WO2001049744A1 - Mouse g-protein coupled receptor mas - Google Patents

Mouse g-protein coupled receptor mas Download PDF

Info

Publication number
WO2001049744A1
WO2001049744A1 PCT/US2001/000054 US0100054W WO0149744A1 WO 2001049744 A1 WO2001049744 A1 WO 2001049744A1 US 0100054 W US0100054 W US 0100054W WO 0149744 A1 WO0149744 A1 WO 0149744A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
seq
sequence
polynucleotide
mas
Prior art date
Application number
PCT/US2001/000054
Other languages
French (fr)
Inventor
Pamela Lane
Ping Tsui
Nabil Elshourbagy
Original Assignee
Smithkline Beecham Corporation
Smithkline Beecham Plc.
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 Smithkline Beecham Corporation, Smithkline Beecham Plc. filed Critical Smithkline Beecham Corporation
Publication of WO2001049744A1 publication Critical patent/WO2001049744A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to newly identified polypeptides and polyn ⁇ cleotides encoding such polypeptides, to their use in identifying compounds that may be agonists and/or antagonists that are potentially useful in therapy, and to production of such polypeptides and polynucleotides.
  • the drug discovery process is currently undergoing a fundamental revolution as it embraces 'functional genomics,' that is, high throughput genome- or gene-based biology. This approach is rapidly superseding earlier approaches based on 'positional cloning'. A phenotype, that is a biological function or genetic disease, would be identified and this would then be tracked back to the responsible gene, based on its genetic map position.
  • G-protein coupled (GPC) receptors such as those for adrenergic agents and dopamme (Kobilka, B.K., et al., Proc. Natl Acad.
  • G-protems themselves, effector proteins, e.g., phosphohpase C, adenyl cyclase, and phosphodiesterase, and actuator proteins, e.g , protein k ase A and protein kmase C (Simon, M.I., et al., Science, 1991, 252:802-8).
  • effector proteins e.g., phosphohpase C, adenyl cyclase, and phosphodiesterase
  • actuator proteins e.g , protein k ase A and protein kmase C (Simon, M.I., et al., Science, 1991, 252:802-8).
  • GTP adenylate cyclase
  • a G-protem connects the hormone receptor to adenylate cyclase.
  • G-protein was shown to exchange GTP for bound GDP when activated by a hormone receptor.
  • the GTP-carrymg form then binds to activated adenylate cyclase.
  • the G-protem serves a dual role, as an intermediate that relays the signal from receptor to effector, and as a clock that controls the duration of the signal.
  • G-protem coupled receptors The membrane protein gene superfamily of G-protem coupled receptors has been characterized as having seven putative transmembrane domains. The domains are believed to represent transmembrane ⁇ -hehces connected by extracellular or cytoplasmic loops.
  • G-protem coupled receptors include a wide range of biologically active receptors, such as hormone, viral, growth factor and neuroreceptors.
  • G-protem coupled receptors (otherwise known as 7TM receptors) have been characterized as including these seven conserved hydrophobic stretches of about 20 to 30 ammo acids, connecting at least eight divergent hydrophihc loops.
  • the G-protein family of coupled receptors includes dopamine receptors which bind to neuroleptic drugs used for treating psychotic and neurological disorders.
  • members of this family include, but are not limited to calcitonm, adrenergic, endothehn, cAMP, adenosme, musca ⁇ nic, acetylchohne, serotonin, histamine, thrombin, kimn, follicle stimulating hormone, opsins, endothehal differentiation gene-1 , rhodopsins, odorant, and cytomegalovirus receptors.
  • G-protein coupled receptors have single conserved cysteme residues in each of the first two extracellular loops which form disulfide bonds that are believed to stabilize functional protein structure.
  • the 7 transmembrane regions are designated as TM1 , TM2, TM3, TM4, TM5, TM6, and TM7.
  • TM3 has been implicated in signal transduction Phosphorylation and hpidation (palmitylation or farnesylation) of cysteme residues can influence signal transduction of some G-protein coupled receptors.
  • Most G-protem coupled receptors contain potential phosphorylation sites within the third cytoplasmic loop and/or the carboxy terminus. For several G-protem coupled receptors, such as the ⁇ -adrenoreceptor, phosphorylation by protein kmase A and/or specific receptor kinases mediates receptor desensitization
  • the ligand binding sites of G-protein coupled receptors are believed to comprise hydrophihc sockets formed by several G-protem coupled receptor transmembrane domains, said sockets bemg surrounded by hydrophobic residues of the G-protem coupled receptors.
  • the hydrophihc side of each G-protem coupled receptor transmembrane helix is postulated to face inward and form a polar ligand binding site.
  • TM3 has been implicated in several G-protein coupled receptors as having a ligand binding site, such as the TM3 aspartate residue TM5 serines, a TM6 asparagine and TM6 or TM7 phenylalanmes or tyrosmes are also implicated in ligand binding
  • G-protem coupled receptor 3 can be mtracellularly coupled by heterot ⁇ me ⁇ c G-protems to various mtracellular enzymes, ion c hannels and transporters (see, Johnson et al., Endoc. Rev , 1989, 10:317-331). Different G-protem ⁇ -subumts preferentially stimulate particular effectors to modulate various biological functions in a cell.
  • G-protein coupled receptors Phosphorylation of cytoplasmic residues of G- 5 protein coupled receptors has been identified as an important mechanism for the regulation of G- protem coupling of some G-protein coupled receptors.
  • G-protein coupled receptors are found in numerous sites within a mammalian host.
  • the present invention relates to Mus musculus mas, in particular Mus musculus mas polypeptides and Mus musculus mas polynucleotides, recombinant materials and methods for their production.
  • the invention relates to methods for identifying agonists and antagonists/inhibitors of the Mus musculus mas gene.
  • This invention further relates to the 15 generation of in vitro and in vivo comparison data relating to the polynucleotides and polypeptides in order to predict oral absorption and pharmacokmetics in man of compounds that either agonize or antagonize the biological activity of such polynucleotides or polypeptides.
  • the present invention further relates to methods for creating transgenic animals, which overexpress or underexpress or have regulatable expression of a mas gene and "knock-out" animals, preferably mice, which an animal no longer expresses a mas gene. Furthermore, this invention relates to transgenic and knock-out animals obtained by using these methods.
  • Such animal models are expected to provide valuable insight into the potential pharmacological and toxicological effects 5 in humans of compounds that are discovered by the aforementioned screening methods as well as other methods
  • An understanding of how a Mus musculus mas gene functions in these animal models is expected to provide an insight into treating and preventing human diseases including, but not limited to.
  • infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV-2; pam, cancers; diabetes, obesity; anorexia: bulimia; asthma; 0 Parkinson's disease; acute heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectons; myocardial infarction; stroke; ulcers; asthma; allergies; benign prostatic hypertrophy; migraine; vomiting; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, depression, delirium, dementia, and severe mental retardation: and dyskmesias, such as Huntmgton's disease or Gilles dela Tourett's syndrome, hereinafter referred to as "the Diseases", amongst others.
  • the Diseases Huntmgton's disease or Gilles dela Tourett's syndrome
  • the present invention relates to Mus musculus mas polypeptides.
  • Such polypeptides include isolated polypeptides comprising an ammo acid sequence having at least a
  • polypeptides include those comprising the amino acid of SEQ ID NO:2
  • variants and fragments thereof and portions of such polypeptides in (a) to (e) that generally contain at least 30 ammo acids, more preferably at least 50 ammo acids, thereof.
  • Polypeptides of the present invention are believed to be members of the mas family of polypeptides. They are. therefore, of interest, because understanding the biological activities of mas in mouse would help to understand the biological function of its human counterpart, human mas, and other related genes.
  • 7TM receptors G-Protem Coupled Receptors
  • the polypeptides of the present invention can be used to establish assays to predict oral absorbtion and pharmacokmetics m man and thus enhance compound and formulation design, among others.
  • a polypeptide of the present invention exhibits at least one biological activity of Mus musculus mas.
  • Polypeptides of the present invention also includes variants of the aforementioned polypeptides, including alleles and splice variants.
  • Such polypeptides vary from the reference polypeptide by insertions, deletions, and substitutions that may be conservative or non-conservative.
  • Particularly preferred variants are those in which several, for instance from 50 to 30, from 30 to 20, from 20 to 10, from 10 to 5, from 5 to 3, from 3 to 2, from 2 to 1 or 1 ammo acids are inserted, substituted, or deleted, in any combination.
  • Preferred fragments of polypeptides of the present invention include an isolated polypeptide comprising an ammo acid sequence having at least 15, 20, 30, 40, 50 or 100 contiguous ammo acids from the ammo acid sequence of SEQ ID NO:2, or an isolated polypeptide comprising an ammo acid sequence having at least 15, 20, 30, 40, 50 or 100 contiguous ammo acids truncated or deleted from the ammo acid sequence of SEQ ID NO:2.
  • biologically active fragments that mediate activities of mas, including those with a similar activity or an improved activity, or with a decreased undesirable activity.
  • Fragments of the polypeptides of the invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, these variants may be employed as intermediates for producing the full-length polypeptides of the invention
  • polypeptides of the present invention may be in the form of a "mature" protein or may be a part of a larger protein such as a fusion protein. It is often advantageous to include an additional ammo acid sequence that contains secretory or leader sequences, pro-sequences, sequences that aid in purification, for instance, multiple histidine residues, or an additional sequence for stability during recombinant production.
  • the present invention also includes variants of the aforementioned polypeptides, that is polypeptides that vary from the referents by conservative ammo acid substitutions, whereby a residue is substituted by another with like characteristics.
  • Typical substitutions are among Ala, Val, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr.
  • Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 ammo acids are substituted, deleted, or added in any combination.
  • Polypeptides of the present invention can be prepared m any suitable manner.
  • Such polypeptides include isolated naturally occurring polypeptides, recomb antly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood m the art.
  • the present invention relates to Mus musculus mas polynucleotides.
  • Such polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide having at least a 95% identity, to the ammo acid sequence of SEQ ID N0:2, over the entire length of SEQ ID NO:2.
  • polypeptides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identity are more highly preferred, and those with at least a 99% identity are most highly preferred.
  • Such polynucleotides include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO: l encoding the polypeptide of SEQ ID NO:2.
  • polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence having at least a 95% identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO:2, over the entire coding region.
  • polynucleotides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identity are more highly preferred, and those with at least a 99% identity are most highly preferred.
  • polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence having at least a 95% identity, to SEQ ID NO: 1 over the entire length of SEQ ID NO: 1.
  • polynucleotides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identify are more highly preferred, and those with at least a 99% identity are most highly preferred.
  • Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO: l , as well as the polynucleotide of SEQ ID NO: 1.
  • the invention also provides polynucleotides that are complementary to all the above described polynucleotides.
  • the nucleotide sequence of SEQ ID NO: 1 shows homology with mouse mas.
  • the nucleotide sequence of SEQ ID NO: 1 is a cDNA sequence and comprises a polypeptide encoding sequence (nucleotide 1 to 975) encoding a polypeptide of 324 ammo acids, the polypeptide of SEQ ID NO:2.
  • the nucleotide sequence encoding the polypeptide of SEQ ID NO:2 may be identical to the polypeptide encoding sequence of SEQ ID NO: 1 or it may be a sequence other than SEQ ID NO: l , which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ID NO:2.
  • the polypeptide of SEQ ID NO:2 is structurally related to other proteins of the mas family, having homology and/or structural similarity with mouse mas.
  • Preferred polypeptides anc polynucleotides of the present invention are expected to have, inter aha, similar biological functionans/properties to their homologous polypeptides and polynucleotides.
  • polypeptides and polynucleotides of the present invention have at least one mas activity.
  • Polynucleotides of the present invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of Mus musculus liver, using the expressed sequence tag (EST) analysis (Adams, M.D., et al. Science (1991) 252: 1651- 1656; Adams, M.D. et al, Nature (1992) 355:632-634; Adams, M.D., et al. Nature (1995) 377 Supp.: 3-174). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesized using well known and commercially available techniques.
  • the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide m reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions.
  • a marker sequence that facilitates purification of the fused polypeptide can be encoded.
  • the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, Inc.) and described in Gentz, et al . Proc Natl Acad ci USA (1989) 86:821- 824, or is an HA tag.
  • the polynucleotide may also comprise non-coding 5' and 3 " sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ⁇ bosome binding sites and sequences that stabilize mRNA.
  • polynucleotides encoding polypeptide variants that comprise the ammo acid sequence of SEQ ID NO:2 and in which several, for instance from 50 to 30, from 30 to 20, from 20 to 10. from 10 to 5, from 5 to 3, from 3 to 2, from 1 to 1 or 1 ammo acid residues are substituted, deleted or added, in any combination.
  • Particularly preferred probes will have between 30 and 50 nucleotides, but may have between 100 and 200 contiguous nucleotides of the polynucleotide of SEQ ID NO. l .
  • a preferred embodiment of the invention is a polynucleotide of consisting of or comprising nucleotide ATG to the nucleotide immediately upstream of or including nucleotide TGA set forth in SEQ ID NO: 1 , both of which encode a mas polypeptide.
  • the invention also includes a polynucleotide consisting of or comprising a polynucleotide of the formula: X-(R 1 ) m -(R 2 )-(R 3 ) n -Y wherein, at the 5' end of the molecule, X is hydrogen, a metal or a modified nucleotide residue, or together with Y defines a covalent bond, and at the 3' end of the molecule, Y is hydrogen, a metal, or a modified nucleotide residue, or together with X defines the covalent bond, each occurrence of R j and R3 is independently any nucleic acid residue or modified nucleic acid residue, m is an integer between 1 and 3000 or zero, n is an integer between 1 and 3000 or zero, and R 2 is a nucleic acid sequence or modified nucleic acid sequence of the invention, particularly the nucleic acid sequence set forth in SEQ ID NO: l or a modified nucleic acid sequence thereof.
  • R 2 is oriented so that its 5' end nucleic acid residue is at the left, bound to R ] and its 3' end nucleic acid residue is at the right, bound to R3.
  • Any stretch of nucleic acid residues denoted by either R j and/or R 2 , where m and/or n is greater than 1, may be either a heteropolymer or a homopolymer, preferably a heteropolymer.
  • the polynucleotide of the above formula is a closed, circular polynucleotide, which can be a double-stranded polynucleotide wherein the formula shows a first strand to which the second strand is complementary.
  • m and/or n is an integer between 1 and 1000.
  • Other preferred embodiments of the invention are provided where m is an integer between 1 and 50, 100 or 500, and n is an integer between 1 and 50, 100, or 500.
  • Polynucleotides that are identical, or are substantially identical to a nucleotide sequence of SEQ ID NO: 1 may be used as hybridization probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides of the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding homologs and orthologs from species other than Mus musculus) that have a high sequence identity to SEQ ID NO.1. Typically these nucleotide sequences are 95% identical to that of the referent.
  • Preferred probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides, and may even have at least 100 nucleotides. Particularly preferred primers will have between 20 and 25 nucleotides.
  • a polynucleotide encoding a polypeptide of the present invention, mcludmg homologs and orthologs from a species other than Mus musculus may be obtained by a process comprising the steps of screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: l or a fragment thereof, preferably of at least 15 nucleotides in length; and isolating full-length cDNA and genomic clones comprising said polynucleotide sequence.
  • Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM t ⁇ sodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters m O.l x SSC at about 65°C.
  • the present invention also includes isolated polynucleotides, preferably of at least 100 nucleotides in length, obtained by screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: l or a fragment thereof, preferably of at least 15 nucleotides.
  • an isolated cDNA sequence will be incomplete, that the region coding for the polypeptide is cut short at the 5' end of the cDNA. This is a consequence of reverse transc ⁇ ptase, an enzyme with inherently low 'processivity' (a measure of the ability of the enzyme to remain attached to the template during the polymerization reaction), failing to complete a DNA copy of the mRNA template during 1 st strand cDNA synthesis.
  • PCR Nucleic acid amplification
  • Recombmant polypeptides of the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, m a further aspect, the present invention relates to expression systems comprising a polynucleotide or polynucleotides of the present invention, to host cells which are genetically engmeered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
  • host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention.
  • Introduction of polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis, et al, BASIC METHODS IN MOLECULAR BIOLOGY (1986) and Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
  • Preferred methods of introducing polynucleotides into host cells include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, micro jection, catiomc hpid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection
  • bacterial cells such as streptococci, staphylococci, E coh, Streptomyces and Bacillus subtihs cells
  • fungal cells such as yeast cells and Aspergillus cells
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells
  • plant cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells
  • chromosomal, episomal and virus-derived systems e.g , vectors derived from bacterial plasmids, from bacte ⁇ ophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacte ⁇ ophage genetic elements, such as cosmids and phagemids.
  • the expression systems may comprise control regions that regulate as well as engender expression
  • any system or vector that is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used
  • the appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook, et al, MOLECULAR CLONING, A LABORATORY MANUAL (supra).
  • a polypeptide of the present invention is to be expressed for use in screening assays, it is generally preferred that the polypeptide be produced at the surface of the cell. In this event, the cells may be harvested prior to use in the screening assay. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the polypeptide.
  • Polypeptides of the presen invention can be recovered and purified from recombinant cell cultures by well-known methods lr eluding ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectm chromatography. Most preferably, high performance liquid chromatography is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
  • the polynucleotide sequences of the present invention are also valuable for chromosome localization studies.
  • the polynucleotide sequence, or fragment(s) thereof, is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome.
  • the mapping of these sequences to human chromosomes according to the present invention is an important first step in correlating homologous human polynucleotide sequences with gene associated disease in humans.
  • Precise chromosomal localizations for a polynucleotide sequence can be determined using Radiation Hybrid (RH) Mapping (Walter, M., et al (1994) Nature Genetics 7, 22-28), for example.
  • RH Radiation Hybrid
  • a number of RH panels are available, including mouse, rat, baboon, zebrafish and human.
  • RH mapping panels are available from a number of sources, for example Research Genetics (Huntsville, AL, USA).
  • PCR reactions are performed using primers, designed to the polynucleotide sequence of interest, on the RH DNAs of the panel.
  • Each of these DNAs contains random genomic fragments from the species of interest.
  • These PCRs result in a number of scores, one for each RH DNA m the panel, indicating the presence or absence of the PCR product of the polynucleotide sequence of interest
  • These scores are compared with scores created using PCR products from genomic sequences of known location, usually using an on-line resource such as that available at the Whitehead Institute for Biomedical Research in Cambridge, Massachusetts, USA website (http:// www. genome wi.mit.edu/).
  • Mus musculus mas gene products can be expressed in transgenic animals. Animals of any species, including, but not limited to: mice, rats, rabbits, guinea pigs, dogs, cats, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, chimpanzees, may be used to generate mas transgenic animals.
  • This invention further relates to a method of producing transgenic animals, preferably Mus musculus, over-expressing mas, which method may comprise the introduction of several copies of a segment comprising at least the polynucleotide sequence encoding SEQ ID NO:2 with a suitable promotor into the cells of a Mus musculus embryo, or the cells of another species, at an early stage
  • This invention further relates to a method of producing transgenic animals, preferably Mus musculus, under-expressing or regulatably expressing mas, which method may comprise the introduction of a weak promoter or a regulatable promoter (e g. , an mducible or repressible promoter) respectively, expressibly linked to the polynucleotide sequence of SEQ ID NO: l into the cells of a Mus musculus embryo at an early stage
  • a weak promoter or a regulatable promoter e g. , an mducible or repressible promoter
  • This invention also relates to transgenic animals, characterized in that they are obtained by a method, as defined above Any technique known in the art may be used to introduce a Mus musculus mas transgene into animals to produce a founder line of animals. Such techniques include, but are not limited to pronuclear micromjection (U.S. Patent No. 4,873, 191 ); retrovirus mediated gene transfer into germ lines (Van der Putten, et al . Proc Natl Acad Sci , USA 82. 6148-6152 ( 1985); gene targeting m embryonic stem cells (Thompson, et al , Cell 56: 313-321 (1989), electropolation of embryos (Lo, Mol.
  • a further aspect of the present invention involves gene targeting by homologous recombination in embryonic stem cells to produce a transgenic animal with a mutation in a mas gene ("knock-out" mutation).
  • knock-out animals there is mactivation of the mas gene or altered gene expression, such that the animals are useful to study the function of the mas gene, thus providing animals models of human disease, which are otherwise not readily available through spontaneous, chemical or irradiation mutagenesis.
  • Another aspect of the present invention involves the generation of so-called "knock-m” animals in which a portion of a wild-type gene is fused to the cDNA of a heterologous gene.
  • This invention further relates to a method of producing "knock-out" animals, preferably 5 mice, no longer expressing mas.
  • a Mus musculus mas cDNA SEQ ID NO. l
  • SEQ ID NO. l a Mus musculus mas cDNA
  • the method used to create a knockout mouse is characterized in that: a suitable mutation is produced in the polynucleotide sequence of the murme mas genomic 10 clone, which inhibits the expression of a gene encoding murme mas, or inhibits the activity of the gene product, said modified murme mas polynucleotide is introduced into a homologous segment of murme genomic DNA, combined with an appropriate marker, so as to obtain a labeled sequence comprising said modified murme genomic DNA; i s said modified murme genomic DNA comprising the modified polynucleotide is transfected mto embryonic stem cells and correctly targeted events selected in vitro: then said stem cells are remjected into a mouse embryo; then said embryo is implanted into a female recipient and brought to term as a chimera which transmits said mutation through the germhne; and
  • mice 20 homozygous recombinant mice are obtained at the F2 generation which are recognizable by the presence of the marker.
  • a mutation is generated in a murme mas allele by the introduction of a DNA construct comprising DNA of a gene encoding murme mas, which murme
  • the mutation is targeted to the allele by way of the DNA construct
  • the DNA of the gene encoding mur e mas comprised in the construct may be foreign to the species of which the recipient is a member, may be native to the species and foreign only to the individual recipient, may be a construct comprised of synthetic or natural genetic components, or a mixture of these
  • the mutation may constitute an insertion, deletion, substitution, or combination 0 thereof.
  • the DNA construct can be introduced into cells by, for example, calcium-phosphate DNA co-precipitation It is preferred that a mutation be introduced into cells using electroporation, micromjection, virus infection, hgand-DNA conjugation, virus-hgand-DNA conjugation, or hposomes.
  • Another embodiment of the instant invention relates to "knock-out" animals, preferably mice, obtained by a method of producing recombinant mice as defined above, among others.
  • Another aspect of this invention provides for in vitro mas "knock-outs", i.e., tissue cultures
  • mice Animals of any species, including, but not limited to: mice, rats, rabbits, guinea pigs, dogs, cats, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, chimpanzees, may be used to generate in vitro mas "knock-outs". Methods for "knocking out” genes in vitro are described in Galh-Tahadoros, et al , Journal of Immunological Methods 181 : 1 -15 (1995). Transgenic, "knock-m", and “knock-out” animals, as defined above, are a particularly advantageous model, from a physiological point of view, for studying mas.
  • Such animals will be valuable tools to study the functions of a mas gene. Moreover, such animal models are expected to provide information about potential toxicological effects in humans of any compounds discovered by an aforementioned screening method, among others.
  • An understanding of how a Mus musculus mas gene functions in these animal models is expected to provide an insight into treating and preventing human diseases including, but not limited to: infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV -2; pam; cancers; diabetes, obesity; anorexia; bulimia; asthma; Parkinson's disease; acute heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectons; myocardial infarction; stroke; ulcers; asthma; allergies; bemgn prostatic hypertrophy; migraine; vomiting; psychotic and neurological disorders, including anxiety, schizophrenia, mamc depression, depression, delirium, dementia, and severe mental retardation; and dyskmesias, such as Huntmgton's disease or Gilles
  • Polypeptides of the present invention are responsible for many biological functions, including many disease states, m particular the Diseases mentioned herein. It is, therefore, an aspect of the invention to devise screening methods to identify compounds that stimulate (agonists) or that inhibit (antagonists) the function of the polypeptide, such as agonists, antagonists and inhibitors. Accordingly, m a further aspect, the present invention provides for a method of screening compounds to identify those that stimulate or inhibit the function of the polypeptide. In general, agonists or antagonists may be employed for therapeutic and prophylactic purposes for the Diseases mentioned herein mentioned. Compounds may be identified from a variety of sources, for example, cells, cell-free preparations, chemical libraries, and natural product mixtures.
  • Such agonists and antagonists so-identified may be natural or modified substrates, hgands, receptors, enzymes, etc., as the case may be, o the polypeptide; or may be structural or functional mimetics thereof (see Cohgan, et al , CURRENT PROTOCOLS IN IMMUNOLOGY 1(2): Chapter 5 (1991)).
  • the screening method may simply measure the binding of a candidate compound to the polypeptide, or to cells or membranes bearing the polypeptide, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound.
  • a screening method may involve measuring or, qualitatively or quantitatively, detecting the competition of binding of a candidate compound to the polypeptide with a labeled competitor (e.g., agonist or antagonist).
  • screening methods may test whether the candidate compound results in a signal generated by an agonist or antagonist of the polypeptide, using detection systems appropriate to cells bearing the polypeptide.
  • Antagonists are generally assayed in the presence of a known agonist and an effect on activation by the agonist by the presence of the candidate compound is observed.
  • screening methods may simply comprise the steps of mixing a candidate compound with a solution comprising a polypeptide of the present invention, to form a mixture, measuring Mus musculus mas activity m the mixture, and comparing a Mus musculus mas activity of the mixture to a control mixture which contains no candidate compound.
  • Polypeptides of the present invention may be employed in conventional low capacity screening methods and also in high-throughput screening (HTS) formats.
  • HTS formats include not only the well-established use of 96- and, more recently, 384-well microtiter plates but also emerging methods such as the nanowell method described by Schullek, et al , Anal Biochem., 246, 20-29, (1997).
  • Fusion proteins such as those made from Fc portion and Mus musculus mas polypeptide, as herein desc ⁇ bed, can also be used for high-throughput screening assays to identify antagonists of antagonists of the polypeptide of the present invention (see D. Bennett, et al. J Mol Recognition, 8 52-58 (1995), and K. Johanson, et al , J Bwl Chem., 270(16):9459-9471 (1995)).
  • One screening technique includes the use of cells which express the receptor of this invention (for example, transfected CHO cells) in a system which measures extracellular pH or intracellular calcium changes caused by receptor activation.
  • compounds may be contacted with cells expressing the receptor polypeptide of the present invention
  • a second messenger response e.g., signal transduction, pH changes, or changes m calcium level, is then measured to determine whether the potential compound activates or inhibits the receptor.
  • Another method involves screening for receptor inhibitors by determining inhibition or stimulation of receptor-mediated cAMP and/or adenylate cyclase accumulation.
  • Such a method mvolves transfecting a eukaryotic cell with the receptor of this invention to express the receptor on the cell surface. The cell is then exposed to potential antagonists in the presence of the receptor of this invention. The amount of cAMP accumulation is then measured. If the potential antagonist binds the receptor, and thus inhibits receptor binding, the levels of receptor-mediated cAMP, or adenylate cyclase, activity will be reduced or increased.
  • Another methods for detecting agonists or antagonists for the receptor of the present invention is the yeast based technology as described in U.S. Patent No. 5,482,835.
  • polypeptide antagonists include antibodies or, in some cases, ohgopeptides or proteins that are closely related to ligands, substrates, receptors, enzymes, etc , as the case may be, of a mas polypeptide, e g., a fragment of a ligand, substrate, receptor, enzyme, etc., or small molecules which bind to a mas polypeptide but do not elicit a response, so that an activity of a mas polypeptide is prevented
  • the present invention relates to a screening kit for identifying agonists, antagonists, inhibitors, ligands, receptors, substrates, enzymes, etc for polypeptides of the present invention; or compounds which decrease or enhance the production of such polypeptides, which compounds comprise a member selected from the group consisting of.
  • polypeptide of the present invention (c) a cell membrane expressing a polypeptide of the present invention; which polypeptide is preferably that of SEQ ID NO.2
  • kits may comprise a substantial component.
  • polypeptide of the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by.
  • the present invention relates to the use of Mus musculus mas polypeptides, polynucleotides, and recombinant materials thereof in selection screens to identify compounds which are neither agonists nor antagonist/inhibitors of Mus musculus mas.
  • the data from such a selection screen is expected to provide in vitro and in vivo comparisons and to predict oral absorption, pharmacokmetics in humans. The ability to make such a comparison of data will enhance formulation design through the identification of compounds with optimal development characteristics, i.e . high oral bioavailabihty, UID (once a day) dosmg, reduced drug interactions, reduced variability, and reduced food effects, among others.
  • Allele refers to one or more alternative forms of a gene occurring at a given locus in the genome.
  • “Fragment” of a polypeptide sequence refers to a polypeptide sequence that is shorter than the reference sequence but that retains essentially the same biological function or activity as the reference polypeptide.
  • “Fragment” of a polynucleotide sequence refers to a polynucleotide sequence that is shorter than the reference sequence of SEQ ID NO: l .
  • Fusion protein refers to a protein encoded by two, often unrelated, fused genes or fragments thereof.
  • EP-A-0 464 discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • employing an immunoglobulin Fc region as a part of a fusion protein is advantageous for use m therapy and diagnosis resulting m, for example, improved pharmacokmetic properties [see, e.g , EP-A 0232 262].
  • “Homolog” is a generic term used in the art to indicate a polynucleotide or polypeptide sequence possessing a high degree of sequence relatedness to a reference sequence. Such relatedness may be quantified by determining the degree of identity and/or similarity between the two sequences as hereinbefore defined. Falling withm this generic term are the terms, "ortholog", and “paralog”. "Ortholog” refers to polynucleotides/genes or polypeptide that are homologs via speciation. that is closely related and assumed to have commend descent based on structural and functional considerations. "Paralog” refers to polynucleotides/genes or polypeptide that are homologs via gene duplication, for instance, duplicated variants withm a genome.
  • Identity reflects a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, determined by comparing the sequences.
  • identity refers to an exact nucleotide to nucleotide or ammo acid to ammo acid correspondence of the two polynucleotide or two polypeptide sequences, respectively, over the length of the sequences being compared.
  • a "% identity” may be determined.
  • the two sequences to be compared are aligned to give a maximum correlation between the sequences. This may include inserting "gaps" m either one or both sequences, to enhance the degree of alignment.
  • a % identity may be determined over the whole length of each of the sequences bemg compared (so-called global alignment), that is particularly suitable for sequences of the same or very similar length, or over shorter, defined lengths (so-called local alignment), that is more suitable for sequences of unequal length.
  • Similarity is a further, more sophisticated measure of the relationship between two polypeptide sequences.
  • similarity means a comparison between the amino acids of two polypeptide chains, on a residue by residue basis, taking into account not only exact correspondences between a between pairs of residues, one from each of the sequences being compared (as for identity) but also, where there is not an exact correspondence, whether, on an evolutionary basis, one residue is a likely substitute for the other. This likelihood has an associated 'score' from which the "%. similarity" of the two sequences can then be determined.
  • BESTFIT is more suited to comparing two polynucleotide or two polypeptide sequences that are dissimilar in length, the program assuming that the shorter sequence represents a portion of the longer.
  • GAP aligns two sequences, finding a "maximum similarity", according to the algorithm of Neddleman and Wunsch (J Mo I Biol , 48, 443-453, 1970).
  • GAP is more suited to comparing sequences that are approximately the same length and an alignment is expected over the entire length.
  • the parameters " 3ap Weight” and "Length Weight” used m each program are 50 and 3, for polynucleotide sequences and 12 and 4 for polypeptide sequences, respectively.
  • % identities and similarities are determined when the two sequences bemg compared are optimally aligned.
  • Other programs for determining identity and/or similarity between sequences are also known in the art, for instance the BLAST family of programs (Altschul S.F., et al , J.
  • the BLOSUM62 ammo acid substitution matrix (Hemkoff S. and Hemkoff J G , Proc Nat Acad Sci USA, 89. 10915-10919 (1992)) is used in polypeptide sequence comparisons including where nucleotide sequences are first translated mto ammo acid sequences before comparison.
  • the program BESTFIT is used to determine the % identity of a query polynucleotide or a polypeptide sequence with respect to a polynucleotide or a polypeptide sequence of the present invention, the query and the reference sequence being optimally aligned and the parameters of the program set at the default value, as hereinbefore described
  • a polynucleotide sequence having, for example, at least 95% identity to a reference polynucleotide sequence is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference sequence Such point mutations are selected from the group consisting of at least one nucleotide deletion, substitution, mcludmg transition and transversion, or insertion.
  • point mutations may occur at the 5' or 3' terminal positions of the reference polynucleotide sequence or anywhere between these terminal positions, interspersed either individually among the nucleotides m the reference sequence or in one or more contiguous groups withm the reference sequence.
  • a polynucleotide sequence having at least 95% identity to a reference polynucleotide sequence up to 5% of the nucleotides of the in the reference sequence may be deleted, substituted or inserted, or any combination thereof, as hereinbefore described.
  • % identities such as 96%, 97%, 98%, 99% and 100%.
  • a polypeptide sequence having, for example, at least 95% identity to a reference polypeptide sequence is identical to the reference sequence except that the polypeptide sequence may include up to five point mutations per each 100 ammo acids of the reference sequence.
  • Such point mutations are selected from the group consisting of at least one ammo acid deletion, substitution, including conservative and non-conservative substitution, or insertion. These point mutations may occur at the ammo- or carboxy- terminal positions of the reference polypeptide sequence or anywhere between these terminal positions, interspersed either individually among the ammo acids in the reference sequence or m one or more contiguous groups within the reference sequence.
  • a sequence polypeptide sequence having at least 95% identity to a reference polypeptide sequence up to 5% of the ammo acids of the m the reference sequence may be deleted, substituted or inserted, or any combination thereof, as hereinbefore desc ⁇ bed.
  • % identities such as 96%, 97%, 98%, 99%, and 100%.
  • Polynucleotide embodiments further include an isolated polynucleotide comprising a polynucleotide sequence having at least a 95, 97 or 100% identity to the reference sequence of SEQ ID NO: l , wherein said polynucleotide sequence may be identical to the reference sequence of SEQ ID NO: l or may include up to a certain integer number of nucleotide alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or m one or more contiguous groups within the reference sequence, and wherein said number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ ID NO: l by the integer defining the percent identity
  • n n is the number of nucleotide alterations
  • x n is the total number of nucleotides in SEQ ID NO: 1
  • y is 0 95 for 95%. 0.97 for 97% or 1 00 for 100%.
  • is the symbol for the multiplication operator, and wherein any non-mteger product of x n and y is rounded down to the nearest integer prior to subtracting it from x n .
  • Alterations of a polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 may create nonsense, missense or frameshift mutations m this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
  • Polypeptide embodiments further include an isolated polypeptide comprising a polypeptide having at least a 95, 97 or 100% identity to a polypeptide reference sequence of SEQ ID NO:2, wherein said polypeptide sequence may be identical to the reference sequence of SEQ ID NO:2 or may include up to a certain integer number of amino acid alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the ammo- or carboxy-termmal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the ammo acids in the reference sequence or in one or more contiguous groups withm the reference sequence, and wherein said number of ammo acid alterations is determined by multiplying the total number of ammo acids m SEQ ID NO:2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of ammo acids in SEQ ID
  • n a is the number of ammo acid alterations
  • x a is the total number of amino acids in SEQ ID NO:2
  • y is 0.95 for 95%, 0.97 for 97% or 1.00 for 100%
  • is the symbol for the multiplication operator, and wherein any non-mteger product of x a and y is rounded down to the nearest integer prior to subtracting it from x a .
  • isolated means altered “by the hand of man” from its natural state, / e , if it occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living organism is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated”, as the term is employed herein.
  • a polynucleotide or polypeptide that is introduced into an organism by transformation, genetic manipulation or by any other recombinant method is "isolated” even if it is still present in said organism, which organism may be living or non-living.
  • “Knock-in” refers to the fusion of a portion of a wild-type gene to the cDNA of a heterologous gene
  • “Knock-out” refers to partial or complete suppression of the expression of a protein encoded by an endogenous DNA sequence m a cell.
  • the “knock-out” can be affected by targeted deletion of the whole or part of a gene encoding a protein, in an embryonic stem cell. As a result, the deletion may prevent or reduce the expression of the protein in any cell in the whole animal in which it is normally expressed.
  • RNA Variant refers to cDNA molecules produced from RNA molecules initially transcribed from the same genomic DNA sequence but which have undergone alternative RNA splicing.
  • Alternative RNA splicing occurs when a primary RNA transcript undergoes splicing, generally for the removal of introns, which results m the production of more than one mRNA molecule each of that may encode different ammo acid sequences.
  • the term splice variant also refers to the proteins encoded by the above cDNA molecules.
  • Transgenic animal refers to an animal to which exogenous DNA has been introduced while the animal is still in its embryonic stage. In most cases, the transgenic approach aims at specific modifications of the genome, e g , by introducing whole transc ⁇ ptional units into the genome, or by up- or down-regulat g pre-existing cellular genes. The targeted character of certain of these procedures sets transgenic technologies apart from experimental methods which random mutations are conferred to the germhne, such as administration of chemical mutagens or treatment with ionizing solution.
  • Polynucleotide generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • Polynucleotides include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that may be smgle-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA
  • polynucleotide also includes DNAs or RNAs comprising one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons.
  • Modified bases include, for example, tritylated bases and unusual bases such as inosine.
  • polynucleotide embraces chemically, enzy atically or metabohcally modified forms of polynucleotides as typically found in nature, as well as the chemical forms of DNA and RNA characteristic of viruses and cells.
  • Polynucleotide also embraces relatively short polynucleotides, often referred to as ohgonucleotides.
  • Polypeptide refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, / e , peptide isosteres.
  • Polypeptide refers to both short chains, commonly referred to as peptides, ohgopeptides or ohgomers, and to longer chains, generally referred to as proteins. Polypeptides may comprise ammo acids other than the 20 gene-encoded amino acids.
  • Polypeptides include amino acid sequences modified either by natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well desc ⁇ bed m basic texts and in more detailed monographs, as well as m a voluminous research literature. Modifications may occur anywhere m a polypeptide, including the peptide backbone, the ammo acid side-chams and the amino or carboxyl terrnim. It will be appreciated that the same type of modification may be present to the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may comprise many types of modifications.
  • Polypeptides may be branched as a result of ubiquitmation, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from post-translation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP- ⁇ bosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a hpid or hpid derivative, covalent attachment of phosphotidyhnositol, cross-linking, cychzation, disulfide bond formation, demethylation, formation of covalent cross-lmks, formation of cysteme, formation of pyroglutamate, formylation, gamma- carboxylation, glycosylation, GPI anchor formation, hydroxylation, lodination, methylation, my ⁇ stoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as argmylation, and ubiquitmation (see, for instance, PROTE
  • Variant refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, but retains essential properties.
  • a typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the am o acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result m ammo acid substitutions, additions, deletions, fusions and truncations m the polypeptide encoded by the reference sequence, as discussed below.
  • a typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical.
  • a variant and reference polypeptide may differ m ammo acid sequence by one or more substitutions, additions, deletions in any combination.
  • a substituted or inserted ammo acid residue may or may not be one encoded by the genetic code.
  • a variant of a polynucleotide or polypeptide may be a naturally occurring such as an allehc variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
  • Example 1 Mammalian Cell Expression
  • the receptors of the present invention are expressed in either human embryonic kidney 293 (HEK293) cells or adherent dhfr CHO cells.
  • HEK293 human embryonic kidney 293
  • adherent dhfr CHO cells typically all 5" and 3 * untranslated regions (UTRs) are removed from the receptor cD A prior to insertion into a pCDN or pCDNA3 vector.
  • the cells are transfected with individual receptor cDNAs by lipofectm and selected in the presence of 400 mg/ml G418. After 3 weeks of selection, individual clones are picked and expanded for further analysis.
  • HEK293 or CHO cells transfected with the vector alone serve as negative controls.
  • To isolate cell lines stably expressing the individual receptors about 24 clones are typically selected and analyzed by Northern blot analysis. Receptor mRNAs are generally detectable m about 50% of the G418-res ⁇ stant clones analyzed.
  • a bank of over 600 putative receptor ligands has been assembled for screening.
  • the bank comp ⁇ ses: transmitters, hormones and chemokines known to act via a human seven transmembrane (7TM) receptor; naturally occurring compounds which may be putative agonists for a human 7TM receptor, non-mammalian, biologically active peptides for which a mammalian counterpart has not yet been identified; and compounds not found m nature, but which activate 7TM receptors with unknown natural ligands.
  • This bank is used to initially screen the receptor for known ligands, using both functional (i.e . calcium, cAMP, microphysiometcr, oocyte electrophysiology, etc, see below) as well as binding assays.
  • Ligand binding assays provide a direct method for ascertaining receptor pharmacology and are adaptable to a high throughput format.
  • the purified ligand for a receptor is radiolabeled to high specific activity (50-2000 Ci/mmol) for binding studies. A determination is then made that the process of radiolabelmg does not dimmish the activity of the ligand towards its receptor.
  • Assay conditions for buffers, ions, pH and other modulators such as nucleotides are optimized to establish a workable signal to noise ratio for both membrane and whole cell receptor sources.
  • specific receptor binding is defined as total associated radioactivity minus the radioactivity measured in the presence of an excess of unlabeled competing ligand. Where possible, more than one competing ligand is used to define residual nonspecific binding.
  • RNA transcripts from linearized plasmid templates encoding the receptor cDNAs of the invention are synthesized in vitro with RNA polymerases in accordance with standard procedures. In vitro transcripts are suspended in water at a final concentration of 0.2 mg/ml. Ovarian lobes are removed from adult female toads, Stage V defolhculated oocytes are obtained, and RNA transcripts ( 10 ng/oocyte) are injected in a 50 nl bolus using a microinjection apparatus. Two electrode voltage clamps are used to measure the currents from individual Xenopus oocytes m response to agonist exposure. Recordings are made in Ca2+ free Barth's medium at room temperature. The Xenopus system can be used to screen known ligands and tissue/cell extracts for activating ligands. Example 5. Microphysiomet ⁇ c Assays
  • Activation of a wide variety of secondary messenger systems results m extrusion of small amounts of acid from a cell.
  • the acid formed is largely as a result of the increased metabolic activity required to fuel the mtracellular signaling process.
  • the pH changes in the media surrounding the cell are very small but are detectable by the CYTOSENSOR rmcrophysiometer (Molecular Devices Ltd., Menlo Park, CA).
  • the CYTOSENSOR is thus capable of detecting the activation of a receptor which is coupled to an energy utilizing mtracellular signaling pathway such as the G-protem coupled receptor of the present invention
  • the 7TM receptor of the invention is also functionally screened (using calcium, cAMP, microphysiometer, oocyte electrophysiology, etc., functional screens) against tissue extracts to identify natural ligands. Extracts that produce positive functional responses can be sequentially subfractionated until an activating ligand is isolated identified.
  • HEK 293 cells expressing recombinant receptors are loaded with fura 2 and in a single day > 150 selected ligands or tissue/cell extracts are evaluated for agonist induced calcium mobilization Similarly, HEK 293 cells expressing recombinant receptors are evaluated for the stimulation or inhibition of c AMP production using standard cAMP quantitation assays . Agonists presenting a calcium transient or cAMP fluctuation are tested in vector control cells to determine if the response is unique to the transfected cells expressing receptor.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Mus musculus mas polypeptides and polynucleotides and method for producing such polypeptides by recombinant techniques are disclosed. Also disclosed are methods for screening for compounds that either agonize or antagonize Mus musculus mas. Such compounds are expected to be useful in treatment of human diseases, including, but not limited to: infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV-2; pains; cancers; diabetes, obesity; anorexia; bulimia, asthma; Parkinson's disease; acute heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectoris; myocardial infarction; stroke; ulcers; asthma; allergies; benign prostatic hypertrophy; migraine; vomiting; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, depression, delirium, dementia, and severe mental retardation; and dyskinesias, such as Huntington's disease or Gilles dela Tourett's syndrome.

Description

Mouse G-Protein Coupled Receptor Mas
Field of the Invention
This invention relates to newly identified polypeptides and polynμcleotides encoding such polypeptides, to their use in identifying compounds that may be agonists and/or antagonists that are potentially useful in therapy, and to production of such polypeptides and polynucleotides.
Background of the Invention
The drug discovery process is currently undergoing a fundamental revolution as it embraces 'functional genomics,' that is, high throughput genome- or gene-based biology. This approach is rapidly superseding earlier approaches based on 'positional cloning'. A phenotype, that is a biological function or genetic disease, would be identified and this would then be tracked back to the responsible gene, based on its genetic map position.
Functional genomics relies heavily on the various tools of bioinformatics to identify gene sequences of potential interest from the many molecular biology databases now available There is a continuing need to identify and characterize further genes and their related polypeptides/proteins, as targets for drug discovery.
It is well established that many medically significant biological processes are mediated by proteins participating m signal transduction pathways that involve G-protems and/or second messengers, e.g., cAMP (Lefkowitz, Nature, 1991, 351 :353-354) Herein these proteins are referred to as proteins participating in pathways with G-protems or PPG proteins. Some examples of these proteins include the G-protein coupled (GPC) receptors, such as those for adrenergic agents and dopamme (Kobilka, B.K., et al., Proc. Natl Acad. Sci., USA, 1987, 84:46-50; Kobilka, B K , et al , Science, 1987, 238:650-656; Bunzow, J.R., et al, Nature, 1988, 336:783-787), G-protems themselves, effector proteins, e.g., phosphohpase C, adenyl cyclase, and phosphodiesterase, and actuator proteins, e.g , protein k ase A and protein kmase C (Simon, M.I., et al., Science, 1991, 252:802-8).
For example, m one form of signal transduction, the effect of hormone binding is activation of the enzyme, adenylate cyclase, inside the cell. Enzyme activation by hormones is dependent on the presence of the nucleotide, GTP. GTP also influences hormone binding A G-protem connects the hormone receptor to adenylate cyclase. G-protein was shown to exchange GTP for bound GDP when activated by a hormone receptor. The GTP-carrymg form then binds to activated adenylate cyclase. Hydrolysis of GTP to GDP, catalyzed by the G-protem itself, returns the G-protem to its basal, inactive form. Thus, the G-protem serves a dual role, as an intermediate that relays the signal from receptor to effector, and as a clock that controls the duration of the signal.
The membrane protein gene superfamily of G-protem coupled receptors has been characterized as having seven putative transmembrane domains. The domains are believed to represent transmembrane α-hehces connected by extracellular or cytoplasmic loops. G-protem coupled receptors include a wide range of biologically active receptors, such as hormone, viral, growth factor and neuroreceptors.
G-protem coupled receptors (otherwise known as 7TM receptors) have been characterized as including these seven conserved hydrophobic stretches of about 20 to 30 ammo acids, connecting at least eight divergent hydrophihc loops. The G-protein family of coupled receptors includes dopamine receptors which bind to neuroleptic drugs used for treating psychotic and neurological disorders. Other examples of members of this family include, but are not limited to calcitonm, adrenergic, endothehn, cAMP, adenosme, muscaπnic, acetylchohne, serotonin, histamine, thrombin, kimn, follicle stimulating hormone, opsins, endothehal differentiation gene-1 , rhodopsins, odorant, and cytomegalovirus receptors.
Most G-protein coupled receptors have single conserved cysteme residues in each of the first two extracellular loops which form disulfide bonds that are believed to stabilize functional protein structure. The 7 transmembrane regions are designated as TM1 , TM2, TM3, TM4, TM5, TM6, and TM7. TM3 has been implicated in signal transduction Phosphorylation and hpidation (palmitylation or farnesylation) of cysteme residues can influence signal transduction of some G-protein coupled receptors. Most G-protem coupled receptors contain potential phosphorylation sites within the third cytoplasmic loop and/or the carboxy terminus. For several G-protem coupled receptors, such as the β-adrenoreceptor, phosphorylation by protein kmase A and/or specific receptor kinases mediates receptor desensitization
For some receptors, the ligand binding sites of G-protein coupled receptors are believed to comprise hydrophihc sockets formed by several G-protem coupled receptor transmembrane domains, said sockets bemg surrounded by hydrophobic residues of the G-protem coupled receptors. The hydrophihc side of each G-protem coupled receptor transmembrane helix is postulated to face inward and form a polar ligand binding site. TM3 has been implicated in several G-protein coupled receptors as having a ligand binding site, such as the TM3 aspartate residue TM5 serines, a TM6 asparagine and TM6 or TM7 phenylalanmes or tyrosmes are also implicated in ligand binding G-protem coupled receptor 3 can be mtracellularly coupled by heterotπmeπc G-protems to various mtracellular enzymes, ion c hannels and transporters (see, Johnson et al., Endoc. Rev , 1989, 10:317-331). Different G-protem α-subumts preferentially stimulate particular effectors to modulate various biological functions in a cell. Phosphorylation of cytoplasmic residues of G- 5 protein coupled receptors has been identified as an important mechanism for the regulation of G- protem coupling of some G-protein coupled receptors. G-protein coupled receptors are found in numerous sites within a mammalian host.
Over the past 15 years, nearly 350 therapeutic agents targeting 7 transmembrane (7 TM) receptors have been successfully introduced onto the market l o Summary of the Invention
The present invention relates to Mus musculus mas, in particular Mus musculus mas polypeptides and Mus musculus mas polynucleotides, recombinant materials and methods for their production. In another aspect, the invention relates to methods for identifying agonists and antagonists/inhibitors of the Mus musculus mas gene. This invention further relates to the 15 generation of in vitro and in vivo comparison data relating to the polynucleotides and polypeptides in order to predict oral absorption and pharmacokmetics in man of compounds that either agonize or antagonize the biological activity of such polynucleotides or polypeptides. Such a comparison of data will enable the selection of drugs with optimal pharmacokmetics in man, i.e.. good oral bioavailabi ty. blood-brain barrier penetration, plasma half life, and minimum drug interaction 0 The present invention further relates to methods for creating transgenic animals, which overexpress or underexpress or have regulatable expression of a mas gene and "knock-out" animals, preferably mice, which an animal no longer expresses a mas gene. Furthermore, this invention relates to transgenic and knock-out animals obtained by using these methods. Such animal models are expected to provide valuable insight into the potential pharmacological and toxicological effects 5 in humans of compounds that are discovered by the aforementioned screening methods as well as other methods An understanding of how a Mus musculus mas gene functions in these animal models is expected to provide an insight into treating and preventing human diseases including, but not limited to. infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV-2; pam, cancers; diabetes, obesity; anorexia: bulimia; asthma; 0 Parkinson's disease; acute heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectons; myocardial infarction; stroke; ulcers; asthma; allergies; benign prostatic hypertrophy; migraine; vomiting; psychotic and neurological disorders, including anxiety, schizophrenia, manic depression, depression, delirium, dementia, and severe mental retardation: and dyskmesias, such as Huntmgton's disease or Gilles dela Tourett's syndrome, hereinafter referred to as "the Diseases", amongst others.
Description of the Invention
In a first aspect, the present invention relates to Mus musculus mas polypeptides. Such polypeptides include isolated polypeptides comprising an ammo acid sequence having at least a
95%o identity, most preferably at least a 97-99% identity, to that of SEQ ID NO:2 over the entire length of SEQ ID NO:2. Such polypeptides include those comprising the amino acid of SEQ ID
NO:2.
(a) an isolated polypeptide encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 1 ;
(b) an isolated polypeptide comprising a polypeptide sequence having at least a 95%, 97%, 98%, or 99% identity to the polypeptide sequence of SEQ ID NO:2;
(c) an isolated polypeptide comprising the polypeptide sequence of SEQ ID NO:2;(d) an isolated polypeptide having at least a 95%, 97%, 98%, or 99% identity to the polypeptide sequence of SEQ ID NO:2;
(e) the polypeptide sequence of SEQ ID NO:2; and
(f) variants and fragments thereof: and portions of such polypeptides in (a) to (e) that generally contain at least 30 ammo acids, more preferably at least 50 ammo acids, thereof.
Polypeptides of the present invention are believed to be members of the mas family of polypeptides. They are. therefore, of interest, because understanding the biological activities of mas in mouse would help to understand the biological function of its human counterpart, human mas, and other related genes. In addition, 7TM receptors (G-Protem Coupled Receptors), more than any other gene family, are the targets for pharmaceutical intervention. Furthermore, the polypeptides of the present invention can be used to establish assays to predict oral absorbtion and pharmacokmetics m man and thus enhance compound and formulation design, among others. These properties, either alone or in the aggregate, are hereinafter referred to as "Mus musculus mas activity" or "Mus musculus mas polypeptide activity" or "biological activity of mas." Preferably, a polypeptide of the present invention exhibits at least one biological activity of Mus musculus mas.
Polypeptides of the present invention also includes variants of the aforementioned polypeptides, including alleles and splice variants. Such polypeptides vary from the reference polypeptide by insertions, deletions, and substitutions that may be conservative or non-conservative. Particularly preferred variants are those in which several, for instance from 50 to 30, from 30 to 20, from 20 to 10, from 10 to 5, from 5 to 3, from 3 to 2, from 2 to 1 or 1 ammo acids are inserted, substituted, or deleted, in any combination. Particularly preferred primers will have between 20 and 25 nucleotides Preferred fragments of polypeptides of the present invention include an isolated polypeptide comprising an ammo acid sequence having at least 15, 20, 30, 40, 50 or 100 contiguous ammo acids from the ammo acid sequence of SEQ ID NO:2, or an isolated polypeptide comprising an ammo acid sequence having at least 15, 20, 30, 40, 50 or 100 contiguous ammo acids truncated or deleted from the ammo acid sequence of SEQ ID NO:2. Also preferred are biologically active fragments that mediate activities of mas, including those with a similar activity or an improved activity, or with a decreased undesirable activity. Also included are those fragments that are antigemc or immunogenic m an animal, especially in a human. Particularly preferred are fragments comprising receptors or domains of enzymes that confer a function essential for viability of Mus musculus or the ability to initiate, or maintain cause the Diseases m an individual, particularly a human
Fragments of the polypeptides of the invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, these variants may be employed as intermediates for producing the full-length polypeptides of the invention
The polypeptides of the present invention may be in the form of a "mature" protein or may be a part of a larger protein such as a fusion protein. It is often advantageous to include an additional ammo acid sequence that contains secretory or leader sequences, pro-sequences, sequences that aid in purification, for instance, multiple histidine residues, or an additional sequence for stability during recombinant production.
The present invention also includes variants of the aforementioned polypeptides, that is polypeptides that vary from the referents by conservative ammo acid substitutions, whereby a residue is substituted by another with like characteristics. Typical substitutions are among Ala, Val, Leu and He; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr. Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 ammo acids are substituted, deleted, or added in any combination.
Polypeptides of the present invention can be prepared m any suitable manner. Such polypeptides include isolated naturally occurring polypeptides, recomb antly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood m the art.
In a further aspect, the present invention relates to Mus musculus mas polynucleotides. Such polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide having at least a 95% identity, to the ammo acid sequence of SEQ ID N0:2, over the entire length of SEQ ID NO:2. In this regard, polypeptides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identity are more highly preferred, and those with at least a 99% identity are most highly preferred. Such polynucleotides include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO: l encoding the polypeptide of SEQ ID NO:2.
Further polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence having at least a 95% identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO:2, over the entire coding region. In this regard, polynucleotides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identity are more highly preferred, and those with at least a 99% identity are most highly preferred.
Further polynucleotides of the present invention include isolated polynucleotides comprising a nucleotide sequence having at least a 95% identity, to SEQ ID NO: 1 over the entire length of SEQ ID NO: 1. In this regard, polynucleotides which have at least a 97% identity are highly preferred, while those with at least a 98-99% identify are more highly preferred, and those with at least a 99% identity are most highly preferred. Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO: l , as well as the polynucleotide of SEQ ID NO: 1.
The invention also provides polynucleotides that are complementary to all the above described polynucleotides. The nucleotide sequence of SEQ ID NO: 1 shows homology with mouse mas. The nucleotide sequence of SEQ ID NO: 1 is a cDNA sequence and comprises a polypeptide encoding sequence (nucleotide 1 to 975) encoding a polypeptide of 324 ammo acids, the polypeptide of SEQ ID NO:2. The nucleotide sequence encoding the polypeptide of SEQ ID NO:2 may be identical to the polypeptide encoding sequence of SEQ ID NO: 1 or it may be a sequence other than SEQ ID NO: l , which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ID NO:2. The polypeptide of SEQ ID NO:2 is structurally related to other proteins of the mas family, having homology and/or structural similarity with mouse mas. Preferred polypeptides anc polynucleotides of the present invention are expected to have, inter aha, similar biological functians/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides and polynucleotides of the present invention have at least one mas activity. Polynucleotides of the present invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of Mus musculus liver, using the expressed sequence tag (EST) analysis (Adams, M.D., et al. Science (1991) 252: 1651- 1656; Adams, M.D. et al, Nature (1992) 355:632-634; Adams, M.D., et al. Nature (1995) 377 Supp.: 3-174). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesized using well known and commercially available techniques.
When polynucleotides of the present invention are used for the recombinant production of polypeptides of the present invention, the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide m reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions. For example, a marker sequence that facilitates purification of the fused polypeptide can be encoded. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, Inc.) and described in Gentz, et al . Proc Natl Acad ci USA (1989) 86:821- 824, or is an HA tag. The polynucleotide may also comprise non-coding 5' and 3" sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, πbosome binding sites and sequences that stabilize mRNA.
Further embodiments of the present invention include polynucleotides encoding polypeptide variants that comprise the ammo acid sequence of SEQ ID NO:2 and in which several, for instance from 50 to 30, from 30 to 20, from 20 to 10. from 10 to 5, from 5 to 3, from 3 to 2, from 1 to 1 or 1 ammo acid residues are substituted, deleted or added, in any combination. Particularly preferred probes will have between 30 and 50 nucleotides, but may have between 100 and 200 contiguous nucleotides of the polynucleotide of SEQ ID NO. l .
A preferred embodiment of the invention is a polynucleotide of consisting of or comprising nucleotide ATG to the nucleotide immediately upstream of or including nucleotide TGA set forth in SEQ ID NO: 1 , both of which encode a mas polypeptide.
The invention also includes a polynucleotide consisting of or comprising a polynucleotide of the formula: X-(R1)m-(R2)-(R3)n-Y wherein, at the 5' end of the molecule, X is hydrogen, a metal or a modified nucleotide residue, or together with Y defines a covalent bond, and at the 3' end of the molecule, Y is hydrogen, a metal, or a modified nucleotide residue, or together with X defines the covalent bond, each occurrence of Rj and R3 is independently any nucleic acid residue or modified nucleic acid residue, m is an integer between 1 and 3000 or zero, n is an integer between 1 and 3000 or zero, and R2 is a nucleic acid sequence or modified nucleic acid sequence of the invention, particularly the nucleic acid sequence set forth in SEQ ID NO: l or a modified nucleic acid sequence thereof. In the polynucleotide formula above, R2 is oriented so that its 5' end nucleic acid residue is at the left, bound to R] and its 3' end nucleic acid residue is at the right, bound to R3. Any stretch of nucleic acid residues denoted by either Rj and/or R2, where m and/or n is greater than 1, may be either a heteropolymer or a homopolymer, preferably a heteropolymer. Where, in a preferred embodiment, X and Y together define a covalent bond, the polynucleotide of the above formula is a closed, circular polynucleotide, which can be a double-stranded polynucleotide wherein the formula shows a first strand to which the second strand is complementary. In another preferred embodiment m and/or n is an integer between 1 and 1000. Other preferred embodiments of the invention are provided where m is an integer between 1 and 50, 100 or 500, and n is an integer between 1 and 50, 100, or 500.
Polynucleotides that are identical, or are substantially identical to a nucleotide sequence of SEQ ID NO: 1 , may be used as hybridization probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides of the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding homologs and orthologs from species other than Mus musculus) that have a high sequence identity to SEQ ID NO.1. Typically these nucleotide sequences are 95% identical to that of the referent. Preferred probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides, and may even have at least 100 nucleotides. Particularly preferred primers will have between 20 and 25 nucleotides.
A polynucleotide encoding a polypeptide of the present invention, mcludmg homologs and orthologs from a species other than Mus musculus, may be obtained by a process comprising the steps of screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: l or a fragment thereof, preferably of at least 15 nucleotides in length; and isolating full-length cDNA and genomic clones comprising said polynucleotide sequence. Such hybridization techniques are well known to the skilled artisan Preferred stringent hybridization conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCl, 15mM tπsodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters m O.l x SSC at about 65°C. Thus, the present invention also includes isolated polynucleotides, preferably of at least 100 nucleotides in length, obtained by screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: l or a fragment thereof, preferably of at least 15 nucleotides.
The skilled artisan will appreciate that, in many cases, an isolated cDNA sequence will be incomplete, that the region coding for the polypeptide is cut short at the 5' end of the cDNA. This is a consequence of reverse transcπptase, an enzyme with inherently low 'processivity' (a measure of the ability of the enzyme to remain attached to the template during the polymerization reaction), failing to complete a DNA copy of the mRNA template during 1 st strand cDNA synthesis. There are several methods available and well known to those skilled in the art to obtain full- length cDNAs, or extend short cDNAs, for example, those based on the method of Rapid Amplification of cDNA ends (RACE) (see, for example, Frohman, et al , Proc Natl Acad. Sci , USA 85, 8998-9002, 1988). Recent modifications of the technique, exemplified by the Marathon1 technology (Clontech Laboratories Inc.), for example, have significantly simplified the search for longer cDNAs. In the Marathon™ technology, cDNAs have been prepared from mRNA extracted from a chosen tissue and an 'adaptor' sequence hgated onto each end. Nucleic acid amplification (PCR) is then carried out to amplify the 'missing' 5' end of the cDNA using a combination of gene specific and adaptor specific ohgonucleotide primers. The PCR reaction is then repeated using 'nested' primers, that is, primers designed to anneal withm the amplified product (typically an adaptor specific primer that anneals further 3' in the adaptor sequence and a gene specific primer that anneals further 5' m the known gene sequence). The products of this reaction can then be analyzed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or carrying out a separate full-length PCR using the new sequence information for the design of the 5' primer. Recombmant polypeptides of the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, m a further aspect, the present invention relates to expression systems comprising a polynucleotide or polynucleotides of the present invention, to host cells which are genetically engmeered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
For recombinant production, host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention. Introduction of polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis, et al, BASIC METHODS IN MOLECULAR BIOLOGY (1986) and Sambrook, et al, MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989). Preferred methods of introducing polynucleotides into host cells include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, micro jection, catiomc hpid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection
Representative examples of appropriate hosts include bacterial cells, such as streptococci, staphylococci, E coh, Streptomyces and Bacillus subtihs cells; fungal cells, such as yeast cells and Aspergillus cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells, animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells; and plant cells
A great variety of expression systems can be used, for instance, chromosomal, episomal and virus-derived systems, e.g , vectors derived from bacterial plasmids, from bacteπophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteπophage genetic elements, such as cosmids and phagemids. The expression systems may comprise control regions that regulate as well as engender expression Generally, any system or vector that is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used The appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook, et al, MOLECULAR CLONING, A LABORATORY MANUAL (supra).
If a polypeptide of the present invention is to be expressed for use in screening assays, it is generally preferred that the polypeptide be produced at the surface of the cell. In this event, the cells may be harvested prior to use in the screening assay. If the polypeptide is secreted into the medium, the medium can be recovered in order to recover and purify the polypeptide. If produced lntracellularly, the cells must first be lysed before the polypeptide is recovered Polypeptides of the presen invention can be recovered and purified from recombinant cell cultures by well-known methods lr eluding ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectm chromatography. Most preferably, high performance liquid chromatography is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
The polynucleotide sequences of the present invention are also valuable for chromosome localization studies. The polynucleotide sequence, or fragment(s) thereof, is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome. The mapping of these sequences to human chromosomes according to the present invention is an important first step in correlating homologous human polynucleotide sequences with gene associated disease in humans.
Precise chromosomal localizations for a polynucleotide sequence (gene fragment etc) can be determined using Radiation Hybrid (RH) Mapping (Walter, M., et al (1994) Nature Genetics 7, 22-28), for example. A number of RH panels are available, including mouse, rat, baboon, zebrafish and human. RH mapping panels are available from a number of sources, for example Research Genetics (Huntsville, AL, USA). To determine the chromosomal location of a polynucleotide sequence using these panels, PCR reactions are performed using primers, designed to the polynucleotide sequence of interest, on the RH DNAs of the panel. Each of these DNAs contains random genomic fragments from the species of interest. These PCRs result in a number of scores, one for each RH DNA m the panel, indicating the presence or absence of the PCR product of the polynucleotide sequence of interest These scores are compared with scores created using PCR products from genomic sequences of known location, usually using an on-line resource such as that available at the Whitehead Institute for Biomedical Research in Cambridge, Massachusetts, USA website (http:// www. genome wi.mit.edu/). Once a polynucleotide sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data for that species. Also, as a consequence of synteny, where knowledge of the position of a gene on a chromosome of one species can be used to determine the likely position of the orthologous gene on the chromosome of another species, this knowledge can then be used to identify candidate genes for human disease Thus the localization of a polynucleotide sequence of interest to a specific mouse chromosomal location can be used to predict the localization of the orthologous human gene on the corresponding human chromosome From this data, potential disease association may be inferred from genetic map sources such as, for example, V. McKusick, Mendehan Inheritance in Man (available on-lme through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (co- lnheπtance of physically adjacent genes). Mus musculus mas gene products can be expressed in transgenic animals. Animals of any species, including, but not limited to: mice, rats, rabbits, guinea pigs, dogs, cats, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, chimpanzees, may be used to generate mas transgenic animals.
This invention further relates to a method of producing transgenic animals, preferably Mus musculus, over-expressing mas, which method may comprise the introduction of several copies of a segment comprising at least the polynucleotide sequence encoding SEQ ID NO:2 with a suitable promotor into the cells of a Mus musculus embryo, or the cells of another species, at an early stage
This invention further relates to a method of producing transgenic animals, preferably Mus musculus, under-expressing or regulatably expressing mas, which method may comprise the introduction of a weak promoter or a regulatable promoter (e g. , an mducible or repressible promoter) respectively, expressibly linked to the polynucleotide sequence of SEQ ID NO: l into the cells of a Mus musculus embryo at an early stage
This invention also relates to transgenic animals, characterized in that they are obtained by a method, as defined above Any technique known in the art may be used to introduce a Mus musculus mas transgene into animals to produce a founder line of animals. Such techniques include, but are not limited to pronuclear micromjection (U.S. Patent No. 4,873, 191 ); retrovirus mediated gene transfer into germ lines (Van der Putten, et al . Proc Natl Acad Sci , USA 82. 6148-6152 ( 1985); gene targeting m embryonic stem cells (Thompson, et al , Cell 56: 313-321 (1989), electropolation of embryos (Lo, Mol. Cell Bwl 3: 1803-1814 ( 1983 ); and sperm-mediated gene transfer (Lavitrano, et al . Cell 57 717-723 ( 1989); etc. For a review of such techniques, see Gordon, Intl Rev Cytol 1 15 ' 171 -229 ( 1989).
A further aspect of the present invention involves gene targeting by homologous recombination in embryonic stem cells to produce a transgenic animal with a mutation in a mas gene ("knock-out" mutation). In such so-called "knock-out" animals, there is mactivation of the mas gene or altered gene expression, such that the animals are useful to study the function of the mas gene, thus providing animals models of human disease, which are otherwise not readily available through spontaneous, chemical or irradiation mutagenesis. Another aspect of the present invention involves the generation of so-called "knock-m" animals in which a portion of a wild-type gene is fused to the cDNA of a heterologous gene.
This invention further relates to a method of producing "knock-out" animals, preferably 5 mice, no longer expressing mas. By using standard cloning techniques, a Mus musculus mas cDNA (SEQ ID NO. l ) can be used as a probe to screen suitable libraries to obtain the muπne mas genomic DNA clone. Using the murme genomic clone, the method used to create a knockout mouse is characterized in that: a suitable mutation is produced in the polynucleotide sequence of the murme mas genomic 10 clone, which inhibits the expression of a gene encoding murme mas, or inhibits the activity of the gene product, said modified murme mas polynucleotide is introduced into a homologous segment of murme genomic DNA, combined with an appropriate marker, so as to obtain a labeled sequence comprising said modified murme genomic DNA; i s said modified murme genomic DNA comprising the modified polynucleotide is transfected mto embryonic stem cells and correctly targeted events selected in vitro: then said stem cells are remjected into a mouse embryo; then said embryo is implanted into a female recipient and brought to term as a chimera which transmits said mutation through the germhne; and
20 homozygous recombinant mice are obtained at the F2 generation which are recognizable by the presence of the marker.
Various methods for producing mutations in non-human animals are contemplated and well known in the art. In a preferred method, a mutation is generated in a murme mas allele by the introduction of a DNA construct comprising DNA of a gene encoding murme mas, which murme
25 gene contains the mutation. The mutation is targeted to the allele by way of the DNA construct The DNA of the gene encoding mur e mas comprised in the construct may be foreign to the species of which the recipient is a member, may be native to the species and foreign only to the individual recipient, may be a construct comprised of synthetic or natural genetic components, or a mixture of these The mutation may constitute an insertion, deletion, substitution, or combination 0 thereof. The DNA construct can be introduced into cells by, for example, calcium-phosphate DNA co-precipitation It is preferred that a mutation be introduced into cells using electroporation, micromjection, virus infection, hgand-DNA conjugation, virus-hgand-DNA conjugation, or hposomes.
Another embodiment of the instant invention relates to "knock-out" animals, preferably mice, obtained by a method of producing recombinant mice as defined above, among others. Another aspect of this invention provides for in vitro mas "knock-outs", i.e., tissue cultures
Animals of any species, including, but not limited to: mice, rats, rabbits, guinea pigs, dogs, cats, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, chimpanzees, may be used to generate in vitro mas "knock-outs". Methods for "knocking out" genes in vitro are described in Galh-Tahadoros, et al , Journal of Immunological Methods 181 : 1 -15 (1995). Transgenic, "knock-m", and "knock-out" animals, as defined above, are a particularly advantageous model, from a physiological point of view, for studying mas. Such animals will be valuable tools to study the functions of a mas gene. Moreover, such animal models are expected to provide information about potential toxicological effects in humans of any compounds discovered by an aforementioned screening method, among others. An understanding of how a Mus musculus mas gene functions in these animal models is expected to provide an insight into treating and preventing human diseases including, but not limited to: infections such as bacterial, fungal, protozoan and viral infections, particularly infections caused by HIV-1 or HIV -2; pam; cancers; diabetes, obesity; anorexia; bulimia; asthma; Parkinson's disease; acute heart failure; hypotension; hypertension; urinary retention; osteoporosis; angina pectons; myocardial infarction; stroke; ulcers; asthma; allergies; bemgn prostatic hypertrophy; migraine; vomiting; psychotic and neurological disorders, including anxiety, schizophrenia, mamc depression, depression, delirium, dementia, and severe mental retardation; and dyskmesias, such as Huntmgton's disease or Gilles dela Tourett's syndrome
Polypeptides of the present invention are responsible for many biological functions, including many disease states, m particular the Diseases mentioned herein. It is, therefore, an aspect of the invention to devise screening methods to identify compounds that stimulate (agonists) or that inhibit (antagonists) the function of the polypeptide, such as agonists, antagonists and inhibitors. Accordingly, m a further aspect, the present invention provides for a method of screening compounds to identify those that stimulate or inhibit the function of the polypeptide. In general, agonists or antagonists may be employed for therapeutic and prophylactic purposes for the Diseases mentioned herein mentioned. Compounds may be identified from a variety of sources, for example, cells, cell-free preparations, chemical libraries, and natural product mixtures. Such agonists and antagonists so-identified may be natural or modified substrates, hgands, receptors, enzymes, etc., as the case may be, o the polypeptide; or may be structural or functional mimetics thereof (see Cohgan, et al , CURRENT PROTOCOLS IN IMMUNOLOGY 1(2): Chapter 5 (1991)).
The screening method may simply measure the binding of a candidate compound to the polypeptide, or to cells or membranes bearing the polypeptide, or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound. Alternatively, a screening method may involve measuring or, qualitatively or quantitatively, detecting the competition of binding of a candidate compound to the polypeptide with a labeled competitor (e.g., agonist or antagonist). Further, screening methods may test whether the candidate compound results in a signal generated by an agonist or antagonist of the polypeptide, using detection systems appropriate to cells bearing the polypeptide. Antagonists are generally assayed in the presence of a known agonist and an effect on activation by the agonist by the presence of the candidate compound is observed. Further, screening methods may simply comprise the steps of mixing a candidate compound with a solution comprising a polypeptide of the present invention, to form a mixture, measuring Mus musculus mas activity m the mixture, and comparing a Mus musculus mas activity of the mixture to a control mixture which contains no candidate compound.
Polypeptides of the present invention may be employed in conventional low capacity screening methods and also in high-throughput screening (HTS) formats. Such HTS formats include not only the well-established use of 96- and, more recently, 384-well microtiter plates but also emerging methods such as the nanowell method described by Schullek, et al , Anal Biochem., 246, 20-29, (1997).
Fusion proteins, such as those made from Fc portion and Mus musculus mas polypeptide, as herein descπbed, can also be used for high-throughput screening assays to identify antagonists of antagonists of the polypeptide of the present invention (see D. Bennett, et al. J Mol Recognition, 8 52-58 (1995), and K. Johanson, et al , J Bwl Chem., 270(16):9459-9471 (1995)).
One screening technique includes the use of cells which express the receptor of this invention (for example, transfected CHO cells) in a system which measures extracellular pH or intracellular calcium changes caused by receptor activation. In this technique, compounds may be contacted with cells expressing the receptor polypeptide of the present invention A second messenger response, e.g., signal transduction, pH changes, or changes m calcium level, is then measured to determine whether the potential compound activates or inhibits the receptor.
Another method involves screening for receptor inhibitors by determining inhibition or stimulation of receptor-mediated cAMP and/or adenylate cyclase accumulation. Such a method mvolves transfecting a eukaryotic cell with the receptor of this invention to express the receptor on the cell surface. The cell is then exposed to potential antagonists in the presence of the receptor of this invention. The amount of cAMP accumulation is then measured. If the potential antagonist binds the receptor, and thus inhibits receptor binding, the levels of receptor-mediated cAMP, or adenylate cyclase, activity will be reduced or increased.
Another methods for detecting agonists or antagonists for the receptor of the present invention is the yeast based technology as described in U.S. Patent No. 5,482,835.
Examples of potential polypeptide antagonists include antibodies or, in some cases, ohgopeptides or proteins that are closely related to ligands, substrates, receptors, enzymes, etc , as the case may be, of a mas polypeptide, e g., a fragment of a ligand, substrate, receptor, enzyme, etc., or small molecules which bind to a mas polypeptide but do not elicit a response, so that an activity of a mas polypeptide is prevented
Thus, in another aspect, the present invention relates to a screening kit for identifying agonists, antagonists, inhibitors, ligands, receptors, substrates, enzymes, etc for polypeptides of the present invention; or compounds which decrease or enhance the production of such polypeptides, which compounds comprise a member selected from the group consisting of.
(a) a polypeptide of the present invention;
(b) a recombinant cell expressing a polypeptide of the present invention; or
(c) a cell membrane expressing a polypeptide of the present invention; which polypeptide is preferably that of SEQ ID NO.2
It will be appreciated that in any such kit, (a), (b) or (c) may comprise a substantial component.
It will also be readily appreciated by the skilled artisan that a polypeptide of the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by.
(a) determining m the first instance the three-dimensional structure of the polypeptide,
(b) deducing the three-dimensional structure for the likely reactive or binding sιte(s) of an agonist, antagonist or inhibitor;
(c) synthesizing candidate compounds that are predicted to bind to or react with the deduced binding or reactive site, and (d) testing whether the candidate compounds are indeed agonists, antagonists or inhibitors.
It will be further appreciated that this will normally be an iterative process.
In an alternative preferred embodiment, the present invention relates to the use of Mus musculus mas polypeptides, polynucleotides, and recombinant materials thereof in selection screens to identify compounds which are neither agonists nor antagonist/inhibitors of Mus musculus mas. The data from such a selection screen is expected to provide in vitro and in vivo comparisons and to predict oral absorption, pharmacokmetics in humans. The ability to make such a comparison of data will enhance formulation design through the identification of compounds with optimal development characteristics, i.e . high oral bioavailabihty, UID (once a day) dosmg, reduced drug interactions, reduced variability, and reduced food effects, among others.
The following definitions are provided to facilitate understanding of certain terms used frequently herein.
"Allele" refers to one or more alternative forms of a gene occurring at a given locus in the genome. "Fragment" of a polypeptide sequence refers to a polypeptide sequence that is shorter than the reference sequence but that retains essentially the same biological function or activity as the reference polypeptide. "Fragment" of a polynucleotide sequence refers to a polynucleotide sequence that is shorter than the reference sequence of SEQ ID NO: l .
"Fusion protein" refers to a protein encoded by two, often unrelated, fused genes or fragments thereof. In one example, EP-A-0 464 discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof. In many cases, employing an immunoglobulin Fc region as a part of a fusion protein is advantageous for use m therapy and diagnosis resulting m, for example, improved pharmacokmetic properties [see, e.g , EP-A 0232 262]. On the other hand, for some uses, it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected, and purified.
"Homolog" is a generic term used in the art to indicate a polynucleotide or polypeptide sequence possessing a high degree of sequence relatedness to a reference sequence. Such relatedness may be quantified by determining the degree of identity and/or similarity between the two sequences as hereinbefore defined. Falling withm this generic term are the terms, "ortholog", and "paralog". "Ortholog" refers to polynucleotides/genes or polypeptide that are homologs via speciation. that is closely related and assumed to have commend descent based on structural and functional considerations. "Paralog" refers to polynucleotides/genes or polypeptide that are homologs via gene duplication, for instance, duplicated variants withm a genome.
"Identity" reflects a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, determined by comparing the sequences. In general, identity refers to an exact nucleotide to nucleotide or ammo acid to ammo acid correspondence of the two polynucleotide or two polypeptide sequences, respectively, over the length of the sequences being compared. For sequences where there is not an exact correspondence, a "% identity" may be determined. In general, the two sequences to be compared are aligned to give a maximum correlation between the sequences. This may include inserting "gaps" m either one or both sequences, to enhance the degree of alignment. A % identity may be determined over the whole length of each of the sequences bemg compared (so-called global alignment), that is particularly suitable for sequences of the same or very similar length, or over shorter, defined lengths (so-called local alignment), that is more suitable for sequences of unequal length.
"Similarity" is a further, more sophisticated measure of the relationship between two polypeptide sequences. In general, "similarity" means a comparison between the amino acids of two polypeptide chains, on a residue by residue basis, taking into account not only exact correspondences between a between pairs of residues, one from each of the sequences being compared (as for identity) but also, where there is not an exact correspondence, whether, on an evolutionary basis, one residue is a likely substitute for the other. This likelihood has an associated 'score' from which the "%. similarity" of the two sequences can then be determined.
Methods for comparing the identity and similarity of two or more sequences are well known in the art. Thus for instance, programs available in the Wisconsin Sequence Analysis Package, version 9.1 (Devereux J., et al, Nucleic Acids Res, 12, 387-395, 1984, available from Genetics Computer Group, Madison, Wisconsin, USA), for example the programs BESTFIT and GAP, may be used to determine the % identity between two polynucleotides and the % identity and the % similarity between two polypeptide sequences. BESTFIT uses the "local homology" algorithm of Smith and Waterman (J. Mol Biol, 147: 195-197, 1981, Advances in Applied Mathematics, 2, 482- 489, 1981) and finds the best single region of similarity between two sequences. BESTFIT is more suited to comparing two polynucleotide or two polypeptide sequences that are dissimilar in length, the program assuming that the shorter sequence represents a portion of the longer. In comparison, GAP aligns two sequences, finding a "maximum similarity", according to the algorithm of Neddleman and Wunsch (J Mo I Biol , 48, 443-453, 1970). GAP is more suited to comparing sequences that are approximately the same length and an alignment is expected over the entire length. Preferably, the parameters " 3ap Weight" and "Length Weight" used m each program are 50 and 3, for polynucleotide sequences and 12 and 4 for polypeptide sequences, respectively. Preferably, % identities and similarities are determined when the two sequences bemg compared are optimally aligned. Other programs for determining identity and/or similarity between sequences are also known in the art, for instance the BLAST family of programs (Altschul S.F., et al , J. Mol Biol , 215, 403-410, 1990, Altschul S.F., et al, Nucleic Acids Res , 25:389-3402, 1997, available from the National Center for Biotechnology Information (NCBI), Bethesda, Maryland, USA and accessible through the home page of the NCBI at www.ncbi.nlm.mh.gov) and FASTA (Pearson W R, Methods in Enzymology, 183: 63-99 (1990); Pearson W R and Lipman D.J., Proc Nat Acad Sci USA, 85: 2444-2448 (1988) (available as part of the Wisconsin Sequence Analysis Package)
Preferably, the BLOSUM62 ammo acid substitution matrix (Hemkoff S. and Hemkoff J G , Proc Nat Acad Sci USA, 89. 10915-10919 (1992)) is used in polypeptide sequence comparisons including where nucleotide sequences are first translated mto ammo acid sequences before comparison.
Preferably, the program BESTFIT is used to determine the % identity of a query polynucleotide or a polypeptide sequence with respect to a polynucleotide or a polypeptide sequence of the present invention, the query and the reference sequence being optimally aligned and the parameters of the program set at the default value, as hereinbefore described Alternatively, for instance, for the purposes of interpreting the scope of a claim including mention of a "% identity" to a reference polynucleotide, a polynucleotide sequence having, for example, at least 95% identity to a reference polynucleotide sequence is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference sequence Such point mutations are selected from the group consisting of at least one nucleotide deletion, substitution, mcludmg transition and transversion, or insertion. These point mutations may occur at the 5' or 3' terminal positions of the reference polynucleotide sequence or anywhere between these terminal positions, interspersed either individually among the nucleotides m the reference sequence or in one or more contiguous groups withm the reference sequence. In other words, to obtain a polynucleotide sequence having at least 95% identity to a reference polynucleotide sequence, up to 5% of the nucleotides of the in the reference sequence may be deleted, substituted or inserted, or any combination thereof, as hereinbefore described. The same applies mutatis mutandis for other % identities such as 96%, 97%, 98%, 99% and 100%. For the purposes of interpreting the scope of a claim including mention of a "% identity" to a reference polypeptide, a polypeptide sequence having, for example, at least 95% identity to a reference polypeptide sequence is identical to the reference sequence except that the polypeptide sequence may include up to five point mutations per each 100 ammo acids of the reference sequence. Such point mutations are selected from the group consisting of at least one ammo acid deletion, substitution, including conservative and non-conservative substitution, or insertion. These point mutations may occur at the ammo- or carboxy- terminal positions of the reference polypeptide sequence or anywhere between these terminal positions, interspersed either individually among the ammo acids in the reference sequence or m one or more contiguous groups within the reference sequence. In other words, to obtain a sequence polypeptide sequence having at least 95% identity to a reference polypeptide sequence, up to 5% of the ammo acids of the m the reference sequence may be deleted, substituted or inserted, or any combination thereof, as hereinbefore descπbed. The same applies mutatis mutandis for other % identities such as 96%, 97%, 98%, 99%, and 100%.
A preferred meaning for "identity" for polynucleotides and polypeptides, as the case may be, are provided in (1) and (2) below.
(1 ) Polynucleotide embodiments further include an isolated polynucleotide comprising a polynucleotide sequence having at least a 95, 97 or 100% identity to the reference sequence of SEQ ID NO: l , wherein said polynucleotide sequence may be identical to the reference sequence of SEQ ID NO: l or may include up to a certain integer number of nucleotide alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or m one or more contiguous groups within the reference sequence, and wherein said number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ ID NO: l by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of nucleotides in SEQ ID NO: l , or: nn < xn - (xn . y),
wherein nn is the number of nucleotide alterations, xn is the total number of nucleotides in SEQ ID NO: 1 , y is 0 95 for 95%. 0.97 for 97% or 1 00 for 100%. and • is the symbol for the multiplication operator, and wherein any non-mteger product of xn and y is rounded down to the nearest integer prior to subtracting it from xn. Alterations of a polynucleotide sequence encoding the polypeptide of SEQ ID NO:2 may create nonsense, missense or frameshift mutations m this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
(2) Polypeptide embodiments further include an isolated polypeptide comprising a polypeptide having at least a 95, 97 or 100% identity to a polypeptide reference sequence of SEQ ID NO:2, wherein said polypeptide sequence may be identical to the reference sequence of SEQ ID NO:2 or may include up to a certain integer number of amino acid alterations as compared to the reference sequence, wherein said alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the ammo- or carboxy-termmal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the ammo acids in the reference sequence or in one or more contiguous groups withm the reference sequence, and wherein said number of ammo acid alterations is determined by multiplying the total number of ammo acids m SEQ ID NO:2 by the integer defining the percent identity divided by 100 and then subtracting that product from said total number of ammo acids in SEQ ID NO:2, or
na < xa - (xa • y),
wherein na is the number of ammo acid alterations, xa is the total number of amino acids in SEQ ID NO:2, y is 0.95 for 95%, 0.97 for 97% or 1.00 for 100%, and • is the symbol for the multiplication operator, and wherein any non-mteger product of xa and y is rounded down to the nearest integer prior to subtracting it from xa. "Isolated" means altered "by the hand of man" from its natural state, / e , if it occurs in nature, it has been changed or removed from its original environment, or both. For example, a polynucleotide or a polypeptide naturally present in a living organism is not "isolated," but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is "isolated", as the term is employed herein. Moreover, a polynucleotide or polypeptide that is introduced into an organism by transformation, genetic manipulation or by any other recombinant method is "isolated" even if it is still present in said organism, which organism may be living or non-living.
"Knock-in" refers to the fusion of a portion of a wild-type gene to the cDNA of a heterologous gene
"Knock-out" refers to partial or complete suppression of the expression of a protein encoded by an endogenous DNA sequence m a cell. The "knock-out" can be affected by targeted deletion of the whole or part of a gene encoding a protein, in an embryonic stem cell. As a result, the deletion may prevent or reduce the expression of the protein in any cell in the whole animal in which it is normally expressed.
"Splice Variant" as used herein refers to cDNA molecules produced from RNA molecules initially transcribed from the same genomic DNA sequence but which have undergone alternative RNA splicing. Alternative RNA splicing occurs when a primary RNA transcript undergoes splicing, generally for the removal of introns, which results m the production of more than one mRNA molecule each of that may encode different ammo acid sequences. The term splice variant also refers to the proteins encoded by the above cDNA molecules.
"Transgenic animal" refers to an animal to which exogenous DNA has been introduced while the animal is still in its embryonic stage. In most cases, the transgenic approach aims at specific modifications of the genome, e g , by introducing whole transcπptional units into the genome, or by up- or down-regulat g pre-existing cellular genes. The targeted character of certain of these procedures sets transgenic technologies apart from experimental methods which random mutations are conferred to the germhne, such as administration of chemical mutagens or treatment with ionizing solution.
"Polynucleotide" generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. "Polynucleotides" include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double- stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double- stranded regions, hybrid molecules comprising DNA and RNA that may be smgle-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions. In addition, "polynucleotide" refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA The term "polynucleotide" also includes DNAs or RNAs comprising one or more modified bases and DNAs or RNAs with backbones modified for stability or for other reasons. "Modified" bases include, for example, tritylated bases and unusual bases such as inosine. A variety of modifications may be made to DNA and RNA; thus, "polynucleotide" embraces chemically, enzy atically or metabohcally modified forms of polynucleotides as typically found in nature, as well as the chemical forms of DNA and RNA characteristic of viruses and cells. "Polynucleotide" also embraces relatively short polynucleotides, often referred to as ohgonucleotides. "Polypeptide" refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds, / e , peptide isosteres. "Polypeptide" refers to both short chains, commonly referred to as peptides, ohgopeptides or ohgomers, and to longer chains, generally referred to as proteins. Polypeptides may comprise ammo acids other than the 20 gene-encoded amino acids. 'Polypeptides" include amino acid sequences modified either by natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well descπbed m basic texts and in more detailed monographs, as well as m a voluminous research literature. Modifications may occur anywhere m a polypeptide, including the peptide backbone, the ammo acid side-chams and the amino or carboxyl terrnim. It will be appreciated that the same type of modification may be present to the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may comprise many types of modifications. Polypeptides may be branched as a result of ubiquitmation, and they may be cyclic, with or without branching. Cyclic, branched and branched cyclic polypeptides may result from post-translation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-πbosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a hpid or hpid derivative, covalent attachment of phosphotidyhnositol, cross-linking, cychzation, disulfide bond formation, demethylation, formation of covalent cross-lmks, formation of cysteme, formation of pyroglutamate, formylation, gamma- carboxylation, glycosylation, GPI anchor formation, hydroxylation, lodination, methylation, myπstoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as argmylation, and ubiquitmation (see, for instance, PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E Creighton, W. H. Freeman and Company, New
York, 1993; Wold, F., Post-translational Protein Modifications: Perspectives and Prospects, pgs. 1- 12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, 1983; Seifter, et al , "Analysis for protein modifications and nonprotein cofactors", Meth. Enzymol (1990) 182:626-646 and Rattan, et al, "Protein Synthesis: Post-translational Modifications and Aging", Ann NY Acad Sci (1992) 663:48-62).
"Variant" refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, but retains essential properties. A typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the am o acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result m ammo acid substitutions, additions, deletions, fusions and truncations m the polypeptide encoded by the reference sequence, as discussed below. A typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical. A variant and reference polypeptide may differ m ammo acid sequence by one or more substitutions, additions, deletions in any combination. A substituted or inserted ammo acid residue may or may not be one encoded by the genetic code. A variant of a polynucleotide or polypeptide may be a naturally occurring such as an allehc variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
All publications including, but not limited to, patents and patent applications, cited in this specification or to which this patent application claims pπoπty, are herein incorporated by reference as if each individual publication were specifically and individually indicated to be incorporated by reference herein as though fully set forth.
Examples
Example 1 : Mammalian Cell Expression
The receptors of the present invention are expressed in either human embryonic kidney 293 (HEK293) cells or adherent dhfr CHO cells. To maximize receptor expression, typically all 5" and 3* untranslated regions (UTRs) are removed from the receptor cD A prior to insertion into a pCDN or pCDNA3 vector. The cells are transfected with individual receptor cDNAs by lipofectm and selected in the presence of 400 mg/ml G418. After 3 weeks of selection, individual clones are picked and expanded for further analysis. HEK293 or CHO cells transfected with the vector alone serve as negative controls. To isolate cell lines stably expressing the individual receptors, about 24 clones are typically selected and analyzed by Northern blot analysis. Receptor mRNAs are generally detectable m about 50% of the G418-resιstant clones analyzed.
Example 2. Ligand bank for binding and functional assays
A bank of over 600 putative receptor ligands has been assembled for screening. The bank compπses: transmitters, hormones and chemokines known to act via a human seven transmembrane (7TM) receptor; naturally occurring compounds which may be putative agonists for a human 7TM receptor, non-mammalian, biologically active peptides for which a mammalian counterpart has not yet been identified; and compounds not found m nature, but which activate 7TM receptors with unknown natural ligands. This bank is used to initially screen the receptor for known ligands, using both functional (i.e . calcium, cAMP, microphysiometcr, oocyte electrophysiology, etc, see below) as well as binding assays.
Example 3. Ligand Binding Assays
Ligand binding assays provide a direct method for ascertaining receptor pharmacology and are adaptable to a high throughput format. The purified ligand for a receptor is radiolabeled to high specific activity (50-2000 Ci/mmol) for binding studies. A determination is then made that the process of radiolabelmg does not dimmish the activity of the ligand towards its receptor. Assay conditions for buffers, ions, pH and other modulators such as nucleotides are optimized to establish a workable signal to noise ratio for both membrane and whole cell receptor sources. For these assays, specific receptor binding is defined as total associated radioactivity minus the radioactivity measured in the presence of an excess of unlabeled competing ligand. Where possible, more than one competing ligand is used to define residual nonspecific binding.
Example 4 Functional Assay in Xenopus Oocytes Capped RNA transcripts from linearized plasmid templates encoding the receptor cDNAs of the invention are synthesized in vitro with RNA polymerases in accordance with standard procedures. In vitro transcripts are suspended in water at a final concentration of 0.2 mg/ml. Ovarian lobes are removed from adult female toads, Stage V defolhculated oocytes are obtained, and RNA transcripts ( 10 ng/oocyte) are injected in a 50 nl bolus using a microinjection apparatus. Two electrode voltage clamps are used to measure the currents from individual Xenopus oocytes m response to agonist exposure. Recordings are made in Ca2+ free Barth's medium at room temperature. The Xenopus system can be used to screen known ligands and tissue/cell extracts for activating ligands. Example 5. Microphysiometπc Assays
Activation of a wide variety of secondary messenger systems results m extrusion of small amounts of acid from a cell. The acid formed is largely as a result of the increased metabolic activity required to fuel the mtracellular signaling process. The pH changes in the media surrounding the cell are very small but are detectable by the CYTOSENSOR rmcrophysiometer (Molecular Devices Ltd., Menlo Park, CA). The CYTOSENSOR is thus capable of detecting the activation of a receptor which is coupled to an energy utilizing mtracellular signaling pathway such as the G-protem coupled receptor of the present invention
Example 6 Extract/Cell Supernatant Screening
A large number of mammalian receptors exist for which there remains, as yet, no cognate activating ligand (agonist). Thus, active ligands for these receptors may not be included within the ligands banks as identified to date. Accordingly, the 7TM receptor of the invention is also functionally screened (using calcium, cAMP, microphysiometer, oocyte electrophysiology, etc., functional screens) against tissue extracts to identify natural ligands. Extracts that produce positive functional responses can be sequentially subfractionated until an activating ligand is isolated identified.
Example 7' Calcium and cAMP Functional Assays
7TM receptors which are expressed in HEK 293 cells have been shown to be coupled functionally to activation of PLC and calcium mobilization and/or cAMP stimulation or inhibition Basal calcium levels in the HEK 293 cells m receptor-transfected or vector control cells were observed to be in the normal, 100 nM to 200 nM, range. HEK 293 cells expressing recombinant receptors are loaded with fura 2 and in a single day > 150 selected ligands or tissue/cell extracts are evaluated for agonist induced calcium mobilization Similarly, HEK 293 cells expressing recombinant receptors are evaluated for the stimulation or inhibition of c AMP production using standard cAMP quantitation assays . Agonists presenting a calcium transient or cAMP fluctuation are tested in vector control cells to determine if the response is unique to the transfected cells expressing receptor.
SEQUENCE INFORMATION SEQ ID NO: 1
ATGGACCAGTCAAATATGACATCCCTTGCTGAAGAGAAAGCCATGAATACCTCCAGCAGAAATGCCTCCCTGGG GAGCTCACACCCACCCATTCCCATAGTGCACTGGGTCATCATGAGCATCTCCCCTCTGGGCTTTGTGGAGAATG GGATCCTCCTCTGGTTCCTCTGCTTCCGGATGAGGAGAAATCCCTTCACGGTCTACATTACCCACTTGTCCATT GCTGATATCTCTCTTCTGTTCTGTATTTTTATCCTGTCCATTGACTATGCTTTAGACTATGAACTCTCTTCTGG CCATCACTACACAATCGTGACATTATCGGTGACTTTTCTATTTGGCTACAACACGGGCCTCTATCTGCTGACAG CCATCAGTGTGGAGAGGTGCCTATCGGTCCTCTACCCCATATGGTACAGATGCCACCGCCCCAAGCACCAGTCA GCATTCGTCTGTGCCCTTCTGTGGGCACTTTCGTGCTTGGTGACCACCATGGAGTATGTCATGTGTATTGACAG CGGAGAAGAGAGTCATTCTCGGAGTGACTGCCGGGCGGTCATCATCTTCATAGCCATCCTCAGCTTCTTGGTCT TCACTCCGCTCATGTTAGTGTCCAGCACCATCTTGGTGGTGAAGATACGGAAGAACACGTGGGCCTCCCATTCT TCGAAGCTGTACATTGTCATCATGGTCACCATTATCATATTCCTCATCTTTGCCATGCCCATGCGGGTCCTCTA CTTGCTGTACTACGAGTACTGGTCAGCCTTTGGGAACCTGCATAACATCTCCTTGCTTTTCTCCACCATCAACA GCAGCGCCAACCCTTTCATCTACTTTTTTGTGGGCAGCAGTAAGAAGAAGCGCTTCAGGGAGTCCTTAAAAGTC GTCCTGACCAGGGCTTTCAAAGATGAAATGCAACCCAGGCGCCAGGAGGGCAACGGCAACACTGTATCCATTGA GACTGTGGTCTGA
SEQ ID NO:2
MDQSN TSLAEEKAMNTSSRNAS GSSHPPIPIVH VIMSISPLGFVENGILLWFLCFRMRRNPFTVYITHLSI ADISLLFCIFILSIDYA DYELSSGHHYTIVTLSVTFLFGYNTGLYLLTAISVERCLΞVLYPIWYRCHRPKHQS AFVCAL ALSC VTTMEYVMCIDSGEESHΞRSDCRAVIIFIAILSF VFTP MLVSSTI WKIRKNT ASHS SKLYIVIMVTIIIFLIFAMPMRV YL YYEY SAFGN HNIS LFSTINSSANPFIYFFVGΞSKKKRFRESLKV V TRAFKDE QPRRQEGNGNTVSIETW.

Claims

What is claimed is:
1. An isolated polynucleotide selected from the group consisting of:
(I) an isolated polynucleotide comprising a nucleotide sequence encoding a polypeptide having at least a 95% identity to the ammo acid sequence of SEQ ID NO:2, over the entire length of SEQ ID NO:2;
(n) an isolated polynucleotide comprising a nucleotide sequence having at least a 95% identity over its entire length to a nucleotide sequence encoding the polypeptide of SEQ ID
NO:2;
(m) an isolated polynucleotide comprising a nucleotide sequence having at least a 95% identity to that of SEQ ID NO: 1 over the entire length of SEQ ID NO: 1 ;
(iv) an isolated polynucleotide comprising a nucleotide sequence encoding the polypeptide of SEQ ID NO:2;
(v) an isolated polynucleotide that is the polynucleotide of SEQ ID NO: 1 ; and
(vi) an isolated polynucleotide with a nucleotide sequence of at least 100 nucleotides m length obtained by screening an appropriate library under stringent hybridization conditions with a labeled probe having the sequence of SEQ ID NO: l or a fragment thereof of at least
15 nucleotides; or a nucleotide sequence complementary to said isolated polynucleotide.
2. An isolated polypeptide selected from the group consisting of:
(l) an isolated polypeptide having at least a 95% identity to the amino acid sequence of SEQ ID NO: 2 over the entire length of SEQ ID NO:2;
(π) an isolated polypeptide comprising the ammo acid sequence of SEQ ID NO.2; and (in) an isolated polypeptide that has the ammo acid sequence of SEQ ID NO:2.
3. A method for screening to identify compounds that stimulate or that inhibit a function or level of the polypeptide of Claim 2, comprising a method selected from the group consisting of:
(a) measurmg or, quantitatively or qualitatively, detecting the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound;
(b) measuring the competition of the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof in the presence of a labeled competitor;
(c) testing whether the candidate compound results m a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells or cell membranes bearing the polypeptide; (d) mixing a candidate compound with a solution comprising a polypeptide of Claim 2. to form a mixture, measuring activity of the polypeptide in the mixture, and comparing the activity of the mixture to a control mixture that contains no candidate compound; and
(e) detecting the effect of a candidate compound on the production of mRNA encoding said polypeptide and said polypeptide in cells.
4. An agonist or an antagonist of a polypeptide of Claim 2.
5. An agonist or an antagonist of the Mus musculus mas identified by the method of Claim 3.
6. An expression system comprising a polynucleotide capable of producing a polypeptide of Claim 2 when said expression system is present in a compatible host cell.
7. A process for producing a recombinant host cell comprising the step of introducing the expression vector of Claim 6 into a cell, such that the host cell, under appropriate culture conditions, produces said polypeptide.
8. A recombinant host cell produced by the process of Claim 7.
9. A membrane of a recombinant host cell of Claim 8 expressing said polypeptide.
10. A process for producing a polypeptide comprising culturmg a host cell of Claim 8 under conditions sufficient for the production of said polypeptide and recovering the polypeptide from the culture.
PCT/US2001/000054 2000-01-04 2001-01-03 Mouse g-protein coupled receptor mas WO2001049744A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US17449900P 2000-01-04 2000-01-04
US60/174,499 2000-01-04
US74250800A 2000-12-21 2000-12-21
US09/742,508 2000-12-21

Publications (1)

Publication Number Publication Date
WO2001049744A1 true WO2001049744A1 (en) 2001-07-12

Family

ID=26870293

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/000054 WO2001049744A1 (en) 2000-01-04 2001-01-03 Mouse g-protein coupled receptor mas

Country Status (1)

Country Link
WO (1) WO2001049744A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007121546A1 (en) * 2006-04-26 2007-11-01 Universidade Federal De Minas Gerais Use of mas-g-protein-coupled receptor agonists in the treatment of the metabolic syndrome, its components and its complications

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0711831A2 (en) * 1994-11-14 1996-05-15 Takeda Chemical Industries, Ltd. Rabbit G protein coupled receptor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0711831A2 (en) * 1994-11-14 1996-05-15 Takeda Chemical Industries, Ltd. Rabbit G protein coupled receptor

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007121546A1 (en) * 2006-04-26 2007-11-01 Universidade Federal De Minas Gerais Use of mas-g-protein-coupled receptor agonists in the treatment of the metabolic syndrome, its components and its complications
US8586054B2 (en) 2006-04-26 2013-11-19 Max-Delbruck-Centrum Fur Molekulare Medizin Method for modulating, treating and/or preventing metabolic syndrome using MAS-G-protein-coupled receptor agonists

Similar Documents

Publication Publication Date Title
US6881827B2 (en) Molecular cloning of a 7TM receptor (AXOR34) and screening methods thereof
WO2000049170A1 (en) MURINE 11cby RECEPTOR
WO2000078809A1 (en) Axor16, a g protein coupled receptor
US20020106766A1 (en) Molecular cloning of a galanine-like 7 transmembrane receptor (AXOR12 RAT)
US20020090691A1 (en) Mouse seven trans-membrane receptor EDG4
US6428982B1 (en) Polynucleotides encoding mouse urotensin-II Receptor (UTB-R)
WO2001011022A1 (en) Mouse g-protein coupled receptor edg3
US20020061558A1 (en) Mouse G protein coupled receptor EDG7
WO2000059529A1 (en) Mouse edg1
WO2001027153A1 (en) A murine seven-transmembrane receptor, mus musculus mhneaa81
WO2001010889A1 (en) Rat-g-protein coupled receptor brs3
EP1194551A1 (en) G-protein coupled receptor and dna sequences thereof
WO2001049744A1 (en) Mouse g-protein coupled receptor mas
US20020064813A1 (en) Monkey calcium sensing receptor
US20020064814A1 (en) Dog orexin 1 receptor
WO2001007482A1 (en) Gpr27, a g-protein coupled receptor
WO2000068244A1 (en) 7tm receptor mouse apj
US20020137136A1 (en) Rat G protein coupled receptor, EDG6
EP1171467A1 (en) Axor-27, a g-protein coupled receptor
WO2001040259A2 (en) Monkey orexin 1 receptor
US6355452B1 (en) Human histamine H3 gene variant-2
WO2001068816A1 (en) Human histamine h3 gene variant-3
US20020045216A1 (en) Mouse G protein coupled receptor FMRL-3
US20020045214A1 (en) Rat G-protein coupled receptor AXOR29
WO2001027137A2 (en) Rattus norvegicus (edg3)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): GB JP

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP