat GalRl Receptor (clone BMB77) 293 cells (human embryonic kidney, ATCC) were plated onto a T-25 flask one day prior to transfection, such that they were 50-70% confluent at the time of transfection. 15 ug of rat GalR2 (BMB77) DNA were added to 0.3 ml of Optimem I culture media (GIBCO Life Sciences), and 25 ul of lipofectamine were added to 0.3 ml of Optimem I. The DNA and lipofectamine solutions were combined and incubated at room temperature for 20 minutes. An additional 2.4 ml of Optimem I was added to the DNA/lipofectamine mixture. The media was aspirated from T-25 flask containing 293 cells, washed with Optimem I, and the DNA/lipofectamine mixture was added to the 293 cells. After a 5-6 hour incubation in a 37°C incubator (5% CO2) for 5-6 hours, the DNA/lipofectamine mixture was replaced with culture media (DMEM, 10% fetal bovine serum, 2 mM glutamine and antibiotic/antimycotic.)
The day following transfection, the media was replaced with selecting media (culture media with the addition of 350 ug/ml of Geneticin G-418), and the flask was returned to the 37°C / 5% CO2 incubator. When discrete colonies became apparent, cells were pooled. Growth was monitored, followed by cloning by limited dilution, such that an average of one cell was seeded in each well of a 96-well microtiter culture plate. After about 21 days in culture under selection conditions, those wells containing single colonies were selected and transferred to 24-well culture plates pretreated with poly-1-lysine, following trypsinization. Each of these clones was propagated until sufficient quantities were available for testing in the [125 IJgalanin binding assay, from which one particular clone designated 293-rGalR2-l was selected on the basis of its high level of receptor expression.
Membrane Preparation
The media was removed from each flask of transfected cells, and the cells were washed twice with 20 ml ice-cold phosphate buffered saline. The cells were scraped from the flask in 5 ml of Tris buffer (20 mM Tris-HCl, 5 mM EDTA, pH 7.7), and then transferred to a centrifuge tube. Each flask was rinsed with an additional 5 ml of Tris buffer, and combined in the centrifuge tube. The cells were homogenized in a Polytron PT-3000 (Brinkman Instruments, Mill Valley, New York) for 2 x 10 seconds (12 mM probe, 7000-8000 rpm) and 19 centrifuged at 20,000 x g for 30 minutes at 4°C. The pellet was resuspended in fresh Tris buffer, and centrifuged again at 20,000 x g for 30 minutes at 4°C. Protein concentration was measured using the Bio-Rad kit according to the standard manufacturer's protocol (Bio-Rad Laboratories (Hercules, CA) 500-0001) using bovine IgG as the standard.
f'"IIGalanin Binding Assay for rat GalRl and rat GalR2 clones
The binding assays were performed on 96-well plates (GF/C Millipore Corp., Bedford, MA PN MAFC NOB 50) pretreated with 0.3% polyethylenimine (PEI) for at least 3 hours prior to use. The PEI was aspirated from the plates on a vacuum manifold, and the wells were rinsed with 200 μl of ice-cold binding buffer (25 mM Tris, 10 mM MgCl2, 0.1%
BSA, pH 7.4) immediately before samples were added to the wells. For competition assays, increasing concentrations of peptide were incubated with [I25I]hGaianin (NEN DuPont (Boston, MA) NEX333) and membrane. In a final volume of 200 μl, samples consisted of ~1.25 μg of protein, 50 pM [l25I]hGalanin, and peptide dilution or binding buffer. Nonspecific binding was defined by 100 nM rat galanin. For saturation experiments, increasing concentrations of [l25I]hGalanin were incubated with membrane and 100 nM rat galanin. Samples were incubated for 90 minutes at room temperature with constant shaking. To terminate the reaction, samples were aspirated on a vacuum manifold and rinsed with 3 x 200 μl ice-cold buffer. Samples were then counted on a gamma counter to quantitate the amount of radioactivity. Rat galanin, fragment peptides (1-15)galanin, (1-12)galanin, (1-10
)galanin, chimeric peptide M40, (2-29)rat galanin, (3-29)rat galanin, (5-29)rat galanin, (9- 29)rat galanin, (10-29)rat galanin, (2-30)human galanin, and (3-30)human galanin were synthesized at Bayer Corp. (West Haven, CT). All other peptides were purchased from either Peninsula (Belmont, CA) or Bachem (Torrance, CA).
In Vitro Pharmacnlo v
Saturation analysis for rat GalR2 transiently expressed in COS-7 cells yielded a one- component model with an average K,, value of 0.28 nM (n=2) and a receptor density (BmiX) of 770 fmol/mg protein (Fig. 6). This is very similar to the rat GalR2 receptor stably expressed in 293 cells, which yielded an average K,, value of 0.20 nM (n=2) and a receptor density
(BΠMX) of 2289 fmol/mg protein (data not shown).
Table 1 summarizes the ICJ0 values (50% inhibition of specific binding, as determined using nonlinear regression analysis) of various standard peptides, fragments and chimeras for 20
[12 I]hGalanin binding to membranes expressing the rGalR2 receptor. Transiently expressed rat GalRl is included in Table 1 for comparison of its pharmacological profile to this novel rat GalR2 receptor. The preliminary pharmacological binding profile for rat GalR2 differs from rat GalRl such that rat galanin itself has about 10-fold lower affinity for GalR2, no matter how it is expressed. When comparing transiently expressed GalRl and GaIR2, the chimeric peptide C7 also has about 10-fold lower affinity for GalR2; this difference, however, is not observed with rat GalR2 stably expressed in 293 cells. Rat(l-16)galanin has about 5- to 10-fold lower affinity for GalR2 than GalRl (Table 1). In addition, the ratio of affinities of (1-12) and (l-15)galanin are markedly different for GalRl (22-fold difference), while these two peptides have more equivalent binding affinities for GalR2.
The binding profiles for intron-containing (Y107) and intronless (BMB77 clone) GalR2 receptors transiently expressed are very similar, with the possible exceptions of M15 and (l-12)galanin, which have approximately 5- and 3 -fold higher affinities, respectively, for clone Y107.
21
Table 1
Table 1 summarizes the IC30 values for various standard peptides for [l25I]hGalanin binding to rat GalRl and GalR2 clones. The averages ± standard error of the mean (SEM) represent values from at least three independent experiments. Two independent experiments are represented by the average, followed by the individual values in parentheses. Remaining values without SEM are from a single experiment. Peptide species are indicated with the following prefixes: r = rat, h = human. ND = not determined 22 Abbreviations of Chimers:
Ml 5 = (l-13)Galanin + (5-1 l)Substance P - Galantide (see Bartfai, TIPS 13, 312-317 (1992) M35 = (l-13)Galanin + (2-9)Bradykinin (Bartfai, infra) M40 = (l-13)Galanin + ProPro(AlaLeu)2Ala amide (Bartfai, infra) C7 = ( 1 - 13)Galanin + spantide (see Crawley, Brain Research 600, 268-27? ( 1993)
Having now fully described this invention, it will be appreciated by those skilled in the art that the same can be performed with any wide range of equivalent parameters of composition, conditions, and methods of preparing such recombinant molecules, vectors, transformed hosts and proteins without departing from the spirit or scope of the invention or any embodiment thereof.
23
SEQ ID NO: 1 : Clone BMB77 nucleic acid sequence
1 TCGACCCACG CGTCCGCTCA AGTCTAAAGC AGAGCGAGTC CCAGGACTTG
51 AGCGCGGGAA GCGAATGGAG TCAGGGTCAT TCGATTGCAC CTCTCTCGGC
101 TGCGGGCCGG AGCGGGGTAC CATCCTACAC TCTGGGTGCT CCCTCCTCCT
151 CCCGTCCCCC GCGCACCCCT GCCCTGGCTC CTGGAGCTCG GCAGTCTCGC
201 TGGGGCGCTG CAGCGAGGGA GCAGCGTGCT CACCAAGACC CGGACAGCTG
251 CGGGAGCGGC GTCCACTTTG GTGATACC r fiAATCGCTCC GGCAGCCAGG
301 GCGCGGAGAA CACGAGCCAG GAAGGCGGTA GCGGCGGCTG GCAGCCTGAG
351 GCGGTCCTTG TACCCCTATT TTTCGCGCTC ATCTTCCTCG TGGGCACCGT
401 GGGCAACGCG CTGGTGCTGG CGGTGCTGCT GCGCGGCGGC CAGGCGGTCA
451 GCACCACCAA CCTGTTCATC CTCAACCTGG GCGTGGCCGA CCTGTGTTTC
501 ATCCTGTGCT GCGTGCCTTT CCAGGCCACC ATCTACACCC TGGACGACTG 551 GGTGTTCGGC TCGCTGCTCT GCAAGGCTGT TCATTTCCTC ATCTTTCTCA
601 CTATGCACGC CAGCAGCTTC ACGCTGGCCG CCGTCTCCCT GGACAGGTAT
651 CTGGCCATCC GCTACCCGCT GCACTCCCGA GAGTTGCGCA CACCTCGAAA
701 CGCGCTGGCC GCCATCGGGC TCATCTGGGG GCTAGCACTG CTCTTCTCCG
751 GGCCCTACCT GAGCTACTAC CGTCAGTCGC AGCTGGCCAA CCTGACAGTA 801 TGCCACCCAG CATGGAGCGC ACCTCGACGT CGAGCCATGG ACCTCTGCAC
851 CTTCGTCTTT AGCTACCTGC TGCCAGTGCT AGTCCTCAGT CTGACCTATG
901 CGCGTACCCT GCGCTACCTC TGGCGCACAG TCGACCCGGT GACTGCAGGC
951 TCAGGTTCCC AG^GCGCCΛA ACGCAAGGTG ACACGGATGA TCATCATCGT
1001 GGCGGTGCTT TTCTGCCTCT GTTGGATGCC CCACCACGCG CTTATCCTCT 1051 GCGTGTGGTT TGGTCGCTTC CCGCTCACGC GTGCCACTTA CGCGTTGCGC
1101 ATCCTTTCAC ACCTAGTTTC CTATGCCAAC TCCTGTGTCA ACCCCATCGT
1151 TTACGCTCTG GTCTCCAAGC ATTTCCGTAA AGGTTTCCGC AAAATCTGCG
1201 CGGGCCTGCT GCGCCCTGCC CCGAGGCGAG CTTCGGGCCG AGTGAGCATC 24
1251 CTGGCGCCTG GGAACCATAG TGGCAGCATG CTGGAACAGG AATCCACAGA
1301 CCTGACACAG GTGAGCGAGG CAGCCGGGCC CCTTGTCCCA CCACCCGCAC
1351 TTCCCAACTG CACAGCCTCG AGTAGAACCC TGGATCCGGC TTGTTA4AGG
K01 ACCAAAGGGC ATCTAACAGC TTCTAGACAG TGTGGCCCGA GGATCCCTGG
K51 GGGTTATGCT TGAACGTTAC AGGGTTGAGG CTAAAGACTG AGGATTGATT
1501 GTAGGGAACC TCCAGTTATT AAACGGTGCG GATTGCTAGA GGGTGGCATA
1551 GTCCTTCAAT CCTGGCACCC GAAAAGCAGA TGCAGGAGCA GGAGCAGGAG 1601 CAAAGCCAGC CATGGAGTTT GAGGCCTGCT TGAACTACCT GAGATCCAAT
1651 AATAAAACAT TTCATATGCT GTGAAAAAAA AAAAAAAAAA
25
SEQ ID NO: 2: Clone BMB77 amino acid sequence
1 ATG AAT GGC TCC GGC AGC CAG GGC GCG GAG AAC ACG AGC CAG GAA GGC GGT AGC GGC GGC 1 Met Asn Gly Ser Gly Ser Gin Gly Ala Glu Asn Thr Ser Gin Glu Gly Gly Ser Gly Gly
61 TGG CAG CCT GAG GCG GTC CTT GTA CCC CTA TTT TTC GCG CTC ATC TTC CTC GTG GGC ACC
21 Trp Gin Pro Glu Ala Vβl Leu Val Pro Leu Phe Phe Ala Leu lie Phe Leu Val Gly Thr 121 GTG GGC AAC GCG CTG GTG CTG GCG GTG CTG CTG CGC GGC GGC CAG GCG GTC AGC ACC ACC
41 Val Gly Asn Ala Leu Val Leu Ala Val Leu Leu Arg Gly Gly Gin Ala Val Ser Thr Thr
181 AAC CTG TTC ATC CTC AAC CTG GGC GTG GCC GAC CTG TGT TTC ATC CTG TGC TGC GTG CCT
61 Asn Leu Phe He Leu Asn Leu Gly Val Ala Asp Leu Cys Phe He Leu Cys Cys Val Pro
241 TTC CAG GCC ACC ATC TAC ACC CTG GAC GAC TGG GTG TTC GGC TCG CTG CTC TGC AAG GCT 81 Phe Gin Ala Thr He Tyr Thr Leu Asp Asp Trp Val Phe Gly Ser Leu Leu Cys Lys Ala
301 GTT CAT TTC CTC ATC TTT CTC ACT ATG CAC GCC AGC AGC TTC ACG CTG GCC GCC GTC TCC 101 Val His Phe Leu He Phe Leu Thr Met His Ala Ser Ser Phe Thr Leu Ala Ala Vat Ser
361 CTG GAC AGG TAT CTG GCC ATC CGC TAC CCG CTG CAC TCC CGA GAG TTG CGC ACA CCT CGA
121 Leu Asp Arg Tyr Leu Ala He Arg Tyr Pro Leu His Ser Arg Glu Leu Arg Thr Pro Arg 421 AAC GCG CTG GCC GCC ATC GGG CTC ATC TGG GGG CTA GCA CTG CTC TTC TCC GGG CCC TAC
141 Asn Ala Leu Ala Ala He Gly Leu He Trp Gly Leu Ala Leu Leu Phe Ser Gly Pro Tyr
481 CTG AGC TAC TAC CGT CAG TCG CAG CTG GCC AAC CTG ACA GTA TGC CAC CCA GCA TGG AGC 161 Leu Ser Tyr Tyr Arg Gin Ser Gin Leu Ala Asn Leu Thr Vat Cys His Pro Ala Trp Ser
541 GCA CCT CGA CGT CGA GCC ATG GAC CTC TGC ACC TTC GTC TTT AGC TAC CTG CTG CCA GTG 181 Ala Pro Arg Arg Arg Ala Met Asp Leu Cys Thr Phe Val Phe Ser Tyr Leu Leu Pro Val
601 CTA GTC CTC ACT CTG ACC TAT GCG CGT ACC CTG CGC TAC CTC TGG CGC ACA GTC GAC CCG 201 Leu Val Leu Ser Leu Thr Tyr Ala Arg Thr Leu Arg Tyr Leu Trp Arg Thr Val Asp Pro
661 GTG ACT GCA GGC TCA GGT TCC CAG AGC GCC AAA CGC AAG GTG ACA CGG ATG ATC ATC ATC
221 Val Thr Ala Giy Ser Gly Ser Gin is Ala Lys Arg Lys Val Thr Arg Met He He He
721 GTG GCG GTG CTT TTC TGC CTC TGT TGG ATG CCC CAC CAC GCG CTT ATC CTC TGC GTG TGG
241 Val Ala Val Leu Phe Cys Leu Cys Trp Met Pro His His Ala Leu He Leu Cys Val Trp
781 TTT GGT CGC TTC CCG CTC ACG CGT GCC ACT TAC GCG TTG CGC ATC CTT TCA CAC CTA GTT
261 Phe Gly Arg Phe Pro Leu Thr Arg Ala Thr Tyr Ala Leu Arg He Leu Ser His Leu Val
841 TCC TAT GCC AAC TCC TGT GTC AAC CCC ATC GTT TAC GCT CTG GTC TCC AAG CAT TTC CGT
281 Ser Tyr Ala Asn Ser Cys Val Asn Pro He Vat Tyr Ala Leu Val Ser Lys His Phe Arg
901 AAA GGT TTC CGC AAA ATC TGC GCG GGC CTG CTG CGC CCT GCC CCG AGG CGA GCT TCG CGC 301 Lys Gly Phe Arg Lys He Cys Ala Gly Leu Leu Arg Pro Ala Pro Arg Arg Ala Ser Gly 961 CGA GTG AGC ATC CTG GCG CCT GGG AAC CAT AGT GGC AGC ATG CTG GAA CAG GAA TCC ACA 321 Arg Val Ser He Leu Ala Pro Gly Asn His Ser Gly Ser Met Leu Glu Gin Glu Ser Thr
1021 GAC CTG ACA CAG GTG AGC GAG GCA GCC GGG CCC CTT GTC CCA CCA CCC GCA CTT CCC AAC 341 Asp Leu Thr Gin Val Ser Glu Ala Ala Gly Pro Leu Val Pro Pro Pro Ala Leu Pro Asn
1081 TGC ACA GCC TCG AGT AGA ACC CTG GAT CCG GCT TGT TAA 1119 361 Cys Thr Ala Ser Ser Arg Thr Leu Asp Pro Ala Cys * 372
27
SEQ ID NO: 3: Clone Y107 nucleic acid sequence
1 CCACTTTGGT GATACC 7gA ATGGCTCCGG CAGCCAGGGC GCGGAGAACA
51 CGAGCCAGGA AGGCGGTAGC GGCGGCTGGC AGCCTGAGGC GGTCCTTGTA
101 CCCCTATTTT TCGCGCTCAT CTTCCTCGTG GGCACCGTGG GCAACGCGCT 151 GGTGCTGGCG GTGCTGCTGC GCGGCGGCCA GGCGGTCAGC ACCACCAACC
201 TGTTCATCCT CAACCTGGGC GTGGCCGACC TGTGTTTCAT CCTGTGCTGC
251 GTGCCTTTCC AGGCCACCAT CTACACCCTG GACGACTGGG TGTTCGGCTC
301 GCTGCTCTGC AAGGCTGTTC ATTTCCTCAT CTTTCTCACT ATGCACGCCA
351 GCAGCTTCAC GCTGGCCGCC GTCTCCCTGG ACAGGTAAAG GACCCAGAAA 401 GAAACATCCA GTATGCCCGG AGGGATCTTG ACTGGAAAAG ACTGAATCCT
451 GGTCTGGTGA CCTTAGTTςC CTGCCCTTTC ΛCΛTCΛCTTG GACΛTTCCCA
501 CΛCΛΛGΛG.G GTCΛΛGΛGGG GQT_.TC.TT ATTCTCCTCT GGTTTCCΛCT
551 GΛGTGCΛACΛ TGTGCGTCCT GΛGTΛCGCTQ GnGGGACTCΛ GAΛΛATTTCΛ
601 GCTTTCTTTA GGAGTTTCCT TGCTGTAGTT TGACCCAAGT CTTCTCCAGG 651 TTTCTGTCAG AACTCAGGCA TGAGGGATCT GCCTCCCCTG GTTGTCACCA
701 GΛGGΛTΛΛCΛ ΛTCACTGCCC CCπGΛΛΛTCC ΛGΛCΛCnTTC TπCΛΛCTTTT
751 AGTCTTCGGT GTTTTGGGGG TGCCCCTTCA CGTGGAGTAG GTCGGTGGCC
801 ACATTCCCAG GAGTGACAAT AGCCTAGCAG TGAATCCTCT CGCTTAGCTG
851 ATGCCCCCCC ACTGTCCCCA CAGGTATCTG GCCATCCGCT ACCCGCTGCA 901 CTCCCGAGAG TTGCGCACAC CTCGAAACGC GCTGGCCGCC ATCGGGCTCA
951 TCTGGGGGCT AGCACTGCTC TTCTCCGGGC CCTACCTGAG CTACTACCGT
1001 CAGTCGCAGC TGGCCAACCT GACAGTATGC CACCCAGCAT GGAGCGCACC
1051 TCGACGTCGA GCCATGGACC TCTGCACCTT CGTCTTTAGC TACCTGCTGC
1101 CAGTGCTAGT CCTCAGTCTG ACCTATGCGC GTACCCTGCG CTACCTCTGG 1151 CGCACAGTCG ACCCGGTGAC TGCAGGCTCA GGTTCCCAGj. GCGCCAAACG 28
1201 CAAGGTGACA CGGATGATCA TCATCGT6GC GGTGCTTTTC TGCCTCTGTT
1251 GGATGCCCCA CCACGCGCTT ATCCTCTCCG TGTGGTTTGG TCGCTTCCCG
1301 CTCACGCGTG CCACTTACGC GTTGCGCATC CTTTCACACC TACTTTCCTA
1351 TGCCAACTCC TGTGTCAACC CCATCGTTTA CGCTCTGGTC TCCAAGCATT
1401 TCCGTAAAGG TTTCCGCAAA ATCTGCGCGG GCCTGCTGCG CCCTGCCCCG
1451 AGGCGAGCTT CGGGCCGAGT GAGCATCCTG GCGCCTGGGA ACCATAGTGG
1501 CAGCATCCTG GAACAGGAAT CCACACACCT GACACAGGTG AGCGAGGCAG 1551 CCGGGCCCCT TGTCCCACCA CCCGCACTTC CCAACTGCAC AGCCTCGAGT
1601 AGAACCCTGG ATCCGGCTTG Tf AGGACC AAAGGGCATC TAACAGCTTC
1651 TAGACAGTGT GGCCCGAGGΛ TCCCTGGGGG TTATGCTTGA ACGTTACΛGG
1701 GTTGAGGCTA AAGACTGAGG ATTGATTGTA GGGAACCTCC AGTTATTAAA
1751 CGGTGCGGAT TGCTAGAGGG TGGCATAGTC CTTCAATCCT GGCACCCGAA 1801 AAGCAGATGC AGGAGCAGGA GCAGGAGCAA AGCCAGCCAT GGAGTTTGAG
1851 GCCTGCTTGA ACTACCTGAG ATCCAATAAT AAAACATTTC ATATGCTGTG
1901 AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA AAAAAAAAAA
1951 AAAAAAAA
29
SEQ ID NO: 4: Clone Y107 amino acid sequence (omitting putative intron)
1 ATG AAT GGC TCC GGC AGC CAG GGC GCG GAG AAC ACG AGC CAG GAA GGC GGT AGC GGC GGC 1 Met Asn Gly Ser Gly Ser Gin Gly Ala Glu Asn Thr Ser Gin Glu Gly Gly Ser Gly Gly
61 TGG CAG CCT GAG GCG GTC CTT GTA CCC CTA TTT TTC GCG CTC ATC TTC CTC GTG GGC ACC
21 Trp Gin Pro Glu Ala Val Leu Vβl Pro Leu Phe Phe Ate Leu He Phe Leu Val Gly Thr 121 GTG GGC AAC GCG CTG GTG CTG GCG GTG CTG CTG CGC GGC GGC CAG GCG GTC AGC ACC ACC
41 Vat Gly Asn Ala Leu Val Leu Ala Val Leu Leu Arg Gly Gly Gin Ala Val Ser Thr Thr
181 AAC CTG TTC ATC CTC AAC CTG GGC GTG GCC GAC CTG TGT TTC ATC CTG TGC TGC CTG CCT
61 Asn Leu Phe He Leu Asn Leu Gly Vat Ala Asp Leu Cys Phe He Leu Cys Cys Vat Pro
241 TTC CAG GCC ACC ATC TAC ACC CTG GAC GAC TGG GTG TTC GGC TCG CTG CTC TGC AAC GCT 81 Phe Gin Ala Thr He Tyr Thr Leu Asp Asp Trp Vat Phe Gly Ser Leu Leu Cys Lys Ala
301 GTT CAT TTC CTC ATC TTT CTC ACT ATG CAC GCC AGC AGC TTC ACG CTG GCC GCC GTC TCC 101 Val His Phe Leu He Phe Leu Thr Met His Ala Ser Ser Phe Thr Leu Ala Ala Val Ser
361 CTG GAC AGG TAT CTG GCC ATC CGC TAC CCG CTG CAC TCC CGA GAG TTG CGC ACA CCT CGA 121 Leu Asp Arg Tyr Leu Ala He Arg Tyr Pro Leu His Ser Arg Glu Leu Arg Thr Pro Arg
421 AAC GCG CTG GCC GCC ATC GGG CTC ATC TGG GGG CTA GCA CTG CTC TTC TCC GGG CCC TAC 141 Asn Ala Leu Ala Ala He Gly Leu He Trp Gly Leu Ale Leu Leu Phe Ser Gly Pro Tyr
481 CTG AGC TAC TAC CGT CAG TCG CAG CTG GCC AAC CTG ACA GTA TGC CAC CCA GCA TGG AGC 161 Leu Ser Tyr Tyr Arg Gin Ser Gin Leu Ala Asn Leu Thr Val Cys His Pro Ala Trp Ser
541 GCA CCT CGA CGT CGA GCC ATG GAC CTC TGC ACC TTC GTC TTT AGC TAC CTG CTG CCA GTG 181 Ala Pro Arg Arg Arg Ala Met Asp Leu Cys Thr Phe Val Phe Ser Tyr Leu Leu Pro Vβl
601 CTA GTC CTC AGT CTG ACC TAT GCG CGT ACC CTG CGC TAC CTC TGG CGC ACA GTC GAC CCG 201 Leu Val Leu Ser Leu Thr Tyr Ala Arg Thr Leu Arg Tyr Leu Trp Arg Thr Val Asp Pro
661 GTG ACT GCA GGC TCA GGT TCC CAG CGC GCC AAA CGC AAG GTG ACA CGG ATC ATC ATC ATC
221 Vat Thr Ala Gly Ser Gly Ser Gtn ACS Ala Lys Arg Lys Vat Thr Arg Met He He He 721 GTG GCG GTG CTT TTC TCC CTC TGT TGG ATG CCC CAC CAC GCG CTT ATC CTC TGC GTG TGG
241 Vat Ala Val Leu Phe Cys Leu Cys Trp Met Pro His His Ala Leu He Leu Cys Vβl Trp
781 TTT GGT CGC TTC CCC CTC ACG CGT GCC ACT TAC GCG TTG CGC ATC CTT TCA CAC CTA GTT
261 Phe Gly Arg Phe Pro Leu Thr Arg Ala Thr Tyr Ala Leu Arg He Leu Ser His Leu Val
841 TCC TAT GCC AAC TCC TGT GTC AAC CCC ATC GTT TAC GCT CTC GTC TCC AAG CAT TTC CGT
281 Ser Tyr Ala Asn Ser Cys Val Asn Pro He Val Tyr Ala Leu Val Ser Lys His Phe Arg
901 AAA GGT TTC CGC AAA ATC TGC GCG GGC CTG CTG CGC CCT GCC CCG AGG CGA GCT TCG GGC 301 Lys Gly Phe Arg Lys He Cys Ala Gly Leu Leu Arg Pro Ala Pro Arg Arg Ala Ser Gly O 97/4
30
961 CGA GTG AGC ATC CTG GCG CCT GGG AAC CAT AGT GGC AGC ATG CTG GAA CAG GAA TCC ACA
321 Arg Val Ser He Leu Ala Pro Gly Asn His Ser Gly Ser Met Leu Glu Gin Glu Ser Thr
1021 GAC CTG ACA CAG GTG AGC CAG CCA CCC GGG CCC CTT CTC CCA CCA CCC GCA CTT CCC AAC
341 Asp Leu Thr Gin Val Ser Glu Ala Ala Gly Pro Leu Val Pro Pro Pro Ale Leu Pro Aβn
1081 TGC ACA GCC TCG AGT AGA ACC CTG CAT CCG GCT TGT TAA 1119 361 Cys Thr Ala Ser Ser Arg Thr Leu Asp Pro Ala Cys * 372
31
SEQ ID NO: 5: Human GaIR2 partial nucleic acid sequence
1 CTGCGCGGCG GCCAGGCGGT CAGCACTACC AACCTGCTCA TCCTTAACCT
51 GGGCGTGGCC GACCTGTGTT TCATCCTGTG CTGCGTGCCC TTCCAGGCCA
101 CCATCTACAC CCTGGACGGC TGGGTGTTCG GCTCGCTGCT GTGCAAGGCG 151 GTGCACTTCC TCATCTTCCT CΛCCATGCAC GCCAGCAGCT TCACCCTGGC
201 CGCCGTCTCC CTGGACAGGT ATCTGGCCAT CCCCTACCCG CTGCACTCCC
251 GCGAGCTGCG CACGCCTCGA AACGCGCTGG CAGCCATCGG GCTCATCTGG
301 GGGCTGTCGC TCCTCTTCTC CGGGCCCTAC CTGAGCTACT ACCGCCAGTC
351 GCAGCTGGCC AACCTGACCG TGTGCCΛTCC CGCGTGGAGC GCCCCTCGCC 401 GCCGCGCCAT GGACATCTGC ACCTTCGTCT TCAGCTACCT GCTTCCTGTG
451 CTGGTTCTCG GCCTGACCTA CGCGCGCACC TTGCCCTACC TCTCGCGCGC
501 CGTCGACCCG GTGGCCGCGG GCTCGGGTGC CCGGCGCGCC AAGCGCAAGG
551 TGACACGCAT GATCCTCATC GTGGCCGCGC TCTTCTGCCT CTGCTGGATG
601 CCCCACCACG CGCTCATCCT CTGCGTGTGG TTCGGCCAGT TCCCGCTCAC 651 GCGCGCCACT TATGCGCTTC GCATCCTCTC GCACCTGGTC TCCTACGCCA
701 ACTCCTGCGT CAACCCCATC GTTTACGCGC TGGTCTCCAA GCACTTCCGC
751 AAAGGCTTCC GCACGATCTG CGCGGGCCTG CTGGGCCGTG CCCCAGGCCG
801 AGCCTCGGGC CGTGTGTGCG CTGCCGCGCG GGGCACCCAC AGTGGCAGCG
851 TGTTGGAGCG CGAGTCCAGC GACCTGTTGC ACATGAGCGA GGCGGCGGGG 901 GCCCTTCGTC CCTGCCCCGG CGCTTCCCAG CCATGCATCC TCGAGCCCTG
951 TCCTGGCCCG TCCTGGCAGG GCCCAAAGGC AGGCGACAGC ATCC7G4CGG
1001 TTGATGTGGC CTGAAAGCAC TTAGCCCGCG CGCTGGGATG TCACAGAGTT
1051 GGAGTCATTG TTGCGGGACC GTGGGCCGGA ATT 32
SEQ ID NO: 6: Human GalR2 partial amino acid sequence
1 CTG CGC GGC GGC CAG GCG GTC AGC ACT ACC AAC CTG CTC ATC CTT AAC CTG GGC GTG GCC (51) Leu Arg Gly Gly Gin Ala Val Ser Thr Thr Asn Leu Leu He Leu Asn Leu Gty Vβl Ala
61 GAC CTG TGT TTC ATC CTG TGC TGC GTG CCC TTC CAG GCC ACC ATC TAC ACC CTG GAC GGC Asp Leu Cys Phe He Leu Cys Cys Val Pro Phe Oln Ala Thr He Tyr Thr Leu Asp Cly
121 TGG GTG TTC GGC TCC CTG CTG TGC AAG GCG GTG CAC TTC CTC ATC TTC CTC ACC ATG CAC Trp Val Phe Gty Ser Leu Leu Cys Lys Ala Val His Phe Leu He Phe Leu Thr Met His
181 GCC AGC AGC TTC ACG CTG GCC GCC GTC TCC CTG GAC AGG TAT CTG GCC ATC CGC TAC CCG Ala Ser Ser Phe Thr Leu Ala Ale Vet Ser Leu Asp Arg Tyr Leu Ala He Arg Tyr Pro
241 CTG CAC TCC CGC GAG CTG CGC ACG CCT CGA AAC GCG CTG GCA GCC ATC GGG CTC ATC TGG Leu His Ser Arg Glu Leu Arg Thr Pro Arg Asn Ala Leu Ala Ala He Gly Leu He Trp
301 GGG CTG TCG CTG CTC TTC TCC GGG CCC TAC CTG AGC TAC TAC CGC CAG TCG CAG CTG GCC Gly Leu Ser Leu Leu Phe Ser Cly Pro Tyr Leu Ser Tyr Tyr Arg Gin Ser Gin Leu Ala
361 AAC CTG ACC GTG TGC CAT CCC GCG TGG AGC GCC CCT CGC CGC CGC GCC ATG GAC ATC TGC
Asn Leu Thr Vat Cys His Pro Ale Trp Ser Ala Pro Arg Arg Arg Ala Met Asp He Cys
421 ACC TTC GTC TTC AGC TAC CTG CTT CCT GTG CTG GTT CTC GGC CTG ACC TAC GCG CGC ACC
Thr Phe Val Phe Ser Tyr Leu Leu Pro Val Leu Val Leu Gty Leu Thr Tyr Ala Arg Thr
481 TTG CGC TAC CTC TGG CGC GCC GTC GAC CCG GTG GCC GCG GGC TCG GGT GCC CGG CGC GCC Leu Arg Tyr Leu Trp Arg Ala Val Asp Pro Vat Ala Ala Gly Ser Gly Ale Arg Arg Ala
541 AAG CGC AAG GTG ACA CGC ATG ATC CTC ATC GTG GCC GCG CTC TTC TGC CTC TGC TGG ATG Lys Arg Lys Val Thr Arg Met He Leu He Val Ala Ala Leu Phe Cys Leu Cys Trp Met 601 CCC CAC CAC GCG CTC ATC CTC TGC GTG TGG TTC GGC CAG TTC CCG CTC ACG CGC GCC ACT Pro His His Ala Leu He Leu Cys Val Trp Phe Gly Gtn Phe Pro Leu Thr Arg Ala Thr
661 TAT GCG CTT CGC ATC CTC TCG CAC CTG GTC TCC TAC GCC AAC TCC TGC GTC AAC CCC ATC Tyr Ala Leu Arg He Leu Ser His Leu Val Ser Tyr Ala Asn Ser Cys Val Asn Pro He
721 GTT TAC GCG CTG GTC TCC AAG CAC TTC CGC AAA GGC TTC CGC ACG ATC TGC GCG GGC CTG Vβl Tyr Ala Leu Val Ser Lys His Phe Arg Lys Gty Phe Arg Thr He Cys Ala Gly Leu
781 CTG GGC CGT GCC CCA GGC CGA GCC TCG GGC CGT GTG TGC GCT GCC GCG CGG GGC ACC CAC Leu Gly Arg Ala Pro Gly Arg Ala Ser Gly Arg Vat Cys Ala Ala Ala Arg Gly Thr His
841 AGT GGC AGC GTG TTG GAG CGC GAG TCC AGC GAC CTG TTG CAC ATG AGC GAG GCG GCG GGG
Ser Gly Ser Val Leu Glu Arg Glu Ser Ser Asp Leu Leu His Met Ser Glu Ala Ale Gly 901 GCC CTT CGT CCC TGC CCC GGC GCT TCC CAG CCA TGC ATC CTC GAG CCC TGT CCT GGC CCG
Ale Leu Arg Pro Cys Pro Gly Ala Ser Gin Pro Cys He Leu Glu Pro Cys Pro Gly Pro 961 TCC TGG CAC GGC CCA AAG GCA GGC GAC AGC ATC CTG ACG GTT GAT GTG GCC TCA AACCACT Ser Trp Gin Gly Pro Lys Ala Gly Asp Ser He Leu Thr Vat Asp Val Ala *
1022 TAGCGGGCGCGCTGGGATGTCACAGAGTTGGAGTCATTGTTGGGGGACCGTGGGCCGGAATT

Claims

What is claimed as new and useful is:
1. A polynucleotide molecule coding for GalR2 comprising SEQ ID NO: 1, SEQ ID NO:
3, and SEQ ID NO: 5, or a variant or fragment thereof.
2. A purified and isolated GalR2 protein comprising SEQ ID NO: 2, SEQ ID NO: 4 and
SEQ ID NO: 6 or a variant or fragment thereof.
3. A vector comprising a neucleic acid sequence according to claim 1.
4. A cell transformed or transfected with the vector according to claim 3.
5. A method of identifying a GalR2 receptor agonist or antagonist comprising contacting a membrane prepared from cells according to claim 4 with test material and evaluating affinity of the test material to the GalR2 receptor.
EP97929814A 1996-06-05 1997-06-05 GALANIN RECEPTOR GalR2 Withdrawn EP0907734A2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US665034 1996-06-05
US08/665,034 US6410686B1 (en) 1996-06-05 1996-06-05 Galanin receptor 2 protein
US86803497A 1997-06-03 1997-06-03
PCT/US1997/009787 WO1997046681A2 (en) 1996-06-05 1997-06-05 GALANIN RECEPTOR GalR2
2003-05-23

Publications (1)

Publication Number Publication Date
EP0907734A2 true EP0907734A2 (en) 1999-04-14

Family

ID=27099113

Family Applications (1)

Application Number Title Priority Date Filing Date
EP97929814A Withdrawn EP0907734A2 (en) 1996-06-05 1997-06-05 GALANIN RECEPTOR GalR2

Country Status (2)

Country Link
EP (1) EP0907734A2 (en)
CA (1) CA2256523A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SE9602822D0 (en) 1996-07-19 1996-07-19 Astra Pharma Inc New receptor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9746681A3 *

Also Published As

Publication number Publication date
CA2256523A1 (en) 1997-12-11

Similar Documents

Publication Publication Date Title
US20060189530A1 (en) Neuropeptide Y receptor Y5 and nucleic acid sequences
US5545549A (en) DNA encoding a human neuropeptide Y/peptide YY (Y2) receptor and uses thereof
EP0579758B1 (en) Parathyroid hormone receptor and dna encoding same
AU741041B2 (en) DNA encoding galanin GALR3 receptors and uses thereof
US5886148A (en) Parathyroid hormone receptor
JPH10262687A (en) New human 11cb splice variant
US5919901A (en) Neuropeptide Y receptor Y5 and nucleic acid sequences
US6410686B1 (en) Galanin receptor 2 protein
US20030082672A1 (en) Dna encoding galanin galr2 receptors and uses thereof
EP0730647A1 (en) Dna sequence coding for a bmp receptor
US20030008350A1 (en) 14273 receptor, a novel G-protein coupled receptor
CA2240427C (en) Growth hormone secretagogue receptor family
US7132260B2 (en) DNA encoding parathyroid hormone receptor
US7150974B1 (en) Parathyroid hormone receptor binding method
CA2363794A1 (en) 14273 receptor, a novel g-protein coupled receptor
JP2005229804A (en) New melanin concentrating hormone receptor
EP0907734A2 (en) GALANIN RECEPTOR GalR2
US20090275050A1 (en) 14273 receptor, a novel G-protein coupled receptor
US6531314B1 (en) Growth hormone secretagogue receptor family
AU699824C (en) DNA sequence coding for a BMP receptor
US20030108928A1 (en) MID 241 receptor, a novel G-protein coupled receptor
MXPA00012953A (en) 14273 receptor, a g-protein coupled receptor

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990105

AK Designated contracting states

Kind code of ref document: A2

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

17Q First examination report despatched

Effective date: 20020412

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20031216