EP1129104A1 - Dna moleküle, die das melanocortin 3 rezeptor protein des rhesus affen kodieren - Google Patents

Dna moleküle, die das melanocortin 3 rezeptor protein des rhesus affen kodieren

Info

Publication number
EP1129104A1
EP1129104A1 EP99971812A EP99971812A EP1129104A1 EP 1129104 A1 EP1129104 A1 EP 1129104A1 EP 99971812 A EP99971812 A EP 99971812A EP 99971812 A EP99971812 A EP 99971812A EP 1129104 A1 EP1129104 A1 EP 1129104A1
Authority
EP
European Patent Office
Prior art keywords
cells
substance
protein
rhesus
expression vector
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Withdrawn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
EP99971812A
Other languages
English (en)
French (fr)
Inventor
Tung M. Fong
Leonardus H. T. Van Der Ploeg
Ruey-Ruey C. Huang
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.)
Merck and Co Inc
Original Assignee
Merck and Co Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck and Co Inc filed Critical Merck and Co Inc
Publication of EP1129104A1 publication Critical patent/EP1129104A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor

Definitions

  • the present invention relates to rhesus monkey (Macaca mulatto)
  • DNA molecules encoding the melanocortin-3 receptor protein belonging to the rhodopsin sub-family of G-protein coupled receptors recombinant vectors comprising DNA molecules encoding rhesus MC-3R, recombinant host cells which contain a recombinant vector encoding rhesus MC-3R, the rhesus MC-3R protein encoded by the DNA molecule, and methods of identifying selective agonists and antagonists of rhesus MC-3R.
  • GPCR G- protein coupled receptors
  • melanocortin receptors bind and are activated by peptides such as ⁇ -, ⁇ , or ⁇ - melanocyte stimulating hormones ( ⁇ -, ⁇ -, ⁇ -MSH) derived from the pro- opiomelanocortin (POMC) gene.
  • ⁇ -, ⁇ -, ⁇ -MSH ⁇ - melanocyte stimulating hormones
  • POMC pro- opiomelanocortin
  • Mountjoy discloses human and mouse DNA molecules which encode MC-1R (also known in the art as ⁇ -MSH-R).
  • the expressed human protein contains 317 amino acids.
  • Roselli-Rehfuss, et al, (1993, Proc. Natl. Acad. Sci 90: 8856-8860) disclose a cDNA clone encoding rat MC-3R cDNA.
  • Chhajlani, et al. (1993, Biochem. Biophys. Res. Comm., 195(2): 866- 873) disclose a DNA molecule which the authors state encodes MC-5R. This clone was initially designated MC2. Fathi, et al. (1995, Neurochemical Research 20(1):107-113) also disclose a DNA molecule thought to encode human MC-5R. There are several sequence discrepancies when compared to the DNA molecule disclosed by Chhajlani, et al., id.
  • MC-4R has been implicated as a key regulator of feeding behavior which regulates body weight through studies with peptide agonists and antagonists (Fan et al, 1997, Nature 385: 165-168) and with a MC-4R knock-out mouse (Huszar et al., 1997, Cell 88: 131-141).
  • the present invention addresses and meets these needs by disclosing an isolated nucleic acid fragment which expresses a form of rhesus MC-3R, recombinant vectors which house this nucleic acid fragment, recombinant host cells which expresses rhesus MC-3R and/or a biologically active equivalent, and pharmacological properties of this rhesus MC-3R protein.
  • the present invention relates to an isolated nucleic acid molecule (polynucleotide) which encodes a novel rhesus monkey (Macaca mulatto) melanocortin-3 receptor (rhMC-3R).
  • the nucleic acid molecules of the present invention are substantially free from other nucleic acids.
  • the present invention relates to an isolated nucleic acid molecule (polynucleotide) which encodes mRNA which expresses a novel rhesus MC-3R, this DNA molecule comprising the nucleotide sequence disclosed herein as SEQ ID NO:l.
  • the present invention also relates to biologically active fragments or mutants of SEQ ID NO:l which encodes mRNA expressing a novel rhesus MC-3R. Any such biologically active fragment and/or mutant will encode either a protein or protein fragment which at least substantially mimics the pharmacological properties of a wild-type MC-3R protein, including but not limited to the rhesus MC-3R receptor protein as set forth in SEQ ID NO:2.
  • any such polynucleotide includes but is not necessarily limited to nucleotide substitutions, deletions, additions, amino-terminal truncations and carboxy-terminal truncations such that these mutations encode mRNA which express a protein or protein fragment of diagnostic, therapeutic or prophylactic use and would be useful for screening for agonists and/or antagonists for MC-3R function.
  • the isolated nucleic acid molecules of the present invention may include a deoxyribonucleic acid molecule (DNA), such as genomic DNA and complementary DNA (cDNA), which may be single (coding or noncoding strand) or double stranded, as well as synthetic DNA, such as a synthesized, single stranded polynucleotide.
  • the isolated nucleic acid molecule of the present invention may also include a ribonucleic acid molecule (RNA).
  • the present invention also relates to recombinant vectors and recombinant hosts, both prokaryotic and eukaryotic, which contain the substantially purified nucleic acid molecules disclosed throughout this specification.
  • the present invention also relates to subcellular membrane fractions of the recombinant host cells (both prokaryotic and eukaryotic as well as both stably and transiently transformed cells) which contain the proteins encoded by the nucleic acids of the present invention.
  • These subcellular membrane fractions will comprise either wild-type or mutant forms of rhesus melanocortin-3 receptor proteins at levels substantially above endogenous levels and hence will be useful in various assays described throughout this specification.
  • the present invention also relates to a substantially purified form of the rhesus melanocortin-3 receptor protein, which comprises the amino acid sequence disclosed in Figure 2 and set forth as SEQ ID NO:2.
  • a preferred aspect of the present invention is disclosed in Figure 2 and is set forth as SEQ ID NO:2, the amino acid sequence of the novel rhesus melanocortin-3 receptor protein.
  • the present invention also relates to biologically active fragments and/or mutants of the rhesus melanocortin-3 receptor protein comprising the amino acid sequence set forth as SEQ ID NO:2, including but not necessarily limited to amino acid substitutions, deletions, additions, amino terminal truncations and carboxy-terminal truncations such that these mutations provide for proteins or protein fragments of diagnostic, therapeutic or prophylactic use and would be useful for screening for agonists and/or antagonists for MC-3R function.
  • the present invention also relates to assays to screen or select for various modulators of MC-3R activity, methods of expressing the MC-3R protein and biological equivalents disclosed herein, recombinant host cells which comprise DNA constructs which express these receptor proteins, and compounds identified through these assays which act as agonists or antagonists of MC-3R activity.
  • the present invention also relates to isolated nucleic acid molecules which are fusion constructions expressing fusion proteins useful in assays to identify compounds which modulate wild-type vertebrate MC-3R activity.
  • a preferred aspect of this portion of the invention includes, but is not limited to, glutathione S- transferase (GST)-MC-3R fusion constructs which include, but are not limited to, either the intracellular domain of rhesus MC-3R as an in-frame fusion at the carboxy terminus of the GST gene, or the extracellular and transmembrane ligand binding domain of MC-3R fused to the amino terminus of GST, or the extracellular and transmembrane domain of MC-3R fused to an immunoglobulin gene by methods known to one of ordinary skill in the art.
  • GST glutathione S- transferase
  • Soluble recombinant GST-MC-3R fusion proteins may be expressed in various expression systems, including Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen).
  • the present invention also relates to assays to screen or select for various modulators of MC-3R activity, methods of expressing the rhesus MC-3R protein and biological equivalents disclosed herein, recombinant host cells which comprise DNA constructs which express these receptor proteins, and compounds identified through these assays which act as agonists or antagonists of MC-3R activity.
  • Any such polynucleotide includes but is not necessarily limited to nucleotide substitutions, deletions, additions, amino-terminal truncations and carboxy-terminal truncations such that these mutations encode mRNA which express a protein or protein fragment of diagnostic, therapeutic or prophylactic use and would be useful for screening for agonists and/or antagonists for vertebrate MC-3R function. It is a further object of the present invention to provide the rhesus MC-
  • MC- 3R-based assays to select for modulators of this receptor protein.
  • These assays are preferably cell based assays whereby a DNA molecule encoding MC-3R is transfected or transformed into a host cell, this recombinant host cell is allowed to grow for a time sufficient to express MC-3R prior to use in various assays described herein.
  • membrane preparations from host cells transfected or transformed with a DNA molecule encoding MC-3R for use in assays to select for modulators of MC-3R activity.
  • MC-3R- based assays to select for modulators of this receptor protein.
  • These assays are preferably cell based assays whereby a DNA molecule encoding rhMC-3R is transfected or transformed into a host cell, this recombinant host cell is allowed to grow for a time sufficient to express MC-3R prior to use in various assays described herein. It is a further object to provide for membrane preparations from host cells transfected or transformed with a DNA molecule encoding rhMC-3R for use in assays to select for modulators of MC-3R activity.
  • rh or refers to —rhesus--.
  • M-3R refers to —melanocortin 3 receptor—.
  • GPCR refers to — G-protein coupled receptor—.
  • mamalian host will refer to any mammal, including a human being.
  • Figure 1 A and Figure IB show the nucleotide sequence which encodes rhesus MC-3R, as set forth in SEQ ID NO:l.
  • Figure 2 shows the one- letter designation of the amino acid sequence of rhesus MC-3R, as also set forth in SEQ ID NO:2 as a three letter designation.
  • Figure 3A and Figure 3B show characterization of rhesus MC-3R.
  • Figure 3 A shows inhibition of [ 125]NDP-a-MSH binding by NDP- ⁇ -MSH, ⁇ 2-MSH or Shu-9119.
  • Figure 3B shows stimulation of cAMP synthesis in response to NDP- ⁇ - MSH or ⁇ 2-MSH.
  • Each curve represents a typical experiment with duplicate measurements. Average data from multiple experiments are listed in Table 2.
  • Figure 4 shows the comparison of binding affinity (IC50) and activation potency (EC50) for the MC-3R from 2 species.
  • Abbreviations are as follows: A, human ACTH(l-24); ⁇ , ⁇ -MSH; ⁇ , ⁇ 2-MSH; M, MT-II; N, NDP- ⁇ -MSH.
  • the present invention relates to an isolated nucleic acid molecule (polynucleotide) which encodes a novel rhesus monkey (Macaco mulatto) melanocortin-3 receptor (rhMC-3R).
  • the nucleic acid molecules of the present invention are substantially free from other nucleic acids.
  • DNA is a preferred nucleic acid.
  • the present invention relates to an isolated nucleic acid molecule (polynucleotide) which encodes mRNA which expresses a novel rhesus MC-3R, this DNA molecule comprising the nucleotide sequence disclosed herein as SEQ ID NO:l, shown herein as follows:
  • the above-exemplified isolated DNA molecule shown in Figure 1A- 1B and set forth as SEQ ID NO:l, contains 1909 nucleotides.
  • This DNA molecule contains an open reading frame from nucleotide 148 to nucleotide 1116, with a "TAG" termination codon at nucleotides 1117-1119.
  • This open reading frame encodes a rhesus MC-3R protein 323 amino acids in length, as shown in Figure 2 and as set forth in SEQ ID NO:2.
  • the present invention also relates to biologically active fragments or mutants of SEQ ID NO:l which encodes mRNA expressing a novel rhesus MC-3R. Any such biologically active fragment and/or mutant will encode either a protein or protein fragment which at least substantially mimics the pharmacological properties of a wild-type MC-3R protein, including but not limited to the rhesus MC-3R receptor protein as set forth in SEQ ID NO:2.
  • any such polynucleotide includes but is not necessarily limited to nucleotide substitutions, deletions, additions, amino-terminal truncations and carboxy-terminal truncations such that these mutations encode mRNA which express a protein or protein fragment of diagnostic, therapeutic or prophylactic use and would be useful for screening for agonists and/or antagonists for MC-3R function.
  • FIG. 1 A - IB a rhesus cDNA molecule encoding a novel MC-3R (SEQ ID NO:l).
  • the isolated nucleic acid molecules of the present invention may include a deoxyribonucleic acid molecule (DNA), such as genomic DNA and complementary DNA (cDNA), which may be single (coding or noncoding strand) or double stranded, as well as synthetic DNA, such as a synthesized, single stranded polynucleotide.
  • DNA deoxyribonucleic acid molecule
  • cDNA complementary DNA
  • synthetic DNA such as a synthesized, single stranded polynucleotide.
  • the isolated nucleic acid molecule of the present invention may also include a ribonucleic acid molecule (RNA).
  • this invention is also directed to those DNA sequences encode RNA comprising alternative codons which code for the eventual translation of the identical amino acid, as shown below:
  • the present invention discloses codon redundancy which may result in differing DNA molecules expressing an identical protein.
  • a sequence bearing one or more replaced codons will be defined as a degenerate variation.
  • mutations either in the DNA sequence or the translated protein which do not substantially alter the ultimate physical properties of the expressed protein. For example, substitution of valine for leucine, arginine for lysine, or asparagine for glutamine may not cause a change in functionality of the polypeptide.
  • DNA sequences coding for a peptide may be altered so as to code for a peptide having properties that are different than those of the naturally occurring peptide.
  • Methods of altering the DNA sequences include but are not limited to site directed mutagenesis. Examples of altered properties include but are not limited to changes in the affinity of an enzyme for a substrate or a receptor for a ligand.
  • 5' and/or 3' RACE may be performed to generate a full-length cDNA sequence. This strategy involves using gene-specific oligonucleotide primers for PCR amplification of rhesus MC-3R cDNA.
  • These gene-specific primers are designed through identification of an expressed sequence tag (EST) nucleotide sequence which has been identified by searching any number of publicly available nucleic acid and protein databases; (2) direct functional expression of the rhesus MC-3R cDNA following the construction of a rhesus MC-3R-containing cDNA library in an appropriate expression vector system; (3) screening a rhesus MC-3R-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a labeled degenerate oligonucleotide probe designed from the amino acid sequence of the rhesus MC-3R protein; (4) screening a rhesus MC-3R-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA encoding the rhesus MC-3R protein.
  • EST expressed sequence tag
  • This partial cDNA is obtained by the specific PCR amplification of rhesus MC-3R DNA fragments through the design of degenerate oligonucleotide primers from the amino acid sequence known for other kinases which are related to the rhesus MC-3R protein; (5) screening a rhesus MC-3R-containing cDNA library constructed in a bacteriophage or plasmid shuttle vector with a partial cDNA or oligonucleotide with homology to a mammalian MC-3R protein.
  • This strategy may also involve using gene-specific oligonucleotide primers for PCR amplification of rhesus MC-3R cDNA identified as an EST as described above; or (6) designing 5' and 3' gene specific oligonucleotides using SEQ ID NO: 1 as a template so that either the full-length cDNA may be generated by known RACE techniques, or a portion of the coding region may be generated by these same known RACE techniques to generate and isolate a portion of the coding region to use as a probe to screen one of numerous types of cDNA and/or genomic libraries in order to isolate a full-length version of the nucleotide sequence encoding rhesus MC-3R.
  • libraries as well as libraries constructed from other cell types-or species types, may be useful for isolating a rhesus MC-3R-encoding DNA or a rhesus MC-3R homologue.
  • Other types of libraries include, but are not limited to, cDNA libraries derived from other rhesus cells.
  • cDNA libraries may be prepared from cells or cell lines which have MC-3R activity.
  • the selection of cells or cell lines for use in preparing a cDNA library to isolate a cDNA encoding rhesus MC-3R may be done by first measuring cell-associated MC-3R activity using any known assay available for such a purpose.
  • Preparation of cDNA libraries can be performed by standard techniques well known in the art. Well known cDNA library construction techniques can be found for example, in Sambrook et al., 1989, Molecular Cloning: A Laboratory Manual; Cold Spring Harbor Laboratory, Cold Spring Harbor, New York. Complementary DNA libraries may also be obtained from numerous commercial sources, including but not limited to Clontech Laboratories, Inc. and Stratagene.
  • DNA encoding rhesus MC-3R may also be isolated from a suitable genomic DNA library. Construction of genomic DNA libraries can be performed by standard techniques well known in the art. Well known genomic DNA library construction techniques can be found in Sambrook, et al., supra.
  • the amino acid sequence or DNA sequence of rhesus MC-3R or a homologous protein may be necessary.
  • the MC-3R protein or a homologous protein may be purified and partial amino acid sequence determined by automated sequenators. It is not necessary to determine the entire amino acid sequence, but the linear sequence of two regions of 6 to 8 amino acids can be determined for the PCR amplification of a partial rhesus MC-3R DNA fragment. Once suitable amino acid sequences have been identified, the DNA sequences capable of encoding them are synthesized.
  • the amino acid sequence can be encoded by any of a set of similar DNA oligonucleotides. Only one member of the set will be identical to the rhesus MC-3R sequence but others in the set will be capable of hybridizing to rhesus MC-3R DNA even in the presence of DNA oligonucleotides with mismatches. The mismatched DNA oligonucleotides may still sufficiently hybridize to the rhesus MC-3R DNA to permit identification and isolation of rhesus MC-3R encoding DNA.
  • nucleotide sequence of a region of an expressed sequence may be identified by searching one or more available genomic databases.
  • Gene-specific primers may be used to perform PCR amplification of a cDNA of interest from either a cDNA library or a population of cDNAs.
  • the appropriate nucleotide sequence for use in a PCR-based method may be obtained from SEQ ID NO: 1, either for the purpose of isolating overlapping 5' and 3' RACE products for generation of a full- length sequence coding for rhesus MC-3R, or to isolate a portion of the nucleotide sequence coding for rhesus MC-3R for use as a probe to screen one or more cDNA- or genomic-based libraries to isolate a full-length sequence encoding rhesus MC-3R or rhesus MC-3R-like proteins.
  • DNA sequences that hybridize to SEQ LD NO:l under stringent conditions include DNA sequences that hybridize to SEQ LD NO:l under stringent conditions.
  • a procedure using conditions of high stringency is as follows: Prehybridization of filters containing DNA is carried out for 2 hours to overnight at 65 °C in buffer composed of 6X SSC, 5X Denhardt's solution, and 100 ⁇ g/ml denatured salmon sperm DNA. Filters are hybridized for 12 to 48 hrs at 65 °C in prehybridization mixture containing 100 ⁇ g ml denatured salmon sperm DNA and 5-20 X lO 6 cpm of 32 P-labeled probe. Washing of filters is done at 37°C for 1 hr in a solution containing 2X SSC, 0.1%
  • interhelical disulfide bond (e.g., between the Cys residues near the top of TM3 and TM5) may play the same function as interloop disulfide bond in most other GPCR's.
  • the present invention also relates to a substantially purified form of the rhesus melanocortin-3 receptor protein, which comprises the amino acid sequence is disclosed in Figure 2 and set forth as SEQ ID NO:2.
  • the present invention also relates to biologically active fragments and/or mutants of the rhesus melanocortin-3 receptor protein comprising the amino acid sequence set forth as SEQ LD NO:2, including but not necessarily limited to amino acid substitutions, deletions, additions, amino terminal truncations and carboxy-terminal truncations such that these mutations provide for proteins or protein fragments of diagnostic, therapeutic or prophylactic use and would be useful for screening for agonists and/or antagonists for MC-3R function.
  • G C N S M N L G (SEQ ID NO:2), comprises the amino acid sequence of wild type rhesus melanocortin-3 receptor protein.
  • MC-3R protein may be recovered to provide MC-3R protein in active form.
  • MC-3R protein purification procedures are available and suitable for use.
  • Recombinant MC-3R protein may be purified from cell lysates and extracts by various combinations of, or individual application of salt fractionation, ion exchange chromatography, size exclusion chromatography, hydroxylapatite adsorption chromatography and hydrophobic interaction chromatography.
  • recombinant MC-3R protein can be separated from other cellular proteins by use of an immunoaffinity column made with monoclonal or polyclonal antibodies specific for full-length MC-3R protein, or polypeptide fragments of MC-3R protein.
  • the present invention also relates to isolated nucleic acid molecules which are fusion constructions expressing fusion proteins useful in assays to identify compounds which modulate wild-type vertebrate MC-3R activity.
  • a preferred aspect of this portion of the invention includes, but is not limited to, glutathione S- transferase (GST)-MC-3R fusion constructs which include, but are not limited to, either the intracellular domain of rhesus MC-3R as an in- frame fusion at the carboxy terminus of the GST gene or the extracellular and transmembrane ligand binding domain of MC3R fused to an GST or immunoglobulin gene by methods known to one of ordinary skill in the art.
  • GST glutathione S- transferase
  • Recombinant GST-MC-3R fusion proteins may be expressed in various expression systems, including Spodoptera frugiperda (Sf21) insect cells (Invitrogen) using a baculovirus expression vector (pAcG2T, Pharmingen).
  • the present invention also relates to subcellular membrane fractions from the recombinant host cells (both prokaryotic and eukaryotic as well as both stably and transiently transformed cells) which contain the nucleic acids of the present invention.
  • These subcellular membrane fractions will comprise either wild-type or mutant forms of rhesus melanocortin-3 receptor proteins at levels substantially above endogenous levels and hence will be useful in various assays described throughout this specification.
  • the present invention also relates to recombinant vectors and recombinant hosts, both prokaryotic and eukaryotic, which contain the substantially purified nucleic acid molecules disclosed throughout this specification.
  • the nucleic acid molecules of the present invention encoding rhMC-3R in whole or in part, can be linked with other DNA molecules, i.e, DNA molecules to which the rhMC-3R are not naturally linked, to form "recombinant DNA molecules" containing the receptor.
  • the novel DNA sequences of the present invention can be inserted into vectors which comprise nucleic acids encoding a rhMC-3R or a functional equivalent. These vectors may be comprised of DNA or RNA; for most cloning purposes DNA vectors are preferred.
  • Typical vectors include plasmids, modified viruses, bacteriophage and cosmids, yeast artificial chromosomes and other forms of episomal or integrated DNA that can encode a rhMC-3R. It is well within the skilled artisan to determine an appropriate vector for a particular gene transfer or other use.
  • mammalian expression vectors may be used to express recombinant rhesus MC-3R in mammalian cells.
  • Expression vectors are defined herein as DNA sequences that are required for the transcription of cloned DNA and the translation of their mRNAs in an appropriate host.
  • Such vectors can be used to express eukaryotic DNA in a variety of hosts such as bacteria, blue green algae, plant cells, insect cells and animal cells. Specifically designed vectors allow the shuttling of DNA between hosts such as bacteria-yeast or bacteria-animal cells.
  • An appropriately constructed expression vector should contain: an origin of replication for autonomous replication in host cells, selectable markers, a limited number of useful restriction enzyme sites, a potential for high copy number, and active promoters.
  • a promoter is defined as a DNA sequence that directs RNA polymerase to bind to DNA and initiate RNA synthesis.
  • a strong promoter is one which causes mRNAs to be initiated at high frequency.
  • Expression vectors may include, but are not limited to, cloning vectors, modified cloning vectors, specifically designed plasmids or viruses.
  • mammalian expression vectors which may be suitable for recombinant rhesus MC-3R expression, include but are not limited to, pcDNA3.neo (Invitrogen), pcDNA3.1 (Invitrogen), pCI-neo (Promega), pLITMUS28, pLITMUS29, pLITMUS38 and pLITMUS39 (New England Bioloabs), pcDNAI, pcDNAIamp (Invitrogen), pcDNA3 (Invitrogen), pMClneo (Stratagene), pXTl (Stratagene), pSG5 (Stratagene), EBO-pSV2-neo (ATCC 37593) pBPV-l(8-2) (ATCC 37110), pdBPV-MMTneo(342-12) (ATCC 37224), pRSVgpt (ATCC 37199), pRSVneo (ATCC 37198), pSV2-
  • bacterial expression vectors may be used to express recombinant rhesus MC-3R in bacterial cells.
  • Commercially available bacterial expression vectors which may be suitable for recombinant rhesus MC-3R expression include, but are not limited to pCR2.1 (Invitrogen), pETl la (Novagen), lambda gtl 1 (Invitrogen), and pKK223-3 (Pharmacia).
  • fungal cell expression vectors may be used to express recombinant rhesus MC-3R in fungal cells.
  • Commercially available fungal cell expression vectors which may be suitable for recombinant rhesus MC-3R expression include but are not limited to pYES2 (Invitrogen) and Pichia expression vector (Invitrogen).
  • insect cell expression vectors may be used to express recombinant receptor in insect cells.
  • Commercially available insect cell expression vectors which may be suitable for recombinant expression of rhesus MC-3R include but are not limited to pBlueBacIII and pBlueBacHis2 (Invitrogen), and pAcG2T (Pharmingen).
  • Expression of rhesus MC-3R DNA may also be performed using in vitro produced synthetic mRNA.
  • Synthetic mRNA can be efficiently translated in various cell-free systems, including but not limited to wheat germ extracts and reticulocyte extracts, as well as efficiently translated in cell based systems, including but not limited to microinj ection into frog oocytes, with microinj ection into frog oocytes being preferred.
  • cDNA molecules including but not limited to the following can be constructed: a cDNA fragment containing the full-length open reading frame for rhesus MC-3R as well as various constructs containing portions of the cDNA encoding only specific domains of the protein or rearranged domains of the protein. All constructs can be designed to contain none, all or portions of the 5' and/or 3' untranslated region of a rhesus MC-3R cDNA. The expression levels and activity of rhesus MC-3R can be determined following the introduction, both singly and in combination, of these constructs into appropriate host cells.
  • this MC-3R cDNA construct is transferred to a variety of expression vectors (including recombinant viruses), including but not limited to those for mammalian cells, plant cells, insect cells, oocytes, bacteria, and yeast cells.
  • expression vectors including recombinant viruses
  • this invention includes modified rhMC-3R polypeptides which have amino acid deletions, additions, or substitutions but that still retain substantially the same biological activity as rhMC-3R. It is generally accepted that single amino acid substitutions do not usually alter the biological activity of a protein (see, e.g., Molecular Biology of the
  • the present invention includes isolated nucleic acid molecules and expressed MC-3R proteins wherein one amino acid substitution is generated and which this protein retains substantially the same biological activity as wild-type rhMC-3R.
  • the present invention also includes isolated nucleic acid molecules and expressed MC-3R proteins wherein two or more amino acid substitution is generated wherein this protein retains substantially the same biological activity as wild-type rhMC-3R.
  • the present invention includes embodiments where the above-described substitutions are conservative substitutions.
  • the present invention includes embodiments where the above-described substitutions do not occur in the ligand-binding domain of rhMC-3R.
  • this invention also includes vectors containing an rhMC-3R gene, host cells containing the vectors, and methods of making substantially pure rhMC-3R protein comprising the steps of introducing the rhMC-3R gene into a host cell, and cultivating the host cell under appropriate conditions such that rhMC-3R is produced.
  • the rhMC-3R so produced may be harvested from the host cells in conventional ways. Therefore, the present invention also relates to methods of expressing the rhesus MC-3R protein and biological equivalents disclosed herein, assays employing these gene products, recombinant host cells which comprise DNA constructs which express these receptor proteins, and compounds identified through these assays which act as agonists or antagonists of MC-3R activity.
  • the cloned rhesus MC-3R cDNA obtained through the methods described above may be recombinantly expressed by molecular cloning into an expression vector (such as pcDNA3.neo, pcDNA3.1 , pCR2.1 , pBlueBacHis2 or pLITMUS28) containing a suitable promoter and other appropriate transcription regulatory elements, and transferred into prokaryotic or eukaryotic host cells to produce recombinant rhesus MC-3R.
  • an expression vector such as pcDNA3.neo, pcDNA3.1 , pCR2.1 , pBlueBacHis2 or pLITMUS28
  • a suitable promoter and other appropriate transcription regulatory elements such as pcDNA3.neo, pcDNA3.1 , pCR2.1 , pBlueBacHis2 or pLITMUS28
  • another aspect of the present invention includes host cells that have been engineered to contain and/or express DNA sequences encoding the rhMC-3R.
  • Such recombinant host cells can be cultured under suitable conditions to produce rhMC-3R or a biologically equivalent form.
  • Recombinant host cells may be prokaryotic or eukaryotic, including but not limited to, bacteria such as E. coli, fungal cells such as yeast, mammalian cells including, but not limited to, cell lines of human, bovine, porcine, monkey and rodent origin, and insect cells including but not limited to Drosophila and silkworm derived cell lines.
  • an expression vector containing DNA encoding a rhesus MC-3R-like protein may be used for expression of rhesus MC-3R in a recombinant host cell.
  • Recombinant host cells may be prokaryotic or eukaryotic, including but not limited to bacteria such as E. coli, fungal cells such as yeast, mammalian cells including but not limited to cell lines of human, bovine, porcine, monkey and rodent origin, and insect cells including but not limited to Drosophila- and silkworm-derived cell lines.
  • one insect expression system utilizes Spodoptera frugiperda (Sf21) insect cells (Invitrogen) in tandem with a baculovirus expression vector (pAcG2T, Pharmingen).
  • mammalian species which may be suitable and which are commercially available, include but are not limited to, L cells L-M(TK") (ATCC CCL 1.3), L cells L-M (ATCC CCL 1.2), Saos-2 (ATCC HTB-85), 293 (ATCC CRL 1573), Raji (ATCC CCL 86), CV-1 (ATCC CCL 70), COS-1 (ATCC CRL 1650), COS-7 (ATCC CRL 1651), CHO-K1 (ATCC CCL 61), 3T3 (ATCC CCL 92), NIH/3T3 (ATCC CRL 1658), HeLa (ATCC CCL 2), C127I (ATCC CRL 1616), BS-C-1 (ATCC CCL 26), MRC-5 (ATCC CCL 171) and
  • the assays described herein as well as protein purification schemes can be carried out with cells that have been transiently or stably transfected or transformed with an expression vector which directs expression of MC-3R.
  • the expression vector may be introduced into host cells via any one of a number of techniques including but not limited to transformation, transfection, protoplast fusion, and electroporation. Transformation is meant to encompass a genetic change to the target cell resulting from an incorporation of DNA.
  • Transfection is meant to include any method known in the art for introducing MC-3R into the test cells. For example, transfection includes calcium phosphate or calcium chloride mediated transfection, lipofection, infection with a retroviral construct containing MC-3R, and electroporation.
  • the expression vector-containing cells are individually analyzed to determine whether they produce human MC-3R protein. Identification of human MC-3R expressing cells may be done by several means, including but not limited to immunological reactivity with anti-human MC-3R antibodies, labeled ligand binding and the presence of host cell-associated human MC-3R activity.
  • the specificity of binding of compounds showing affinity for rhMC- 3R is shown by measuring the affinity of the compounds for recombinant cells expressing the cloned receptor or for membranes from these cells.
  • Expression of the cloned receptor and screening for compounds that bind to rhMC-3R or that inhibit the binding of a known, radiolabeled ligand of rhMC-3R to these cells, or membranes prepared from these cells provides an effective method for the rapid selection of compounds with high affinity for rhMC-3R.
  • ligands need not necessarily be radiolabeled but can also be nonisotopic compounds that can be used to displace bound radiolabeled compounds or that can be used as activators in functional assays.
  • Compounds identified by the above method are likely to be agonists or antagonists of rhMC-3R and may be peptides, proteins, or non-proteinaceous organic molecules. Accordingly, the present invention is directed to methods for screening for compounds which modulate the expression of DNA or RNA encoding a MC-3R protein as well as compounds which effect the function of the MC-3R protein. Methods for identifying agonists and antagonists of other receptors are well known in the art and can be adapted to identify agonists and antagonists of MC-3R. For example, Cascieri et al. (1992, Molec. Pharmacol.
  • the method involves transfecting COS cells with expression vectors containing rat neurokinin receptors, allowing the transfected cells to grow for a time sufficient to allow the neurokinin receptors to be expressed, harvesting the transfected cells and resuspending the cells in assay buffer containing a known radioactively labeled agonist of the neurokinin receptors either in the presence or the absence of the substance, and then measuring the binding of the radioactively labeled known agonist of the neurokinin receptor to the neurokinin receptor.
  • the substance is a potential agonist or antagonist of the neurokinin receptor.
  • binding of the substance such as an agonist or antagonist to MC-3R can be measured by employing a labeled substance or agonist.
  • the substance or agonist can be labeled in any convenient manner known to the art, e.g., radioactively, fluorescently, enzymatically.
  • the specificity of binding of compounds having affinity for MC-3R is shown by measuring the affinity of the compounds for recombinant cells expressing the cloned receptor or for membranes from these cells.
  • Expression of the cloned receptor and screening for compounds that bind to MC-3R or that inhibit the binding of a known, radiolabeled ligand of MC-3R to these cells, or membranes prepared from these cells provides an effective method for the rapid selection of compounds with high affinity for MC-3R.
  • ligands need not necessarily be radiolabeled but can also be nonisotopic compounds that can be used to displace bound radiolabeled compounds or that can be used as activators in functional assays.
  • Compounds identified by the above method are likely to be agonists or antagonists of MC-3R and may be peptides, proteins, or non-proteinaceous organic molecules. Compounds may modulate by increasing or attenuating the expression of DNA or RNA encoding MC-3R, or by acting as an agonist or antagonist of the MC-3R receptor protein. These compounds that modulate the expression of DNA or RNA encoding MC-3R or the biological function thereof may be detected by a variety of assays.
  • the assay may be a simple "yes/no" assay to determine whether there is a change in expression or function.
  • the assay may be made quantitative by comparing the expression or function of a test sample with the levels of expression or function in a standard sample.
  • Kits containing MC-3R, antibodies to MC-3R, or modified MC- 3R may be prepared by known methods for such uses. Therefore, the present invention relates to methods of expressing rhMC-3R in recombinant systems and of identifying agonists and antagonists of rhMC-3R.
  • screening compounds in order to identify potential pharmaceuticals that specifically interact with a target receptor it is necessary to ensure that the compounds identified are as specific as possible for the target receptor. To do this, it is necessary to screen the compounds against as wide an array as possible of receptors that are similar to the target receptor.
  • the rhMC-3R of this invention is believed to function similarly to GPCRs and have similar biological activity. They are useful in understanding the biological and physiological effects in the rhesus to in identify melanocortin active process in primates, followed by human clinical trials. More notable, rhMC-3R agonists will be identified and evaluated for their effects on food intake, weight gain, and metabolic rate to identify novel-anti-obesity agents that are effective in primates. They may also be used to scan for rhesus monkey melanocortin agonists and antagonists; as in particular to test the specificity of identified ligands.
  • the present invention relates in part to methods of identifying a substance which modulates MC-3R receptor activity, which involves: (a) combining a test substance in the presence and absence of a MC-3R receptor protein wherein said MC-3R receptor protein comprises the amino acid sequence as set forth in SEQ LD NO:2; and, (b) measuring and comparing the effect of the test substance in the presence and absence of the MC-3R receptor protein.
  • the present invention includes assays by which MC-3R modulators (such as agonists and antagonists) may be identified. Accordingly, the present invention includes a method for determining whether a substance is a potential agonist or antagonist of MC-3R that comprises:
  • step (c) of the method is practiced are conditions that are typically used in the art for the study of protein-ligand interactions: e.g., physiological pH; salt conditions such as those represented by such commonly used buffers as PBS or in tissue culture media; a temperature of about 4°C to about 55°C.
  • the test cells may be harvested and resuspended in the presence of the substance and the labeled ligand.
  • step (c) is modified in that the cells are not harvested and resuspended but rather the radioactively labeled known agonist and the substance are contacted with the cells while the cells are attached to a substratum, e.g., tissue culture plates.
  • the present invention also includes a method for determining whether a substance is capable of binding to MC-3R, i.e., whether the substance is a potential agonist or an antagonist of MC-3R, where the method comprises:
  • step (b) of the method is practiced are conditions that are typically used in the art for the study of protein-ligand interactions: e.g., physiological pH; salt conditions such as those represented by such commonly used buffers as PBS or in tissue culture media; a temperature of about 4°C to about 55°C.
  • the test cells are harvested and resuspended in the presence of the substance. Chen et al. (1995, Analytical Biochemistry 226: 349-354) describe a colorometric assay which utilizes a recombinant cell transfected with an expression vector encoding a G-protein coupled receptor with a second expression vector containing a promoter with a c AMP responsive element fused to the LacZ gene.
  • Another aspect of this portion of the invention includes a non-radioactive method for determining whether a substance is a potential agonist or antagonist of MC-3R that comprises:
  • step (b) transfecting or transforming the test cells of step (a) with an expression vector which comprises a cAMP-inducible promoter fused to a colorometric gene such a LacZ;
  • step (c) subsequently or concurrently to step (b), exposing the test cells to a substance that is a suspected antagonist of MC-3R;
  • step (a) the method of II wherein the first and second expression vectors of step (a) are replaced with a single expression vector which expresses a chimeric MC-3R protein fused at its C-terminus to a promiscuous G-protein.
  • Another embodiment of the present invention includes a method for determining whether a substance binds and/or is a potential agonist or antagonist of MC-3R wherein membrane preparations from the test cells are utilized in place of the test cells.
  • Such methods comprise the following and may utilized the physiological conditions as noted above:
  • step (c) subsequently or concurrently to step (b), exposing the membranes from the test cells to a substance; (d) measuring the amount of binding of the ligand to the MC-3R in the membranes in the presence and the absence of the substance;
  • the present invention also relates to a method for determining whether a substance is capable of binding to MC-3R comprising:
  • a preferred embodiment of the present invention is determining various ligand binding affinities using 125jM a beled NDP- ⁇ -MSH as the labeled ligand in the presence of varying concentration of unlabeled ligands.
  • the activation of the second messenger pathway may be determined by measuring the intracellular cAMP elicited by agonist at various concentration.
  • the present invention also relates to polyclonal and monoclonal antibodies raised in response to either the rhesus form of MC-3R, or a biologically active fragment thereof. Polyclonal or monoclonal antibodies may be raised against rhesus MC-3R or a synthetic peptide (usually from about 9 to about 25 amino acids in length) from a portion of rhesus MC-3R as disclosed in SEQ LD NO:2.
  • Monospecific antibodies to rhesus MC-3R are purified from mammalian antisera containing antibodies reactive against rhesus MC-3R or are prepared as monoclonal antibodies reactive with rhesus MC-3R using the technique of Kohler and Milstein (1975, Nature 256: 495-497).
  • Monospecific antibody as used herein is defined as a single antibody species or multiple antibody species with homogenous binding characteristics for rhesus MC-3R.
  • Homogenous binding as used herein refers to the ability of the antibody species to bind to a specific antigen or epitope, such as those associated with rhesus MC-3R, as described above.
  • Rhesus MC-3R-specific antibodies are raised by immunizing animals such as mice, rats, guinea pigs, rabbits, goats, horses and the like, with an appropriate concentration of rhesus MC-3R protein or a synthetic peptide generated from a portion of rhesus MC-3R with or without an immune adjuvant.
  • Preimmune serum is collected prior to the first immunization.
  • Each animal receives between about 0.1 ⁇ g and about 1000 ⁇ g of rhesus MC-3R protein associated with an acceptable immune adjuvant.
  • acceptable adjuvants include, but are not limited to, Freund's complete, Freund's incomplete, alum-precipitate, water in oil emulsion containing Corynebacterium parvum and tRNA.
  • the initial immunization consists of rhesus MC-3R protein or peptide fragment thereof in, preferably, Freund's complete adjuvant at multiple sites either subcutaneously (SC), intraperitoneally (LP) or both.
  • SC subcutaneously
  • LP intraperitoneally
  • Each animal is bled at regular intervals, preferably weekly, to determine antibody titer.
  • the animals may or may not receive booster injections following the initial immunization. Those animals receiving booster injections are generally given an equal amount of rhesus MC-3R in Freund's incomplete adjuvant by the same route. Booster injections are given at about three week intervals until maximal titers are obtained. At about 7 days after each booster immunization or about weekly after a single immunization, the animals are bled, the serum collected, and aliquots are stored at about -20°C.
  • Monoclonal antibodies (mAb) reactive with rhesus MC-3R are prepared by immunizing inbred mice, preferably Balb/c, with rhesus MC-3R protein.
  • the mice are immunized by the LP or SC route with about 1 ⁇ g to about 100 ⁇ g, preferably about 10 ⁇ g, of rhesus MC-3R protein in about 0.5 ml buffer or saline incorporated in an equal volume of an acceptable adjuvant, as discussed above. Freund's complete adjuvant is preferred.
  • the mice receive an initial immunization on day 0 and are rested for about 3 to about 30 weeks.
  • Immunized mice are given one or more booster immunizations of about 1 to about 100 ⁇ g of rhesus MC-3R in a buffer solution such as phosphate buffered saline by the intravenous (TV) route.
  • Lymphocytes from antibody positive mice, preferably splenic lymphocytes, are obtained by removing spleens from immunized mice by standard procedures known in the art.
  • Hybridoma cells are produced by mixing the splenic lymphocytes with an appropriate fusion partner, preferably myeloma cells, under conditions which will allow the formation of stable hybridomas.
  • Fusion partners may include, but are not limited to: mouse myelomas P3/NSl/Ag 4-1; MPC-11; S-194 and Sp 2/0, with Sp 2/0 being preferred.
  • the antibody producing cells and myeloma cells are fused in polyethylene glycol, about 1000 mol. wt, at concentrations from about 30% to about 50%.
  • Fused hybridoma cells are selected by growth in hypoxanthine, thymidine and aminopterin supplemented Dulbecco's Modified Eagles Medium (DMEM) by procedures known in the art.
  • DMEM Dulbecco's Modified Eagles Medium
  • Supernatant fluids are collected form growth positive wells on about days 14, 18, and 21 and are screened for antibody production by an immunoassay such as solid phase immunoradioassay (SPLRA) using rhesus MC-3R as the antigen.
  • the culture fluids are also tested in the Ouchterlony precipitation assay to determine the isotype of the mAb.
  • Hybridoma cells from antibody positive wells are cloned by a technique such as the soft agar technique of MacPherson, 1973, Soft Agar Techniques, in Tissue Culture Methods and Applications, Kruse and Paterson, Eds., Academic Press.
  • Monoclonal antibodies are produced in vivo by injection of pristine primed Balb/c mice, approximately 0.5 ml per mouse, with about 2 x 106 to about 6 x l ⁇ 6 hybridoma cells about 4 days after priming. Ascites fluid is collected at approximately 8-12 days after cell transfer and the monoclonal antibodies are purified by techniques known in the art.
  • In vitro production of anti -rhesus MC-3R mAb is carried out by growing the hybridoma in DMEM containing about 2% fetal calf serum to obtain sufficient quantities of the specific mAb. The mAb are purified by techniques known in the art.
  • Antibody titers of ascites or hybridoma culture fluids are determined by various serological or immunological assays which include, but are not limited to, precipitation, passive agglutination, enzyme-linked immunosorbent antibody (ELISA) technique and radioimmunoassay (RIA) techniques. Similar assays are used to detect the presence of rhesus MC-3R in body fluids or tissue and cell extracts.
  • Rhesus MC-3R antibody affinity columns are made, for example, by adding the antibodies to Affigel-10 (Biorad), a gel support which is pre-activated with N-hydroxysuccinimide esters such that the antibodies form covalent linkages with the agarose gel bead support.
  • the antibodies are then coupled to the gel via amide bonds with the spacer arm.
  • the remaining activated esters are then quenched with 1M ethanolamine HCl (pH 8).
  • the column is washed with water followed by 0.23 M glycine HCl (pH 2.6) to remove any non-conjugated antibody or extraneous protein.
  • the column is then equilibrated in phosphate buffered saline (pH 7.3) and the cell culture supernatants or cell extracts containing full-length rhesus MC-3R or rhesus MC-3R protein fragments are slowly passed through the column.
  • the column is then washed with phosphate buffered saline until the optical density (A280) falls to background, then the protein is eluted with 0.23 M glycine-HCl (pH 2.6).
  • the purified rhesus MC-3R protein is then dialyzed against phosphate buffered saline.
  • the assays described above can be carried out with cells that have been transiently or stably transfected or stably transformed with rhMC-3R.
  • Transfection is meant to include any method known in the art for introducing rhMC- 3R into the test cells.
  • transfection includes calcium phosphate or calcium chloride mediated transfection, lipofection, infection with a retroviral construct containing rhMC-3R, and electroporation. Transformation is meant to encompass a genetic change to the target cell resulting from an incorporation of DNA.
  • binding of the substance or agonist to rhMC-3R is measured, such binding can be measured by employing a labeled substance or agonist.
  • the substance or agonist can be labeled in any convenient manner known to the art, e.g., radioactively, fluorescently, enzymatically.
  • the DNA molecules, RNA molecules, recombinant protein and antibodies of the present invention may be used to screen and measure levels of rhesus MC-3R.
  • the recombinant proteins, DNA molecules, RNA molecules and antibodies lend themselves to the formulation of kits suitable for the detection and typing of rhesus MC-3R.
  • a kit would comprise a compartmentalized carrier suitable to hold in close confinement at least one container.
  • the carrier would further comprise reagents such as recombinant MC-3R or anti-MC-3R antibodies suitable for detecting rhesus MC-3R.
  • the carrier may also contain a means for detection such as labeled antigen or enzyme substrates or the like.
  • compositions comprising modulators of rhesus MC-3R may be formulated according to known methods such as by the admixture of a pharmaceutically acceptable carrier. Examples of such carriers and methods of formulation may be found in Remington's Pharmaceutical Sciences. To form a pharmaceutically acceptable composition suitable for effective administration, such compositions will contain an effective amount of the protein, DNA, RNA, modified rhesus MC-3R, or either MC-3R agonists or antagonists including tyrosine kinase activators or inhibitors. Therapeutic or diagnostic compositions of the invention are administered to an individual in amounts sufficient to treat or diagnose disorders. The effective amount may vary according to a variety of factors such as the individual's condition, weight, sex and age. Other factors include the mode of administration. The pharmaceutical compositions may be provided to the individual by a variety of routes such as subcutaneous, topical, oral and intramuscular.
  • chemical derivative describes a molecule that contains additional chemical moieties which are not normally a part of the base molecule. Such moieties may improve the solubility, half-life, absorption, etc. of the base molecule. Alternatively the moieties may attenuate undesirable side effects of the base molecule or decrease the toxicity of the base molecule. Examples of such moieties are described in a variety of texts, such as Remington's Pharmaceutical Sciences.
  • the present invention also has the objective of providing suitable topical, oral, systemic and parenteral pharmaceutical formulations for use in the novel methods of treatment of the present invention.
  • the compositions containing compounds identified according to this invention as the active ingredient can be administered in a wide variety of therapeutic dosage forms in conventional vehicles for administration.
  • the compounds can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixirs, tinctures, solutions, suspensions, syrups and emulsions, or by injection.
  • they may also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous, topical with or without occlusion, or intramuscular form, all using forms well known to those of ordinary skill in the pharmaceutical arts.
  • compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
  • compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the active agents can be administered concurrently, or they each can be administered at separately staggered times.
  • the dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal, hepatic and cardiovascular function of the patient; and the particular compound thereof employed.
  • a physician or veterinarian of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter or arrest the progress of the condition.
  • Optimal precision in achieving concentrations of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of a drug.
  • a rhesus monkey (M. mulatto) genomic DNA library (Clontech) was screened with a random primed probe using human MC-3R (Gantz, et al., 1993,
  • the receptor plasmid was transfected into CHO dhfr- cells using lipofectamin (GLBCO). Transfected cells were selected by G418 for at least 2-3 weeks before binding and functional assays were performed. To determine the apparent binding affinity of peptides, transfected cells in 12- well plate were washed once with PBS and binding was done in HBSS with 25 mM Hepes, 0.5% NaN 3 , 0.1% BSA, and protease inhibitor cocktail (4 ug/ml leupeptin, 40 ug/ml bacitracin, 5 ug/ml aprotinin, 10 uM phospharamidon and 100 uM AEBSF).
  • stably transfected CHO cells were washed once with PBS without Ca ⁇ /Mg " ⁇ , dissociated by non- enzymatic dissociation medium (Specialty Media, Inc.) at 37°C for 10 min.
  • Dissociated cells were resuspended in EBSS buffer containing 25 mM Hepes, 5 mM MgCl 2 , 0.1% BSA and 50 mM LBMX and incubated with agonist at room temperature for 45 min. The agonist activation was terminated by boiling for 4 min.
  • Intracellular cyclic AMP concentration was measured by the Amersham SPA assay system (Huang, et al., 1997, J. Receptor Signal Transduc. Res. 17:599-607).
  • MT-LI is a partial agonist for both rhesus and human MC-3R.
  • Shu-9119 is shown in this Example Section to be an antagonist for both the rhesus and human MC-3R.
  • Rhesus MC-3R is shown to be pharmacologically similar to human MC-3R with respect to all tested agonists.
  • NDP 0.5 ⁇ 0.1(2) 0.9 ⁇ 0.2(4) 0.1 ⁇ 0.05(3) 0.8 ⁇ 0.2(2) 100 100 ⁇ -MSH 11 ⁇ 5(2) 46 ⁇ 12(3) 1 ⁇ 0.2(3) 4 ⁇ 3(4) 100 100 ⁇ 2-MSH 356 ⁇ 144(2) 105 ⁇ 33(2) 3 ⁇ 1(3) 5 ⁇ 3(4) 100 92

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Toxicology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
EP99971812A 1998-11-09 1999-11-05 Dna moleküle, die das melanocortin 3 rezeptor protein des rhesus affen kodieren Withdrawn EP1129104A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10772598P 1998-11-09 1998-11-09
US107725P 1998-11-09
PCT/US1999/025747 WO2000027862A1 (en) 1998-11-09 1999-11-05 Dna molecules encoding the melanocortin 3 receptor protein from rhesus monkey

Publications (1)

Publication Number Publication Date
EP1129104A1 true EP1129104A1 (de) 2001-09-05

Family

ID=22318131

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99971812A Withdrawn EP1129104A1 (de) 1998-11-09 1999-11-05 Dna moleküle, die das melanocortin 3 rezeptor protein des rhesus affen kodieren

Country Status (4)

Country Link
EP (1) EP1129104A1 (de)
JP (1) JP2002542760A (de)
CA (1) CA2349950A1 (de)
WO (1) WO2000027862A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001273383A1 (en) * 2000-07-10 2002-01-21 Deltagen, Inc. Transgenic mice containing targeted gene disruptions

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5837521A (en) * 1993-04-08 1998-11-17 State Of Oregon Nucleic acids encoding the γ-MSH receptor MC3-R

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0027862A1 *

Also Published As

Publication number Publication date
JP2002542760A (ja) 2002-12-17
WO2000027862A1 (en) 2000-05-18
CA2349950A1 (en) 2000-05-18

Similar Documents

Publication Publication Date Title
WO1999015660A1 (en) G-protein coupled glycoprotein hormone receptor hg38
US6645738B1 (en) DNA molecules encoding the melanocortin 5 receptor protein from rhesus monkey
US7029865B2 (en) DNA molecules encoding the melanocortin 4 receptor protein from rhesus monkey
EP1140968B9 (de) Dna-moleküle kodierende spleissvarianten des humanen melanocortin-1-rezeptorproteins
WO2000027862A1 (en) Dna molecules encoding the melanocortin 3 receptor protein from rhesus monkey
US7060463B2 (en) DNA molecules encoding Macaca mulatta androgen receptor
WO1999015545A1 (en) G-protein coupled glycoprotein hormone receptor aomf05
WO2000031108A1 (en) Dna molecules encoding hg51, a g-protein-coupled receptor
EP1007540A1 (de) Dns moleküle die proteine des menschlichen kernrezeptors kodieren
EP1124844A1 (de) An ein g-protein gekoppelter rezeptor ähnlich dem thrombin-rezeptor
JP2004500023A (ja) ヒトエンドセリン変換酵素3をコードするdna分子
US20030119100A1 (en) DNA molecules encoding human nuclear receptor proteins
CA2315273A1 (en) Dna molecules encoding vertebrate nuclear receptor protein, nnr4
CA2456857A1 (en) Dna encoding rat bombesin receptor subtype-3 (brs-3) and uses thereof

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: 20010611

AK Designated contracting states

Kind code of ref document: A1

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

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: 20040531