EP1179006A2 - Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations - Google Patents

Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations

Info

Publication number
EP1179006A2
EP1179006A2 EP00930459A EP00930459A EP1179006A2 EP 1179006 A2 EP1179006 A2 EP 1179006A2 EP 00930459 A EP00930459 A EP 00930459A EP 00930459 A EP00930459 A EP 00930459A EP 1179006 A2 EP1179006 A2 EP 1179006A2
Authority
EP
European Patent Office
Prior art keywords
gpm
protein
seq
cell
nucleic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00930459A
Other languages
German (de)
English (en)
Other versions
EP1179006A4 (fr
Inventor
Stephen M. Lanier
Aya Takesono
Mary Cismowski
Emir Duzic
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
MUSC Foundation for Research Development
OSI Pharmaceuticals LLC
Original Assignee
MUSC Foundation for Research Development
OSI Pharmaceuticals LLC
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 MUSC Foundation for Research Development, OSI Pharmaceuticals LLC filed Critical MUSC Foundation for Research Development
Publication of EP1179006A2 publication Critical patent/EP1179006A2/fr
Publication of EP1179006A4 publication Critical patent/EP1179006A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

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

Definitions

  • G proteins guanine nucleotide binding proteins
  • Membrane receptors G protein-coupled receptors, sensing external stimuli activate heterotrimeric G (G ⁇ ) by facilitating nucleotide exchange and subsequent dissociation of G ⁇ from G ⁇ .
  • a first line of evidence is the fact that a single G protein coupled receptor can couple to multiple effectors.
  • the ⁇ 2A -AR a typical G protein coupled receptor, may regulate phospholipases A 2 , C and D. calcium flux, Na+/H+ exchangers, p21-ras or adenylyl cyclases. Marjamaki et.al. (1997) J. Biol Chem. 272:16466-16473. The specific regulation of any of these effector molecules often depends upon receptor subtype, receptor density and the environment in which the receptor is operating.
  • a third line of evidence relates to the transfer of signal from receptor to G- protein in a reconstituted system.
  • G-protein activation occurs with homogenous preparations of receptor and G-protein following reconstitution in phospholipid vesicles suggesting that initiation of the cellular response to a biological stimuli involves only receptor and G-protein.
  • the efficiency with which the receptor mediates the agonist-induced activation of G-protein and subsequent effector regulation/signal termination is often dramatically less when the purified entities are reconstituted in phospholipid vesicles as compared to what is observed or expected to occur in the intact cell or membrane preparations. Such a discrepancy may reflect technical issues associated with reconstitution studies or the absence of other cellular factors required for driving these events.
  • a fourth line of evidence relates to the ability of nonreceptor proteins/peptides to associate with and regulate the activation state of specific G-proteins. Sato et.al. (1996) J. Biol. Chem. 271 :30052-30060. Overall, these observations indicate the existence of accessory proteins that influence events at the receptor/G-protein or G-protein/effector interface and regulate the key steps of nucleotide exchange and/or GTP hydrolysis. In fact, recent observations in cells transfected with genes encoding components of the signaling cascade or in unperturbed signaling systems endogenous to the cell suggest the existence of cellular factors that augment, direct or brake the transfer of signal from receptor to G-protein. Sato et.al. (1995) J. Biol. Chem. 270:15269-15276; Nanoff et.al.
  • GPM G Protein Modulator
  • an isolated nucleic acid molecule of the present invention has a nucleotide sequence encoding a G protein modulator (GPM) protein (e.g., a nucleotide sequence selected form the group consisting of SEQ ID NO:l , SEQ ID NO:4 and SEQ ID NO:6).
  • GPM G protein modulator
  • the invention also provides for vectors including nucleotide sequences which encode a GPM, as well as host cells including such vectors.
  • Methods for producing GPM proteins comprising culturing such host cell are also provided by the present invention.
  • an isolated protein of the present invention is a G protein modulator (GPM) protein (e.g. , a GPM protein having the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO:5).
  • GPM G protein modulator
  • the invention also provides for pharmaceutical compositions including GPM proteins, fusion proteins including all or a portion of the GPM proteins or the present invention, antibodies that specifically bind GPM proteins (e.g., monoclonal or polyclonal antibodies) as well as pharmaceutical compositions including such antibodies.
  • a method comprises the steps of (a) contacting a cell that expresses a G protein modulator (GPM) protein with a test compound; (b) determining the effect of the test compound on the activity of the GPM protein; and (c) identifying the test compound as a modulator of signal transduction based on the ability of the compound to modulate the activity of the GPM protein in the cell.
  • GPM proteins utilized have an amino acid sequence at least 75% homologous to SEQ ID NO:2 or SEQ ID NO:5.
  • the GPM proteins utilized have an amino acid sequence at least 75% homologous to SEQ ⁇ D NO:2 or SEQ ID NO:5 and comprises at least one tetratricopeptide repeat ("TPR") motif.
  • the GPM proteins utilized have an amino acid sequence at least 75% homologous to SEQ ID NO:2 or SEQ ID NO:5 and comprises at least one G protein regulatory motif (also referred to herein as a "DDQR" repeat motif or "DDQR" motif.
  • the GPM proteins utilized have an amino acid sequence at least 75% homologous to SEQ ID NO:2 or SEQ ID NO:5, comprises at least one G protein regulatory motif (e.g., a motif having the amino acid sequence of SEQ ID NO:8) and stimulate G protein activity in a receptor-independent manner.
  • G protein regulatory motif e.g., a motif having the amino acid sequence of SEQ ID NO:8
  • cells used in assay methods of the present invention have been engineered to express a GPM (e.g., a GPM protein having the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO:5).
  • Preferred cells for use in the assay are yeast cells.
  • the yeast cells have further been engineered to express a G protein ⁇ subunit, a chimeric G protein ⁇ subunit, or a Gpal- G ⁇ i2 chimeric G protein ⁇ subunit.
  • the activity of a test compound on a GPM of the present invention can be monitored, for example, by measuring the activity of a pheromone responsive promoter in the yeast cells.
  • the activity of a test compound on a GPM can be determined by monitoring the ability of the test compound to bind to the GPM protein or by monitoring the ability of the test compound to modulate the interaction of the GPM protein with a target molecule (e.g., a G protein).
  • the present invention further provides methods for modulating a cell-associated activity (e.g., by contacting a cell with an agent which modulates GPM protein activity or GPM nucleic acid expression).
  • methods for treating subjects having a disorder characterized by aberrant GPM protein activity or nucleic acid expression are provided, as well as methods for detecting the presence of GPM in a biological sample.
  • Figure 1 depicts the nucleotide sequence of a GPM II cDNA isolated from the neuroblastoma glioma hybrid cell line NG-108 (SEQ ID NO:l) as well as the predicted amino acid sequences of four peptides (SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, and SEQ ID NO: 13) encoded by the GPM II cDNA.
  • Figure 2 depicts the nucleotide sequence of a GPM II cDNA isolated from a rat brain cDNA library (SEQ ID NO:5) as well as the predicted amino acid sequence amino acid sequence (SEQ ID NO:6).
  • Figure 3 depicts the nucleotide sequence of a GPM I cDNA isolated from the neuroblastoma glioma hybrid cell line NG-108 (SEQ ID NO:7) as well as the predicted amino acid sequence amino acid sequence (SEQ ID NO:8).
  • Figure 4 demonstrates the structural analysis of GPM II.
  • Figure 4 A depicts an alignment of the GPM II amino acid sequence with the human LGN protein (having Accession No.U54999) and the C. elegans protein (having Accession No.U40409) by PILEUP (University of Wisconsin GCG program). Amino acid sequence similarity and identity are indicated below the three sequences by + or residue, respectively.
  • the TPR and DDQR repeat motifs are indicated by shaded and underlined sequences, respectively.
  • TPR refers to the tetratricopeptide repeat motif.
  • Figure 4B depicts the four DDQR repeat domains in the carboxyl terminus half of GPM II, human LGN, the predicted C. elegans protein and related sequences identified by the motif search program MEME.
  • Figure 5 demonstrates the bioactivity of full-length versus truncated GPM II.
  • Figure 5 A depicts a spot growth assay of yeast strain transformed with control plasmid, truncated GPM II (also referred to herein as Activator of G protein Signaling 3 or "AGS3"), or full-length GPM-II/AGS3.
  • Figure 5B depicts an immunoprecipitation of GPM II/AGS3 protein from transformed yeast strains
  • Figure 6 demonstrates the interaction of AGS proteins with G-protein subunits and the relationship of AGS bioactivity to nucleotide binding properties of the G ⁇ subunit.
  • Figure 6A depicts binding of full length GPM I (also referred to herein as "AGS2") and a 74 amino acid peptide beginning at the first in frame MET of frame 1 in the NG108-15 cDNA clone #53 [AGS3 (M577-S650)] to various G-protein subunits (60-80 nM G ⁇ or 40 nM G ⁇ ). Proteins are adsorbed to glutathione matrix and retained G-protein subunits identified by immunoblotting following gel electrophoresis.
  • Figure 6B depicts the effect of GPM proteins on nucleotide binding to purified brain G-protein.
  • Figure 6C depicts the effect of G204A Gi ⁇ 2 substitution and RGS4 on the functionality of GPM proteins as determined by spot growth assays.
  • the present invention is based on the discovery of nucleic acid and protein molecules, referred to herein as G-protein Modulator ("GPM”) nucleic acid and protein molecules, which play a role in or function in G protein-mediated signal transduction in the absence of receptor stimulation.
  • GPM G-protein Modulator
  • the GPM molecules stimulate the activity of one or more G proteins involved in a G protein-mediated signal transduction pathway, e.g., a pheromone response cascade in yeast, to thereby activate G protein-mediated signal transduction independent of G protein-coupled receptor stimulation.
  • the GPM molecules of the present invention stimulate the activity of one or more G proteins involved in a G protein-mediated signal transduction pathway, such that G protein coupled receptor-mediated signal transduction is amplified.
  • the GPM molecules are capable of modulating the activity of G ⁇ subunits, such as a yeast Gpal protein, a mammalian G ⁇ i2 subunit, or a chimeric G ⁇ subunit comprising a portion of the yeast Gpal protein (e.g., the amino-terminal 41 amino acids) linked to a mammalian G ⁇ i2 subunit.
  • G ⁇ subunits such as a yeast Gpal protein, a mammalian G ⁇ i2 subunit, or a chimeric G ⁇ subunit comprising a portion of the yeast Gpal protein (e.g., the amino-terminal 41 amino acids) linked to a mammalian G ⁇ i2 subunit.
  • a GPM protein can include one or more tetratricopeptide repeat ("TPR) motifs.
  • TPR motif is an amino acid motif within a protein which comprises an amino acid sequence having a length of about 5-50 amino acid residues, preferably about 10-45, more preferably about 15-35, and more preferably about 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33 or 34 amino acid residues that has at least 50% amino acid homology, preferably about 60-70%, more preferably about 70-80%, and more preferably about 80-90% amino acid homology with a second TPR motif within the same protein which is separated by about 30-1 10 amino acid residues, preferably about 40-100 amino acid residues.
  • a first TPR motif can be separated form a second TPR motif within the same protein by about 60, 70, 80 or 90 amino acid residues.
  • a TPR motif includes a highly degenerate motif of about 34 ammo acid residues.
  • TPR motifs have been found in as many as 400 proteins and can be present in 3-16 copies per protein (alternatively 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14 or 15 copies per protein) as described, for example, in Chen and Cohen (1997) FEBS Letters 400:136-140; Das et ⁇ l. (1998) EMBO J. 17:1 192-1 199 (1998); and Ramarao and Cohen (1998) Proc. N ⁇ tl. Ac ⁇ d. Sci., USA 95:4007-4012.
  • Particularly preferred TPR motifs can be identified using SMART analysis and are set forth in Figure 4 as they occur in GPM II (AGS3) and human LGN (having Accession No. U54999).
  • a GPM protein can include one or more G protein regulatory ("GPR") motifs, also referred to herein as "DDRQ repeat” motifs or “DDRQ” motifs.
  • GPR motif is an amino acid motif within a protein which comprises an amino acid sequence having a length of about 5-35 amino acid residues, preferably about 10-30, more preferably about 15-25, and more_ ⁇ jreferably about 17, 18, 19, 20, 21, 22 or 23 amino acid residues that has at least 50% amino acid homology, preferably about 60-70%, more preferably about 70-80%, and more preferably about 80-90% amino acid homology with a second GPR motif within the same protein.
  • a GPR core consensus sequence has the sequence [DE]EX[FL]F[DE][LM][LI]X[RK]XQ[SG]XR[ML]DDQR. Amino acid sequences having all or a majority of the amino acids of the consensus sequences defined herein may be recognized as potentially having a specified motif.
  • amino acid sequence having all but one amino acid residue of a consensus sequence may be recognized by one of skill in the art as potentially having such a motif.
  • amino acid sequence having all but two amino acid residues of a consensus sequence may be recognized by one of skill in the art as potentially having such a motif.
  • a GPR motif has the core consensus sequence, [DE](2)FF X(10-12)-D[DE]QR (SEQ ID NO:12).
  • a GPR motif has the core majority profile EEFF-X(10-12)-DDQR (SEQ ID NO: 1 1).
  • a GPR motif has the consensus sequence [DE]-XFFX-[LIVM](2)-X(3)-Q-X(2)-R-[LIVM][D/E](2)-QR (SEQ ID NO: 12).
  • a GPR motif has the majority profile R-PS— -P- EEFFDL-K-QSSRMDD QRCPL— P- --AT (SEQ ID NO:9). It is suggested that the above defined consensus sequence(s) arranged in repeats with a defined spatial relationship constitute a structural feature found at the carboxy terminus of a novel family of proteins that interact with G-proteins and regulate nucleotide exchange/GTP hydrolysis or the interaction of the G-protein with other entities involved in signal transduction.
  • a GPM protein includes at least one G protein regulator ("GPR") motif wherein the GPR motif has at least 50% amino acid homology, preferably about 60-70%, more preferably about 70-80%, and more preferably about 80- 90% amino acid homology with a GPR motif consensus sequence (e.g.. the consensus sequence of SEQ ID NO:9, SEQ ID NO: 10, or SEQ ID NO:l 1).
  • GPR G protein regulator
  • a GPM protein includes at least one G protein regulatory (“GPR") motif wherein the GPR motif has at least 50% amino acid homology, preferably about 60- 70%, more preferably about 70-80%, and more preferably about 80-90% amino acid homology with a GPR motif of GPM II, as described below.
  • FIG. 1 depicts the nucleotide sequence of a GPM II cDNA which was isolated from the neuroblastoma glioma hybrid cell line NG-108.
  • the predicted amino acid sequences of three peptides encoded by the GPM II cDNA are also depicted in Figure 1.
  • a first open reading frame can be found beginning with the first nucleotide of SEQ ID NO: l and ending with the stop codon, TAA at nucleotides 253-255 of SEQ ID NO:l .
  • a GPM II protein is encoded by a nucleic acid sequence comprising nucleotides 1-252 of SEQ ID NO:l ("coding region") and comprises amino acids 1 to 84 of SEQ ID NO:2.
  • This peptide (Frame 1, Peptide A of Figure 1) comprises at least two GPRs at about amino acids 6-25 and from about 40-59 of SEQ ID NO:2.
  • a GPM II protein (Frame 1, Peptide B of Figure 1) is encoded by a nucleic acid sequence comprising nucleotides 31-252 of SEQ ID NO:l ("coding region") and comprises amino acids 1 to 74 of SEQ ID NO:3.
  • This peptide comprises at least one GPR at about amino acids 30-49 of SEQ ID NO:3.
  • a second open reading frame can be found beginning with the second nucleotide of SEQ ID NO:l and ending with the stop codon, TGA at nucleotides 359-361 of SEQ ID NO: l .
  • a GPM II protein is encoded by a nucleic acid sequence comprising nucleotides 2-358 of SEQ ID NO:l ("coding region") and comprises amino acids 1 to 1 19 of SEQ ID NO:4.
  • a GPM II protein is encoded by a nucleic acid sequence comprising nucleotides 17-358 of SEQ ID NO: 1 and comprises amino acids 1 to 1 14 of SEQ ID NO: 13.
  • Figure 2 depicts the nucleotide sequence of a second GPM II cDNA which was isolated from a rat brain cDNA library.
  • the cDNA encodes a predicted protein which is at least 590 amino acid residues in length and contains at least four GPR motifs at about amino acids 469-490, 524-544, 572-591, and 606-625 of SEQ ID NO:6.
  • the cDNA encodes a predicted protein which contains at least four GPR motifs at about amino acids 465-492, 519-544, 570-592, and 604-627 of SEQ ID NO:6.
  • GPM II has been found to be expressed in mRNA isolated from rat brain, rat vascular smooth muscle cells, the NG108-15 cell line and the C6 -B4 glioma cell line but not in rat liver.
  • GPM proteins e.g., GPMJ.L are involved in activation of the pheromone response cascade in yeast cells, and potentially modulate the MEK pathway in mammalian cells
  • the GPM molecules of the present invention can be used in methods for identifying antagonists of G protein signaling, either receptor-dependent or receptor independent.
  • the GPM II nuclejc acid molecule of SEQ ID NO:l was identified from an NG108-15 cDNA expression library based on its ability to stimulate G protein signaling in yeast in the absence of a G protein-coupled receptor, as determined using a high throughput library screening method for non-receptor activators of the pheromone response cascade (described in detail in Examples 1 and 2).
  • a GenBankTM search using the GPM nucleotide sequence of SEQ ID NO:l revealed two EST clones, both of murine origin, which were similar to different regions of the nucleotide sequence of SEQ ID NO:l .
  • the mouse EST, va71fl0.rl (Accession Number AA271904), is identical to a portion (from about nucleotides 971 to 1397) of the 3' untranslated sequence of SEQ ID NO: l .
  • the mouse EST, mi70b09.rl (Accession Number AA036169), is identical to a portion (from about nucleotides 853 to 1358) of the 3' untranslated sequence of SEQ ID NO: 1.
  • GenBankTM search using the GPM nucleotide sequence of SEQ ID NO: l also revealed a murine partial cDNA referred to as mouse C10A (Accession Number L23316).
  • the first 333 nucleotides of the GPM II cDNA exhibit significant homology (one nucleotide difference) to the 3'end of the mouse C10A cDNA.
  • the human polypeptide sequence, LGN named for the existence of at least 10 leu-gly-asn repeats, (GenPeptTM Accession Number U54999), includes at least four GPR motifs which are approximately 50-85% homologous to the GPR motifs of SEQ ID NO:6. (Described in detail in Example 3).
  • elegans open reading frame (Accession Number U40409) also includes a similar four domain motif.
  • the present invention also pertains to proteins which have an amino acid sequence which is substantially homologous to the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, or SEQ ID NO:6, comprise at least four GPR motifs, and have a GPM activity.
  • a protein which has an amino acid sequence which is substantially homologous to a selected amino_ac id sequence is at least 50% homologous to the selected amino acid sequence, e.g., the entire selected amino acid sequence.
  • a protein which has an amino acid sequence which is substantially homologous to a selected amino acid sequence can also be least 60%, preferably at least 70%, more preferably at least 80%, and even more preferably at least 85, 86, 88, 90%, and most preferably at least 95%, 96, 97, 98, or 99% homologous to a selected amino acid sequence.
  • the GPM protein or a biologically active portion or fragment of the invention can have one or more of the following activities: 1) it can interact with (e.g., bind to) a G protein; 2) it can modulate the activity of a G protein; 3) it can interact with (e.g., bind to) a G protein target molecule; 4) it can modulate the activity of a G protein target molecule; 5) it can modulate a G protein-mediated response in a cell, independent of G protein-coupled receptor activation; and 6) it can augment G protein-coupled receptor signaling by modulating a G protein-mediated response in a cell.
  • nucleic acid molecules that encode a GPM protein or biologically active portion thereof, as well as nucleic acid fragments sufficient for use as hybridization probes to identify GPM-encoding nucleic acid (e.g., GPM mRNA).
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • the nucleic acid molecule can be single-stranded or double-strauded, but preferably is double-stranded DNA.
  • an “isolated” nucleic acid molecule is one which is separated from other nucleic acid molecules which are present in the natural source of the nucleic acid.
  • an “isolated” nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated GPM nucleic acid molecule can contain less than about 5 kb, 4kb, 3kb, 2kb, 1 kb, 0.5 kb or 0.1 kb of nucleotide sequences which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized.
  • a nucleic acid molecule of the present invention e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO: 1 , SEQ ID NO:5, SEQ ID NO:7, or a portion thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein.
  • a human GPM II cDNA can be isolated from a human neuronal cDNA library using all or portion of SEQ ID NO: 1 as a hybridization probe and standard hybridization techniques (e.g., as described in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual.
  • nucleic acid molecule encompassing all or a portion of SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7 can be isolated by the polymerase chain reaction using oligonucieotide primers designed based upon the sequence of SEQ ID NO: l, SEQ ID NO:5, or SEQ ID NO:7.
  • mRNA can be isolated from normal neuronal cells (e.g., by the guanidinium-thiocyanate extraction procedure of Chirgwin et.al.
  • cDNA can be prepared using reverse transcriptase (e.g., Moloney MLV reverse transcriptase, available from Gibco/BRL, Bethesda, MD; or AMV reverse transcriptase, available from Seikagaku America, Inc., St. Russia, FL).
  • reverse transcriptase e.g., Moloney MLV reverse transcriptase, available from Gibco/BRL, Bethesda, MD; or AMV reverse transcriptase, available from Seikagaku America, Inc., St. Russia, FL.
  • Synthetic oligonucieotide primers for PCR amplification can be designed based upon the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7.
  • a nucleic acid of the invention can be amplified using cDNA or, alternatively, genomic DNA, as a template and appropriate oligonucieotide primers according to standard PCR amplification techniques.
  • oligonucleotides corresponding to a GPM nucleotide sequence can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • an isolated nucleic acid molecule of the invention comprises the nucleotide sequence shown in SEQ ID NO:l.
  • the sequence of SEQ ID NO:l corresponds to a GPM II cDNA isolated from the neuroblastoma glioma hybrid cell line NG-108.
  • This cDNA comprises sequences encoding the NG108-derived GPM II protein.
  • a GPM protein comprises the peptide encoded by nucleotides 1-252 of SEQ ID NO:l, as well as 3' untranslated sequences (nucleotides 253 to 1478 of SEQ ID NO:l).
  • a GPM protein comprises the peptide encoded by nucleotides 31-252 of SEQ ID NO:l, as well as 5' untranslated sequences from nucleotides 1-30 of SEQ ID NO: l and 3' untranslated sequences (nucleotides 253-1478 of SEQ ID NO: l).
  • the nucleic acid nolecule can comprise only coding regions of SEQ ID NO:l (e.g., nucleotides 1 to 252 or 31 to 252 of SEQ ID NO:l).
  • a GPM protein comprises the peptide encoded by nucleotides 2-358 of SEQ ID NO:l, as well as 3' untranslated sequences (nucleotides 359-1478 of SE ID NO:l).
  • an isolated nucleic acid molecule of the invention comprises the nucleotide sequence shown in SEQ ID NO:5.
  • the sequence of SEQ ID NO:5 corresponds to a GPM II cDNA isolated from rat brain.
  • This cDNA comprises sequences encoding the rat brain GPM II protein (i.e., "the coding region", from nucleotides 1 to 1950), as well as 3' untranslated sequences (nucleotides 1951 to - 2386).
  • the nucleic acid molecule can comprise only the coding region of SEQ ID NO:5 (e.g., nucleotides 1 to 1950).
  • an isolated nucleic acid molecule of the invention comprises the nucleotide sequence shown in SEQ ID NO:7.
  • the sequence of SEQ ID NO:7 corresponds to a GPM I cDNA isolated from the neuroblastoma glioma hybrid cell line NG-108.
  • This cDNA comprises sequences encoding the NG108-derived GPM I protein (i.e., "the coding region", from nucleotides 102 to 440), as well as 5' untranslated sequences (nucleotides 1 to 101) and 3' untranslated sequences (nucleotides 441 to 770).
  • the nucleic acid molecule can comprise only the coding region of SEQ ID NO:6 (e.g., nucleotides 102 to 440).
  • an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, or a portion of any of these nucleotide sequences.
  • a nucleic acid molecule which is the complement of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7 is one which has a nucleotide sequence that directly pairs with that of SEQ ID NO: 1 or 3, according to the rules of Watson and Crick base pairing, wherein A pairs with T and G pairs with C.
  • the complement of the sequence 5' GGATGC 3' would be 3'CCTACG 5" (which, written in the standard 5' to 3' direction, would be 5' GCATCC 3').
  • an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which is at least 60%, even more preferably at least 70%, even more preferably at least 80%, and even more preferably at least 90%, 95%, 96%, 97%, 98%, or 99% homologous to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, or a portion of any of these nucleotide sequences.
  • an isolated nucleotide of the present invention can comprise a nucleotide sequence which is at least 60%, preferably at least 70%, more_rjreferably at least 80%, and even more preferably at least 90%, 95%, 96%, 97%, 98%, or 99% homologous to the 3' untranslated region or to the 5' untranslated region of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7.
  • an isolated nucleic acid molecule of the invention comprises a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, or a portion of any of these nucleotide sequences.
  • the nucleic acid molecule of the invention can comprise only a portion of the coding region of SEQ ID NO: l, SEQ ID NO:5, or SEQ ID NO:7, for example a fragment which can be used as a probe or primer or a fragment encoding a biologically active portion of a GPM protein.
  • the nucleotide sequence determined from the cloning of the GPM genes from a mammal, e.g., the mouse allows for the generation of probes and primers designed for use in identifying and/or cloning GPM homologues in other cell types, e.g. from other tissues, as well as GPM homologues from other mammals, e.g., humans.
  • the probe/primer typically comprises substantially purified oligonucieotide.
  • the oligonucieotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12, preferably about 25, more preferably about 40, 50 or 75 consecutive nucleotides of SEQ ID NO:l , SEQ ID NO:5, or SEQ ID NO:7 sense, an anti-sense sequence of SEQ ID NO: l , SEQ ID NO:5, or SEQ ID NO:7, or naturally occurring mutants thereof.
  • Primers based on the nucleotide sequence in SEQ ID NO: 1 , SEQ ID NO:5, or SEQ ID NO:7 can be used in PCR reactions to clone GPM homologues.
  • Probes based on the GPM nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins.
  • the probe further comprises a label group attached thereto, e.g. the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co-factor.
  • Such probes can be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress a GPM protein, such as by measuring a level of a GPM-encoding nucleic acid in a sample of cells from a subject e.g., detecting GPM mRNA levels or determining whether a genomic GPM gene has been mutated or deleted.
  • the nucleic acid molecule of the invention encodes a protein or portion thereof which includes an amino acid sequence which is sufficiently homologous to an amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO: 8 or SEQ ID NO: 13 such that the protein or portion thereof maintains the ability to modulate a G-protein mediated response in a cell.
  • the language "sufficiently homologous” refers to proteins or portions thereof which have amino acid sequences which include a minimum number of identical or equivalent (e.g., an amino acid residue which has a similar side chain as an amino acid residue in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13) amino acid residues to an amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 such that the protein or portion thereof is able to modulate a G-protein mediated response in a cell.
  • a minimum number of identical or equivalent e.g., an amino acid residue which has a similar side chain as an amino acid residue in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13
  • the protein is at least 60%, preferably at least 70%, more preferably at least 80%, more preferably at least 90% and most preferably at least 95, 96, 97, 98, or 99% homologous to the entire amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO:13.
  • Portions of proteins encoded by the GPM nucleic acid molecule of the invention are preferably biologically active portions of the GPM protein.
  • biologically active portion of GPM is intended to include a portion, e.g., a domain/motif, of GPM that has one or more of the following activities: 1) it can interact with (e.g., bind to) a G protein; 2) it can modulate the activity of a G protein; 3) it can interact with (e.g., bind to) a G protein target molecule; 4) it can modulate the activity of a G protein target molecule; 5) it can modulate a G protein-mediated response in a cell, independent of G protein-coupled receptor activation; and 6) it can augment G protein- coupled receptor signaling by modulating a G protein-mediated response in a cell.
  • a biologically active portion of GPM can comprise at least one GPR motif.
  • Standard binding assays e.g., immunoprecipitations and yeast two-hybrid assays as described herein, can be performed to determine the ability of a GPM protein or a biologically active portion thereof to interact with (e.g., b ud lu) a G protein.
  • the GPM protein or biologically active portion thereof can be introduced into a cell (e.g., transformed or transfected) which has been engineered to grow only in the presence of a GPM protein or biologically active portion thereof, e.g., yeast cell strain 1316/1 183 (described in the Examples) and the ability of the GPM protein or biologically active portion thereof to facilitate growth determined.
  • a GPM protein or biologically active portion thereof e.g., yeast cell strain 1316/1 183 (described in the Examples) and the ability of the GPM protein or biologically active portion thereof to facilitate growth determined.
  • a cell can be transformed or transfected with a G-protein mediated signal transduction responsive reporter construct (e.g., FUSl-luciferase) which responds to G- protein mediated signaling by expressing luciferase, and a nucleic acid encoding the GPM protein or biologically active portion thereof.
  • a G-protein mediated signal transduction responsive reporter construct e.g., FUSl-luciferase
  • reporter activity e.g., luciferase activity
  • control cells include cells which include the G- protein mediated signal transduction responsive reporter construct.
  • An alteration in reporter activity in the cells which include nucleic acid encoding the GPM protein, as compared to reporter activity in the cells without nucleic acid encoding the GPM protein is indicative of a modulation of a G-protein mediated response in the cell.
  • GPM nucleotide sequences shown in SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7 DNA sequence polymorphisms that lead to changes in the amino acid sequences of GPM may exist within a population.
  • Such genetic polymorphism in the GPM gene may exist among individuals within a population due to natural allelic variation.
  • the terms "gene” and “recombinant gene” refer to nucleic acid molecules comprising an open reading frame encoding a GPM protein, preferably a mammalian GPM protein.
  • Such natural allelic variations can typically result in 1-5% variance in the nucleotide sequence of the GPM gene.
  • nucleic acid molecules encoding GPM proteins from other species and thus which have a nucleotide sequence which differs from the sequence of SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, are intended to be within the scope of the invention.
  • human homologues of the GPM cDNAs of the invention can be isolated based on their homology to the GPM nucleic acid disclosed herein using the murine cDNAs, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions.
  • an isolated nucleic acid molecule of the invention is at least 15 nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7.
  • the nucleic acid is at least 30, 50, 100, 250 or 500 nucleotides in length.
  • hybridizes under stringent conditions is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.
  • the conditions are such that sequences at least 65%, more preferably at least 70%, and even more preferably at least 75% or more homologous to each other typically remain hybridized to each other.
  • stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • a preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45°C, followed by one or more washes in 0.2 X SSC, 0.1% SDS_at 50-65°C.
  • SSC 6X sodium chloride/sodium citrate
  • an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NO:l, SEQ ID NO: 5, or SEQ ID NO: 7 corresponds to a naturally-occurring nucleic acid molecule.
  • a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
  • the nucleic acid encodes a natural human GPM.
  • allelic variants of the GPM sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequence of SEQ ID NO: l, SEQ ID NO:5, or SEQ ID NO:7, thereby leading to changes in the amino acid sequence of the encoded GPM proteins, without altering the functional activity of the GPM proteins.
  • nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NO:l , SEQ ID NO:5, or SEQ ID NO:7.
  • non-essential amino acid residue is a residue that can be altered from the wild-type sequences of GPM (e.g., the sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13) without altering the activity of GPM, whereas an "essential" amino acid residue is required for GPM activity.
  • conserved amino acid residues in the GPR motif of GPM II are most likely important for binding to G proteins and are thus essential residues of GPM II.
  • Other amino acid residues, however, may not be essential for activity and thus are likely to be amenable to alteration without altering GPM activity.
  • GPM proteins that contain changes in amino acid residues that are not essential for GPM activity.
  • GPM proteins differ in amino acid sequence from SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13, yet retain at least one of the GPM activities described herein.
  • the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least 60% homologous to the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO:13, and is capable of modulating a G- protein mediated response in a cell.
  • the protein encoded by the nucleic acid molecule is at least 70% homologous to SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13, more preferably at least 80% homologous, even more preferably at least 90% homologous, and most preferably at least 95, 96, 97, 98, or 99% homologous to SEQ ID NO:2, SEQ ID NO-- SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO:13.
  • sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second a ⁇ iino or nucleic acid sequence and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, even more preferably at least 60%, and even more preferably at least 70%, 80%, or 90% of the length of the reference sequence (e.g., when aligning a second sequence to the GPM amino acid sequence of SEQ ID NO:2 having 84 amino acid residues, at least 25, preferably at least 37, more preferably at least 42, even more preferably at least 50, and even more preferably at least 59, 76, or 76 are aligned).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid "homology” is equivalent to amino acid or nucleic acid "identity”
  • the comparison of sequences and determination of percent homology between two sequences can be accomplished using a mathematical algorithm.
  • a preferred, non- limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl.
  • Gapped BLAST can be utilized as described in Altschul et.al., (1997) Nucleic Acids Research 25(17):3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • XBLAST and NBLAST e.g., XBLAST and NBLAST
  • Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller, CABIOS (1989). Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package.
  • a PAM120 weight residue table When utilizing the ALIGN program for comparing amino acid sequence (or for comparing nucleic acid sequences), a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Alternatively, a PAM 250 residue weight Table, a GAP penalty of 10, and a GAP length penalty of 10 can be used.
  • An isolated nucleic acid molecule encoding a GPM protein homologous to the protein of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13, can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NO: 1 , SEQ ID NO:5, or SEQ ID NO:7, such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein.
  • Mutations can be introduced into SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. Those lamilies include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta- branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • a predicted nonessential amino acid residue in a GPM protein is preferably replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of a GPM coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for a GPM activity described herein to identify mutants that retain GPM activity.
  • the encoded protein can be expressed recombinantly and the activity of the protein can be determined using, for example, assays described herein.
  • an antisense nucleic acid comprises a nucleotide sequence which is complementary to a "sense" nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid.
  • the antisense nucleic acid can be complementary to an entire GPM coding strand, or to only a portion thereof.
  • an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding a GPM protein.
  • coding region refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues.
  • the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding GPM.
  • noncoding region refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions).
  • antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing.
  • the antisense nucleic acid molecule can be complementary to the entire coding region of a GPM mRNA, but more preferably is an oligonucieotide which is antisense to only a portion of the coding or noncoding region of a GPM mRNA.
  • the antisense oligonucieotide can be complementary to the region surrounding the translation start site of a GPM mRNA.
  • An antisense oligonucieotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucieotide
  • an antisense nucleic acid e.g., an antisense oligonucieotide
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil,_5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5- (carboxyhydroxylmethyl) uracil, 5-carboxymethylaminornethyl-2-thiouridine, 5- carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1 -methylguanine, 1 -methylinosine, 2,2-dimethylguanine, 2- methyladenine, 2-methylg ⁇ anine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7- methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta- D-mannosylque
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding a GPM protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • An example of a route of administration of an antisense nucleic acid molecule of the invention includes direct injection at a tissue site.
  • an antisense nucleic acid molecule can be modified to target selected cells and then administered systemically.
  • an antisense molecule can be modified such that it specifically binds to a receptor or an antigen expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecule to a peptide or an antibody which binds to a cell surface receptor or antigen.
  • the antisense nucleic acid molecule can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
  • the antisense nucleic acid molecule of the invention is an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which ⁇ contrary to the usual ⁇ -units, the strands run parallel to each other (Gaultier et.al. (1987) Nucleic Acids. Res. 15:6625-6641).
  • the antisense nucleic acid molecule can also comprise a 2'-o- methylribonucleotide (Inoue et. al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et.al. (1987) FEBS Lett. 215:327-330).
  • an antisense nucleic acid of the invention is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)
  • a ribozyme having specificity for a GPM-encoding nucleic acid can be designed based upon the nucleotide sequence of a GPM cD ⁇ A disclosed herein.
  • a derivative of a Tetrahymena L-19 IVS R ⁇ A can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in a GPM-encoding mR ⁇ A. See, e.g., Cech et.al. U.S. Patent No. 4,987,071 and Cech et.al. U.S. Patent No. 5,1 16,742.
  • GPM mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel, D. and Szostak, J.W. (1993) Science 261 :141 1-1418.
  • GPM gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of a GPM gene (e.g., a GPM promoter and/or enhancer) to form triple helical structures that prevent transcription of a GPM gene in target cells.
  • a GPM gene e.g., a GPM promoter and/or enhancer
  • nucleotide sequences complementary to the regulatory region of a GPM gene e.g., a GPM promoter and/or enhancer
  • vectors preferably expression vectors, containing a nucleic acid encoding a GPM protein (or a portion thereof).
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • Other vectors are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as "expression vectors".
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector can be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno- associated viruses), which serve equivalent functions.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid sequence to be expressed.
  • "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to includes promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in mrmv types of host cell and those which direct expression of the nucleotide sequence o.-ly in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., GPM proteins, mutant forms of GPM proteins, fusion proteins, etc.).
  • the recombinant expression vectors of the invention can be designed for expression of GPM proteins in prokaryotic or eukaryotic cells.
  • a GPM protein can be expressed in bacterial cells such as E. coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three purposes: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa. thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D.B. and Johnson, K.S. (1988) Gene 67:31-40), pMAL (New England Biolabs.
  • GPM protein is cloned into a pGEX expression vector to create a vector encoding a fusion protein comprising, from the N- terminus to the C-terminus, GST-thrombin cleavage site-GPM protein.
  • the fusion protein can be purified by affinity chromatography using glutathione-agarose resin. Recombinant GPM protein unfused to GST can be recovered by cleavage of the fusion protein with thrombin.
  • Suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et.al., (1988) Gene 69:301-315) and pET l id (Studier et.al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 60-89).
  • Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid trp-lac fusion promoter.
  • Target gene expression from the pET 1 Id vector relies on transcription from a T7 gnl 0-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident ⁇ prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
  • One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically_cleave the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 119-128).
  • Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et.al. (1992) Nucleic Acids Res. 20:21 1 1-21 18).
  • Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • a GPM expression vector is a yeast expression vector.
  • yeast expression vectors for expression in yeast S. cerivisae include pYepSecl (Baldari, et.al., (1987) Embo J. 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933- 943), pJRY88 (Schultz et.al., (1987) Gene 54: 1 13-123), and pYES2 (Invitrogen Corporation, San Diego, CA).
  • a GPM protein can be expressed in insect cells using baculovirus expression vectors.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et.al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, B. (1987) Nature 329:840) and pMT2PC (Kaufman et.al.
  • the expression vector's control functions are often provided by viral regulatory elements.
  • viral regulatory elements For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.
  • the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements are known in the art.
  • suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et.al. (1987) Genes Dev. 1 :268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBO J.
  • promoters are also encompassed, for example the murine hox promoters (Kessel and Gruss (1990) Science 249:374-379) and the ⁇ -fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
  • the invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to a GPM mRNA.
  • Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • a high efficiency regulatory region the activity of which can be determined by the cell type into which the vector is introduced.
  • host cell and “recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • GPM protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et.al. (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding GPM or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) a GPM protein.
  • the invention further provides methods for producing GPM proteins using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a GPM protein has been introduced) in a suitable medium until GPM protein i. produced.
  • the method further comprises isolating GPM protein from the medium or the host cell.
  • the host cells of the invention can also be used to produce nonhuman transgenic animals.
  • the nonhuman transgenic animals can be used in screening assays designed to identify agents or compounds, e.g., drugs, pharmaceuticals, etc., which are capable of ameliorating detrimental symptoms of selected disorders such as cardiovascular disorders and proliferative disorders.
  • a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which GPM-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous GPM sequences have been introduced into their genome or homologous recombinant animals in which endogenous GPM sequences have been altered.
  • transgenic animal is a nonhuman animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene.
  • Other examples of transgenic animals include nonhuman primates, sheep, dogs, cows, goats, chickens, amphibians, etc.
  • a transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal.
  • a "homologous recombinant animal” is a nonhuman animal, preferably a mammal, more preferably a mouse, in which an endogenous GPM gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
  • a transgenic animal of the invention can be created by introducing GPM- encoding nucleic acid into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retroviral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal.
  • the GPM cDNA sequence of SEQ ID NO: l can be used as a transgene.
  • a human homologue of the GPM genes can be isolated based on hybridization to GPM cDNAs (e.g., hybridization to SEQ ID NO:l, SEQ ID NO:5, or SEQ ID NO:7, described further in subsection I above) and used as a transgene.
  • Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene.
  • a tissue-specific regulatory sequence(s) can be operably linked to a GPM transgene to direct expression of GPM protein to particular cells.
  • transgenic founder animal can be identified based upon the presence of a GPM transgene in its genome and/or expression of GPM mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene encoding GPM can further be bred to other transgenic animals carrying other transgenes.
  • a vector is prepared which contains at least a portion of a GPM gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the GPM gene.
  • the GPM gene can be a human gene (e.g., isolated by hybridization to SEQ ID NO:l , SEQ ID NO:5, or SEQ ID NO:7), but more preferably, is a nonhuman homologue of a huma GPM gene (e.g., SEQ ID NOT , SEQ ID NO:5, or SEQ ID NO:7).
  • a mouse GPM gene can be used to construct a homologous recombination vector suitable for altering an endogenous GPM gene in the mouse genome.
  • the vector is designed such that, upon homologous recombination, the endogenous GPM gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
  • the vector can be designed such that, upon homologous recombination, the endogenous GPM gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous GPM protein).
  • the altered portion of the GPM gene is flanked at its 5' and 3' ends by additional nucleic acid of the GPM gene to allow for homologous recombination to occur between the exogenous GPM gene carried by the vector and an endogenous GPM gene in an embryonic stem cell.
  • the additional flanking GPM nucleic acid is of sufficient length for successful homologous recombination with the endogenous gene.
  • flanking DNA both at the 5' and 3' ends
  • are included in the vector see e.g., Thomas, K.R. and Capecchi, M. R. (1987) Cell 51 :503 for a description of homologous recombination vectors).
  • the vector is introduced into an embryonic stem cell line (e.g. , by electroporation) and cells in which the introduced GPM gene has homologously recombined with the endogenous GPM gene are selected (see e.g., Li, E. et.al. (1992) Cell 69:915).
  • the selected cells are then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see e.g., Bradley, A. in Teratocarcinomas and Embryonic Stem Cells: A GPMctical Approach, E.J. Robertson, ed. (IRL, Oxford, 1987) pp. 1 13-152).
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term.
  • Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene.
  • Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley, A.
  • transgenic nonhumans animals can be produced which contain selected systems which allow for regulated expression of the transgene.
  • One example of such a system is the cre/loxP recombinase system of bacteriophage PI .
  • cre/loxP recombinase system For a description of the cre/loxP recombinase system, see, e.g., Lakso et.al. (1992) PNAS 89:6232-6236.
  • Another example of a recombinase system is the FLP recombinase system of Saccharomyces ⁇ erevisiae (O'Gorman et.al. (1991) Science 251 :1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required.
  • Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
  • Clones of the nonhuman transgenic animals described herein can also be produced according to the methods described in Wilmut, I. et.al. (1997) Nature 385:810- 813.
  • a cell e.g., a somatic cell, from the transgenic animal can be isolated and induced to exit the growth cycle and enter G 0 phase.
  • the quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated.
  • the reconstructed oocyte is then cultured such that it develops to morula or blastocyst and then transferred to pseudopregnant female foster animal.
  • the offspring borne of this female foster animal will be a clone of the animal from which the cell, e.g., the somatic cell, is isolated.
  • Another aspect of the invention pertains to isolated GPM proteins, and biologically active portions thereof, as well as peptide fragments suitable for use as immunogens to raise anti- GPM antibodies.
  • An "isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material when produced by recombinant DNA techniques, or chemical precursors or other chemicals when chemically synthesized.
  • the language “substantially free of cellular material” includes preparations of GPM protein in which the protein is separated from cellular components of the cells in which it is naturally or recombinantly produced.
  • the language "substantially free of cellular material” includes preparations of GPM protein having less than about 30% (by dry weight) of non- GPM protein (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non- GPM protein, still more preferably less than about 10% of non- GPM protein, and most preferably less than about 5% non- GPM protein.
  • non- GPM protein also referred to herein as a "contaminating protein”
  • the GPM protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents lessjthan about 20%, more preferably less than about 10%. and most preferably less than about 5% of the volume of the protein preparation.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of GPM protein in which the protein is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of GPM protein having less than about 30% (by dry weight) of chemical precursors or non- GPM chemicals, more preferably less than about 20% chemical precursors or non- GPM chemicals, still more preferably less than about 10% chemical precursors or non- GPM chemicals, and most preferably less than about 5% chemical precursors or non- GPM chemicals.
  • isolated proteins or biologically active portions thereof lack contaminating proteins from the same animal from which the GPM protein is derived. Typically, such proteins are produced by recombinant expression of, for example, a huma GPM protein in a nonhuman cell.
  • an isolated GPM protein or a portion thereof of the invention can modulate a G- protein mediated response in a cell.
  • the protein or portion thereof comprises an amino acid sequence which is sufficiently homologous to an amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO: 8 or SEQ ID NO: 13 such that the protein or portion thereof maintains the ability to modulate a G-protein mediated response in a cell.
  • the portion of the protein is preferably a biologically active portion as described herein.
  • the GPM protein has an amino acid sequence shown in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13.
  • the GPM protein has an ammo acid sequence which is encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to the nucleotide sequence of SEQ ID NOT, SEQ ID NO:5, or SEQ ID NO:7.
  • the GPM protein has an amino acid sequence which is encoded by a nucleotide sequence that is at least 60%, preferably at least 70%, more preferably at least 80%, and even more preferably at least 90%, 95, 96, 87, 98, or 99% homologous to the nucleotide sequence of SEQ ID NOT, SEQ ID NO:5, or SEQ ID NO:7.
  • a preferred GPM protein of the present invention also preferably possess at least one of the GPM activities described herein.
  • a preferred GPM protein of the present invention includes an amino acid sequence encoded by a nucleotide sequence which hybridizes, e.g., hybridizes under stringent conditions, to the nucleotide sequence SEQ ID NOT , SEQ ID NO:5, or SEQ ID NO:7 and which can modulate a G-protein mediated response in a cell.
  • the GPM protein is substantially homologous to the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 and retains the functional activity of the protein of SEQ ID NO:2, SEQ ID NQ:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail in subsection I above.
  • the GPM protein is a protein which comprises an amino acid sequence which is at least 60%, preferably at least 70%, more preferably at least 80%, and even more preferably at least 85%, 86, 88, 90%, and most preferably at least 95%, 96, 97, 98, or 99% homologous to the entire amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 and which has at least one of the GPM activities described herein.
  • the invention pertains to a full length human protein which is substantially homologous to the entire amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO:13.
  • Biologically active portions of the GPM protein include peptides comprising amino acid sequences derived from the amino acid sequence of the GPM protein, e.g., the amino acid sequence shown in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 or the amino acid sequence of a protein homologous to the GPM protein, which include less amino acids than the full length GPM protein or the full length protein which is homologous to the GPM protein, and exhibit at least one activity of the GPM protein.
  • biologically active portions comprise a domain or motif, e.g., a GPR, with at least one activity of the GPM protein.
  • the domain is a GPR domain derived from a human and is at least 55%, preferably at least 60%, even more preferably at least 70%, and most preferably at least 80% or more (e.g. 90, 95, 96, 97, 98 or 99%) homologous to SEQ ID NOT or SEQ ID NO:4.
  • the biologically active portion of the protein which includes the GPR motif can modulate the activity of a G-protein.
  • other biologically active portions in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the activities described herein.
  • the biologically active portions of the GPM protein include one or more selected domains/motifs or portions thereof having biological activity.
  • GPM proteins are preferably produced by recombinant DNAjechniques. For example, a nucleic acid molecule encoding the protein is cloned into an expression vector (as described above), the expression vector is introduced into a host cell (as described above) and the GPM protein is expressed in the host cell.
  • the GPM protein can then be isolated from the cells by an appropriate purification scheme using standard protein purification technigues.
  • a GPM protein, polypeptide, or peptide can be synthesized chemically using standard peptide synthesis techniques.
  • native GPM protein can be isolated from cells (e.g., endothelial cells), for example using an anti- GPM antibody (described further below).
  • GPM chimeric or fusion proteins As used herein, a GPM "chimeric protein” or “fusion protein” comprises a GPM polypeptide operatively linked to a non- GPM polypeptide.
  • An " GPM polypeptide” refers to a polypeptide having an amino acid sequence corresponding to GPM
  • a non- GPM polypeptide refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to the GPM protein, e.g., a protein which is different from the GPM protein and which is derived from the same or a different organism.
  • the term "operatively linked" is intended to indicate that the GPM polypeptide and the non- GPM polypeptide are fused in-frame to each other.
  • the non- GPM polypeptide can be fused to the N-terminus or C-terminus of the GPM polypeptide.
  • the fusion protein is a GST- GPM fusion protein in which the GPM sequences axe fused to the C-terminus of the GST sequences. Such fusion proteins can facilitate the purification of recombinant GPM.
  • the fusion protein is a GPM protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g...
  • GPM chimeric or fusion protein of the invention is produced by standard recombinant DNA techniques.
  • DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, for example by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et.al. John Wiley & Sons: 1992).
  • anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence
  • many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide).
  • a GPM- encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the GPM protein.
  • the present invention also pertains to homologues of the GPM proteins which function as either a GPM agonist (mimetic) or a GPM antagonist.
  • the GPM agonists and antagonists stimulate or inhibit, respectively, a subset of the biological activities of the naturally occurring form of the GPM protein.
  • specific biological effects can be elicited by treatment with a homologue of limited function.
  • treatment of a subject with a homologue having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the GPM protein.
  • Homologues of the GPM protein can be generated by mutagenesis, e.g., discrete point mutation or truncation of the GPM protein.
  • the term "homologue” refers to a variant form of the GPM protein which acts as an agonist or antagonist of the activity of the GPM protein.
  • An agonist of the GPM protein can retain substantially the same, or a subset, of the biological activities of the GPM protein.
  • An antagonist of the GPM protein can inhibit one or more of the activities of the naturally occurring form of the GPM protein, by, for example, competitively binding to a G-protein or downstream or upstream member of the pheromone response cascade.
  • homologues of the GPM protein can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of the GPM protein for GPM protein agonist or antagonist activity.
  • a variegated library of GPM variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of GPM variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential GPM sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of GPM sequences therein.
  • a degenerate set of potential GPM sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of GPM sequences therein.
  • libraries of fragments of the GPM protein coding can be used to generate a variegated population of GPM fragments for screening and subsequent selection of homologues of a GPM protein.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of a GPM coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with S 1 nuclease, and ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which encodes N-terminal, C-terminal and internal fragments of various sizes of the GPM protein.
  • Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of GPM homologues.
  • the most widely used techniques, which are amenable to high through-put analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of ti.e encoding the gene whose product was detected.
  • Recursive ensemble mutagenesis (REM) a new technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify GPM homologues (Arkin and Yourvan (1992) PNAS £9:781 1-7815; Delgrave et.al. (1993) Protein Engineering 6(3):327-331).
  • GPM protein or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind GPM using standard techniques for polyclonal and monoclonal antibody preparation.
  • the full-length GPM protein can be used or, alternatively, the invention provides antigenic peptide fragments of GPM for use as immunogens.
  • the antigenic peptide of GPM comprises at least 8 amino acid residues of the amino acid sequence shown in SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 and encompasses_an epitope of GPM such that an antibody raised against the peptide forms a specific immune complex with GPM.
  • the antigenic peptide comprises at least 10 amino acid residues, more preferably at least 15 amino acid residues, even more preferably at least 20 amino acid residues, and most preferably at least 30 amino acid residues.
  • Preferred epitopes encompassed by the antigenic peptide are regions of GPM that are located on the surface of the protein, e.g., hydrophilic regions.
  • a GPM immunogen typically is used to prepare antibodies by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal) with the immunogen.
  • An appropriate immunogenic preparation can contain, for example, recombinantly expressed GPM protein or a chemically synthesized GPM peptide.
  • the preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent. Immunization of a suitable subject with an immunogenic GPM preparation induces a polyclonal anti- GPM antibody response.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, . e. , molecules that contain an antigen binding site which specifically binds (immunoreacts with) an antigen, such as GPM.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab') 2 fragments which can be generated by treating the antibody with an enzyme such as pepsin.
  • the invention provides polyclonal and monoclonal antibodies that bind GPM.
  • monoclonal antibody or
  • monoclonal antibody composition refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of GPM.
  • a monoclonal antibody composition thus typically displays a single binding affinity for a particular GPM protein with which it immunoreacts.
  • Polyclonal anti- GPM antibodies can be prepared as described above by immunizing a suitable subject with a GPM immunogen.
  • the anti- GPM antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized GPM.
  • ELISA enzyme linked immunosorbent assay
  • the antibody molecules directed against GPM can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497) (see also, Brown et.al. (1981) J. Immunol. 127:539-46; Brown et.al. (1980) J. Biol. Chem .255:4980-83; Yeh et.al. (1976) PNAS 76:2927-31 ; and Yeh et.al. (1982) Int. J.
  • an immortal cell line typically a myeloma
  • lymphocytes typically splenocytes
  • the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds GPM.
  • any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating an anti- GPM monoclonal antibody (see, e.g., G. Galfre et.al. (1977) Nature 266:55052; Gefter et.al. Somatic Cell Genet., cited suGPM; Lerner, Yale J. Biol. Med., cited suGPM; Kenneth, Monoclonal Antibodies, cited suGPM).
  • the immortal cell line e.g., a myeloma cell line
  • the immortal cell line is derived from the same mammalian species as the lymphocytes.
  • murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line.
  • Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium").
  • HAT medium culture medium containing hypoxanthine, aminopterin and thymidine
  • Any of a number of myeloma cell lines can be used as a fusion partner according to standard techniques, e.g., the P3-NSl/l-Ag4-l , P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines. These myeloma lines are available from ATCC.
  • HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG").
  • PEG polyethylene glycol
  • Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed).
  • Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind GPM, e.g., using a standard ELISA assay.
  • a monoclonal anti- GPM antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with GPM to thereby isolate immunoglobulin library members that bind GPM.
  • Kits for generating and screening phage display libraries are commercially available (e g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAPTM Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, Ladner et.al U.S.
  • recombinant anti- GPM antibodies such as chime ⁇ c and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in Robinson et al International Application No. PCT/US 86/02269; Akira, et.al European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496, Morrison et al European Patent Application 173.494; Neuberger et.al PCT International Publication No. WO 86/01533; Cabilly et al U.S. Patent No. 4,816,567, Cabilly et al European Patent Application 125,023; Better et.al (1988) Science
  • An anti- GPM antibody (e.g., monoclonal antibody) can be used to isolate GPM by standard techniques, such as affinity chromatography or immunoprecipitation.
  • An anti- GPM antibody can facilitate the purification of natural GPM from cells and of recombinantly produced GPM expressed in host cells.
  • an anti- GPM antibody can be used to detect GPM protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the GPM protein.
  • Anti- GPM antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen.
  • Detection can be facilitated by coupling (i.e. , physically linking) the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 12 V 3
  • compositions The GPM nucleic acid molecules, GPM proteins, GPM modulators, and anti-
  • GPM antibodies also referred to herein as "active compounds" of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject, e.g., a human.
  • Such compositions typically comprise the nucleic acid molecule, protein, modulator, or antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, such media can be used in the compositions of the invention.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a GPM protein or anti- GPM antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • the active compound e.g., a GPM protein or anti- GPM antibody
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol sGPMy from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sGPMys or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides]_or retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • the nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et.al (1994) PNAS 91 :3054-3057).
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • a GPM protein of the invention has one or more of the activitiesjiescribed herein and can thus be used, for example, to screen drugs or compounds which modulate GPM protein activity as well as to treat disorders characterized by insufficient production of GPM protein or production of GPM protein forms which have decreased activity compared to wild type GPM.
  • the isolated nucleic acid molecules of the invention can be used to express GPM protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect GPM mRNA (e.g., in a biological sample) or a genetic lesion in a GPM gene, and to modulate GPM activity, as described further below.
  • the anti- GPM antibodies of the invention can be used to detect and isolate GPM protein and modulate GPM protein activity.
  • the invention provides methods for identifying compounds or agents which modulate GPM protein activity and/or GPM nucleic acid expression. These methods are also referred to herein as drug screening assays and typically include the step of screening a candidate/test compound or agent for the ability to interact with (e.g., bind to) a GPM protein, to modulate the interaction of a GPM protein and a target molecule, and/or to modulate GPM nucleic acid expression and/or GPM protein activity, and or to modulate signal transduction mediated at least in part by a GPM protein.
  • Candidate/test compounds or agents which have one or more of these abilities can be used to develop drugs to treat disorders characterized by aberrant or abnormal GPM protein activity and/or GPM nucleic acid expression.
  • Candidate/test compounds such as small molecules, e.g., small organic molecules, and other drug candidates can be obtained, for example, from combinatorial and natural product libraries.
  • the invention provides ,- n- ⁇ -i ⁇ d for identifying a compound that modulates signal transduction in a cell, comprising: contacting a cell that expresses a G protein modulator (GPM) protein with a test compound; determining the effect of the test compound on the activity of the GPM protein; and identifying the test compound as a modulator of signal transduction based on the ability of the compound to modulate the activity of the GPM protein in the cell.
  • GPM G protein modulator
  • the term "identify” as used in the context of "identifying a compound” refers to the identification of compounds for which an activity as a G protein modulator has not been previously recognized or demonstrated.
  • identifying is intended to refer to identifying, screening and/or selecting of test compound, for example selecting active compounds not previously recognized as a modulator of G protein activity for further analysis and testing.
  • compounds "identified” according to the methods of the present invention can be used as test compounds in a second assay to confirm a G protein modulatory activity.
  • compounds “identified” according to the methods of the present invention can be tested for other desireable activities or can be tested, for example, at varying doses to determine the effecacy of the compound.
  • Compounds "identified” according to the methods of the present invention can be also tested in cell culture models or in animal models of disease.
  • the GPM protein comprises an amino acid sequence at least 75% homologous to SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6 rubber SEQ ID NO:8 or SEQ ID NO: 13, comprises at least one G protein regulatory motif (e.g., a GPR motif having at least 75% homology to SEQ ID NO: 10), and stimulates G protein activity in a receptor-independent manner.
  • the GPM protein can comprise a structure as described above in the sections discussing GPM proteins and nucleic acids.
  • the cell has been engineered to express the GPM protein by introducing into the cell an expression vector encoding the GPM protein.
  • the cell can be further engineered to express other proteins, such as a G protein ⁇ subunit.
  • the cell is a yeast cell that has been engineered to express a GPM protein and a mammalian or chimeric G protein ⁇ subunit and the effect of the test compound on the activity of the GPM protein is determined by monitoring a pheromone response pathway in the yeast cells.
  • a preferred chimeric G protein subunit which the yeast cell is engineered to express is a Gpal-G ⁇ i2 chimeric G protein ⁇ subunit (preferably comprising 41 amino-terminal amino acids from yeast Gpal operatively linked to a mammalian G ⁇ i2).
  • the pheromone response pathway in the yeast cells can be monitored, for example, by measuring the activity of a pheromone responsive promoter in the yeast cells.
  • Yeast cell compositions and methods that can be used for screening modulators of GPM proteins are described in further detail below and in the Examples.
  • the effect of the test compound on the activity of the GPM protein is determined by monitoring the ability of the test compound to bind to the GPM protein.
  • the effect of the test compound on the activity of the GPM protein is determined by monitoring the ability of the test compound to modulate the interaction of the GPM protein with a target molecule.
  • the target molecule is a G protein.
  • the invention provides assays for screening candidate/test compounds which interact with (e.g., bind to) GPM protein.
  • the assays are cell-free assays which include the steps of combining a GPM protein or a biologically active portion.
  • a candidate/test compound e.g., under conditions which allow for interaction of (e.g., binding of) the candidate/test compound to the GPM protein or portion thereof to form a complex, and detecting the formation of a complex, in which the ability of the candidate compound to interact with (e.g., bind to) the GPM protein or portion thereof is indicated by the presence of the candidate compound in the complex.
  • Formation of complexes between the GPM protein and the candidate compound can be quantitated, for example, using standard immunoassays.
  • the invention provides screening assays to identify candidate/test compounds which modulate (e.g., stimulate or inhibit) the interaction (and most likely GPM activity as well) between a GPM protein and a molecule (target molecule) with which the GPM protein normally interacts.
  • target molecules includes proteins in the same signaling pathway as the GPM protein, e.g.. G proteins, or proteins which may function upstream (including both stimulators and inhibitors of activity) or downstream of the G protein in the pheromone response pathway.
  • the assays are cell-free assays which include the steps of combining a GPM protein or a biologically active portion thereof, a GPM target molecule (e.g., a G protein) and a candidate/test compound, e.g., under conditions wherein but for the presence of the candidate compound, the GPM protein or biologically active portion thereof interacts with (e.g., binds to) the target molecule, and detecting the formation of a complex which includes the GPM protein and the target molecule or detecting the interaction/reaction of the GPM protein and the target molecule.
  • Detection of complex formation can include direct quantitation of the complex by, for example, measuring inductive effects of the GPM protein.
  • a statistically significant change, such as a decrease, in the interaction of the GPM and target molecule (e.g., in the formation of a complex between the GPM and the target molecule) in the presence of a candidate compound (relative to what is detected in the absence of the candidate compound) is indicative of a modulation (e.g., stimulation or inhibition) of the interaction between the GPM protein and the target molecule.
  • Modulation of the formation of complexes between the GPM protein and the target molecule can be quantitated using, for example, an immunoassay.
  • GPM GPM
  • its target molecule to facilitate separation of complexes from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay.
  • Interaction e.g., binding of
  • GPM to a target molecule, in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix.
  • glutathione-S-transferase/ GPM fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the cell lysates (e.g. 35s-. l beled) and the candidate compound, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads are washed to remove any unbound label, and the matrix immobilized and radiolabel determined directly, or in the supernatant after the complexes are dissociated.
  • the complexes can be dissociated from the matrix, separated by SDS-PAGE, and the level of GPM-binding protein found in the bead fraction quantitated from the gel using standard electrophoretic techniques.
  • GPM or its target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated GPM molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with GPM but which do not interfere with binding of the protein to its target molecule can be derivatized to the wells of the plate, and GPM trapped in the wells by antibody conjugation.
  • preparations of a GPM-binding protein and a candidate compound are incubated in the GPM-presenting wells of the plate, and the amount of complex trapped in the well can be quantitated.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the GPM target molecule, or which are reactive with GPM protein and compete with the target molecule; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the target molecule.
  • the invention also provides cell-based assays for identifying compounds which modulate GPM activity.
  • the cell-based assay involves a method for identifying a compound which modulates (e.g.. stimulates or inhibits) GPM activity including contacting a cell which contains a GPM protein with a test compound and determining the ability of the test compound to modulate the activity of the GPM protein.
  • determining the ability of the test compound to modulate GPM activity includes determining the ability of the test compound to effect (e.g., upregulate or down regulate) G-protein mediated signaling in the cell.
  • G-protein mediated signaling can be determined in a GPM-containing cell prior to contacting the cell with a test compound and compared to the signaling after contacting the cell with a test compound.
  • Compounds which reduce the G-protein mediated signaling can be identified as GPM antagonists whereas compounds which increase the G-protein mediated signaling can be identified as antagonists.
  • modulators of GPM expression can be identified in a method wherein a cell is contacted with a candidate compound and the expression of GPM mRNA or protein in the cell is determined. The level of expression of GPM mRNA or protein in the presence of the candidate compound is compared to the level of expression of GPM mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of GPM nucleic acid expression based on this comparison. For example, when expression of GPM mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of GPM mRNA or protein expression.
  • the candidate compound when expression of GPM mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of GPM mRNA or protein expression.
  • the level of GPM mRNA or protein expression in the cells can be determined by methods described herein for detecting GPM mRN A. or protein.
  • the present invention also provides cell-based assa s foi identifying compounds which modulate the activity of a GPM.
  • Cells for use in cell based assays are engineered to express a heterologous GPM protein.
  • the cells may be further engineered such that the endogenous GPM protein is not expressed in functional form.
  • Cells used to express a heterologous GPM can be, for example, mammalian or yeast in origin.
  • the engineered cells of the present invention are yeast cells.
  • the cells for use in the instant assays are further engineered to express a heterologous G protein coupled receptor which is functionally integrated into the signaling pathway of the cell in which it is expressed.
  • heterologous GPCRs can be expressed in yeast cells and can be made to couple to yeast G proteins resulting in the transduction of signals via the endogenous yeast pheromone system signaling pathway which is normally activated by STE2 or
  • heterologous receptors can be made to couple more effectively to the yeast pheromone system signaling pathway by coexpressing a heterologous G protein subunit or subunits, by expressing a chimeric G protein subunit, or by expressing a chimeric G protein coupled receptor.
  • Methods for preparing engineered yeast cells, and engineered yeast cells themselves, are described in U.S. Patent No. 5,482,835 by King et.al. and PCT Publication WO 94/23025 by Fowlkes et.al., the contents of both of which are hereby expressly incorporated herein by reference.
  • the effect of a test compound on GPM induced G protein activation can be measured by detecting changes in second messenger generation.
  • G-protein-regulated including adenylyl cyclase, cyclic GMP, phosphodiesterases, phosphoinositidase C, and phospholipase A2-
  • G proteins interact with a range of ion channels and are able to inhibit certain voltage-sensitive Ca ++ transients, as well as stimulating cardiac K + channels.
  • yeast cells a reduction in the generation of GPM-induced second messengers or mating factor responses (e.g., growth arrest or shmoo formation) could be measured.
  • the GTPase enzymatic activity by G proteins can be measured in plasma membrane preparations by determining the breakdown of ⁇ 3 p GTP using techniques that are known in the art (For example, see Signal Transduction: A GPMctical Approach. G. Milligan, Ed. Oxford University Press, Oxford England).
  • receptors that modulate cAMP are tested, it will be possible to use standard techniques for cAMP detection, such as competitive assays which quantitate HJcAMP in the presence of unlabelled cAMP.
  • Inositol lipids can be extracted and analyzed using standard lipid extraction techniques. DAG can also be measured using thin-layer chromatography. Water soluble derivatives of all three inositol lipids (IP 1 , IP2, IP3) can also be quantitated using radiolabelling techniques or HPLC.
  • the mobilization of intracellular calcium or the influx of calcium from outside the cell can be measured using standard techniques.
  • the choice of the appropriate calcium indicator, fluorescent, bioluminescent, metallochromic, or Ca++-sensitive microelectrodes depends on the cell type and the magnitude and time constant of the event under study (Borle (1990) Environ Health Perspect 84:45-56).— As-en exemplary method of Ca++ detection, cells could be loaded with the Ca++sensitive fluorescent dye fura-2 or indo-1, using standard methods, and any change in Ca++ measured using a fluorometer.
  • DAG can also be produced from phosphatidyl choline.
  • the breakdown of this phospholipid in response to receptor- mediated signaling can also be measured using a variety of radiolabelling techniques.
  • the activation of phospholipase A2 can easily be quantitated using known techniques, including, for example, the generation of arachadonate in the cell.
  • Such assay formats may be useful when the receptor of interest is a receptor tyrosine kinase.
  • yeast transformed with the FGF receptor and a ligand which binds the FGF receptor could be screened using colony immunoblotting (Lyons and Nelson (1984) Proc. Natl. Acad. Sci. USA 81 :7426-7430) using anti-phosphotyrosine.
  • tests for phosphorylation could be useful when a receptor which may not itself be a tyrosine kinase, activates protein kinases that function downstream in the signal transduction pathway.
  • Multi-kinase cascades allow not only signal amplification but also signal divergence to multiple effectors that are often cell-type specific, allowing a growth factor to stimulate mitosis of one cell and differentiation of another.
  • MAP kinase pathway that appears to mediate both mitogenic, differentiation and stress responses in different cell types. Stimulation of growth factor receptors results in Ras activation followed by the sequential activation of c-Raf, MEK, and p44 and p42 MAP kinases (ERK1 and ERK2). Activated MAP kinase then phosphorylates many key regulatory proteins, including p90RSK and Elk-1 that are phosphorylated when MAP kinase translocates to the nucleus. Homologous pathways exist in mammalian and yeast cells. For instance, an essential part of the S.
  • cerevisiae pheromone signaling pathway is comprised of a protein kinase cascade composed of the products of the STE1 1, STE7, and FUS3/KSS 1 genes (the latter pair are distinct and functionally redundant). Accordingly, phosphorylation and/or activation of members of this kinase cascade can be detected and used to quantitate receptor engagement.
  • Phosphotyrosine specific antibodies are available to measure increases in tyrosine phosphorylation and phospho-specific antibodies are commercially available (New England Biolabs, Beverly, MA). Modified methods for detecting receptor-mediated signal transduction exist and one of skill in the art will recognize suitable methods that may be used to substitute for the example methods listed.
  • an indicator gene can be used for detection.
  • an indicator gene is an unmodified endogenous gene.
  • the instant method can rely on detecting the transcriptional level of such pheromone system pathway responsive endogenous genes as the Barl or Fusl. Fus 2, mating factor, Ste3 Stel3, Kexl, Ste2, Ste6, Stel, sSst2, or Chsl. (Appletauer and Zchstetter (1989) Eur. J. Biochem. 181 :243).
  • the sensitivity of an endogenous indicator gene can be enhanced by manipulating the promoter sequence at the natural locus for the indicator gene.
  • Such manipulation may range from point mutations to the endogenous regulatory elements to gross replacement of all or substantial portions of the regulatory elements.
  • the promoter of the gene can be modified to enhance the transcription of Barl upon activation of the yeast pheromone system pathway. Barl gene transcription is activated upon exposure of yeast cells to mating factor.
  • the sequence of the Barl gene is known in the art (see e.g., U.S. patent 4,613,572). Moreover, the sequences required for ⁇ -factor-enhanced expression of the Barl, and other pheromone responsive genes have been identified. (Appeltauer and Achstetter (1989) Eur. J. Biochem. 181 :243; Hagen et. al. ( 1991 ) Mol. Cell. Biol.
  • the yeast Barl promoter can be engineered by mutagenesis to be more responsive, e.g., to more strongly promoter gene transcription, upon stimulation of the yeast pheromone pathway. Standard techniques for mutagenizing the promoter can be used. In such embodiments, it is desirable that the conserved oligonucieotide motif described by Appeltaure et.al. be conserved.
  • the activity of endogenous yeast proteins can be assayed.
  • the signal transduction pathway of the receptor upregulates expression or otherwise activates an enzyme which is capable of modifying a substrate which can be added to the cell.
  • the signal can be detected by using a detectable substrate, in which case loss of the substrate signal is monitored, or alternatively, by using a substrate which produces a detectable product.
  • the substrate is naturally occurring.
  • the substrate can be non-naturally occurring.
  • BAR1 activity can be measured.
  • the modulation of a receptor by a test compound can result in a change in the transcription of a gene, which is not normally pheromone responsive.
  • the gene is easily detectable.
  • the subject assay can be used to measure Pho5, a secreted acid phosphatase. Acid phosphatase activity can be measured using standard techniques.
  • reporter gene constructs can be used. Reporter gene constructs are prepared by operatively linking a reporter gene with at least one transcriptional regulatory element. If only one transcriptional regulatory element is included it must be a regulatable promoter. At least one of the selected transcriptional regulatory elements must be indirectly or directly regulated by the activity of the selected cell-surface receptor whereby activity of the receptor can be monitored via transcription of the reporter genes.
  • reporter genes and transcriptional regulatory elements are known to those of skill in the art and others may be identified or synthesized by methods known to those of skill in the art.
  • Reporter genes include any gene that expresses a detectable gene product, which may be RNA or protein. Preferred reporter genes are those that are readily detectable.
  • the reporter gene may also be included in the construct in the form of a fusion gene with a gene that includes desired transcriptional regulatory sequences or exhibits other desirable properties.
  • reporter genes include, but are not limited to CAT (chloramphenicol acetyl transferase) (Alton and Vapnek (1979) Nature 282:864-869) luciferase, and other enzyme detection systems, such as beta-galactosidase; firefly luciferase (deWet et.al. (1987) Mol. Cell. Biol. 7:725-737); bacterial luciferase (Engebrecht and Silverman (1984), Proc. Natl. Acad. Sci. 1 :4154-4158; Baldwin et.al. (1984) Biochemistry 23:3663-3667); alkaline phosphatase (Toh et.al. (1989) Eu.
  • CAT chloramphenicol acetyl transferase
  • alkaline phosphatase Toh et.al. (1989) Eu.
  • Transcriptional control elements include, but are not limited to, promoters, enhancers, and repressor and activator binding sites.
  • Suitable transcriptional regulatory elements may be derived from the transcriptional regulatory regions of genes whose expression is rapidly induced, generally within minutes, of contact between the cell surface protein and the effector protein that modulates the activity of the cell surface protein. Examples of such genes include, but are not limited to, the immediate early genes (see, Sheng et. ⁇ /.(1990) Neuron 4:477-485), such as c-fos.
  • Immediate early genes are genes that are rapidly induced upon binding of a ligand to a cell surface protein.
  • the transcriptional control elements that are preferred for use in the gene constructs include transcriptional control elements from immediate early genes, elements derived from other genes that exhibit some or all of the characteristics of the immediate early genes, or synthetic elements that are constructed such that genes in operative linkage therewith exhibit such characteristics.
  • the characteristics of preferred genes from which the transcriptional control elements are derived include, but are not limited to, low or undetectable expression in quiescent cells, rapid induction at the transcriptional level within minutes of extracellular simulation, induction that is transient and independent of new protein synthesis, subsequent shut-off of transcription requires new protein synthesis, and mRNAs transcribed from these genes have a short half-life. It is not necessary for all of these properties to be present.
  • VIP vasoactive intestinal peptide
  • cAMP responsive Fink et.al. (1988) Proc Natl. Acad. Sci 85:6662-6666
  • somatostatin gene promoter cAMP responsive; Montminy et.al (1986) Proc Natl Acad Sci 8.3:6682-6686
  • proenkephalin promoter responsive to cAMP, nicotinic agonists, and phorbol esters
  • Comb et.al (1986) Nature 323:353-356 the phosphoenolpyruvate carboxy-kinase gene promoter (cAMP responsive; Short et.al.
  • cells exposed to both a known agonist and a peptide of unknown activity will be growth arrested if the peptide is neutral or an agonist, but will grow normally if the peptide is an antagonist.
  • the growth arrest response can be used to advantage to discover peptides that function as antagonists.
  • the growth arrest consequent to activation of the pheromone response pathway is an undesirable effect since cells that bind agonists stop growing while surrounding cells that fail to bind agonists will continue to grow. The cells of interest, then, will be overgrown or their detection obscured by the background cells, confounding identification of the cells of interest.
  • the present invention teaches engineering the cell such that: 1) growth arrest does not occur ⁇ as_a result of exogenous signal pathway activation (e.g., by inactivating the FAR1 gene); and or 2) a selective growth advantage is conferred by activating the pathway (e.g., by transforming an auxotrophic mutant with a HIS3 gene under the control of a pheromone-responsive promoter, and applying selective conditions).
  • the promoter may be one which is repressed by the receptor pathway, thereby preventing expression of a product which is deleterious to the cell.
  • a receptor repressed promoter one screens for agonists by linking the promoter to a deleterious gene, and for antagonists, by linking it to a beneficial gene.
  • Repression may be achieved by operably linking a receptor- induced promoter to a gene encoding mRNA which is antisense to at least a portion of the mRNA encoded by the marker gene (whether in the coding or flanking regions), so as to inhibit translation of that mRNA.
  • Repression may also be obtained by linking a receptor-induced promoter to a gene encoding a DNA binding repressor protein, and incorporating a suitable operator site into the promoter or other suitable region of the marker gene.
  • exemplary positively selectable (beneficial) genes include the following: URA3, LYS2, HIS3, LEU2, TRPl; ADE1, 2,3,4,5, 1,8; ARGl, 3, 4, 5, 6, 8; HIS1, 4, 5; ILV1, 2, 5; THR1, 4; TRP2, 3, 4, 5; LEU1, 4; MET2, 3, 4,8,9, 14, 16,19; URA1, 2,4,5, 10; H0M3,6; ASP3; CHOI; ARO 2, 1; CYS3; OLE1; IN01,2,4; PR01,3.
  • Countless other genes are potential selective markers. The above are involved in well-characterized biosynthetic pathways.
  • the imidazoleglycerol phosphate dehydratase (IGP dehydratase) gene (HIS3) is preferred because it is both quite sensitive and can be selected over a broad range of expression levels.
  • the cell is auxotrophic for histidine (requires histidine for growth) in the absence of activation. Activation leads to synthesis of the enzyme and the cell becomes prototrophic for histidine (does not require histidine). Thus the selection is for growth in the absence of histidine. Since only a few molecules per cell of IGP dehydratase are required for histidine prototrophy, the assay is very sensitive.
  • a reporter gene e.g., the fusl-lacZ reporter plasmid can be introduced along with the GPM.
  • the addition of an antagonist should result in a decrease in ⁇ -galactosidase units over that observed in the absence of the GPM, demonstrating the ability of the antagonist to interact in a negative fashion with the GPM.
  • cells can be selected for resistance to aminotriazole (AT), a drug that inhibits the activity of IGP dehydratase.
  • AT aminotriazole
  • Cells with low, fixed level of expression of HIS3 are sensitive to the drug, while cells with higher levels are resistant.
  • the amount of AT can be selected to inhibit cells with a_basal level of
  • HIS3 expression (whatever that level is) but allow growth of cells with an induced level of expression. In this case selection is for growth in the absence of histidine and in the presence of a suitable level of AT.
  • Suitable genes include: URA3 (orotidine-5'-phosphate decarboxylase; inhibits growth on 5-fluoroorotic acid), LYS2 (2-aminoadipate reductase; inhibits growth on ⁇ -aminoadipate as sole nitrogen source), CYH2 (encodes ribosomal protein L29; cycloheximide-sensitive allele is dominant to resistant allele), CAN1 (encodes arginine permease; null allele confers resistance to the arginine analog canavanin), and other recessive drug-resistant markers.
  • the reporter gene affects yeast cell growth.
  • the natural response to signal transduction via the yeast pheromone system response pathway is for cells to undergo growth arrest. This is a preferred way to select for antagonists of a ligand/receptor pair that stimulates a the pathway. An antagonist would inhibit the activation of the pathway; hence, the cell would be able to grow.
  • the FAR1 gene may be considered an endogenous counterselectable marker.
  • the FAR1 gene is preferably inactivated when screening for agonist activity.
  • the reporter gene may also be a screenable gene.
  • the screened characteristic may be a change in cell morphology, metabolism or other screenable features.
  • Suitable markers include beta-galactosidase (Xgal, C12FDG, Salmon-gal, Magenta-Gal (latter two from Biosynth Ag)), alkaline phosphatase. horseradish peroxidase. exo-glucanase (product of yeast exbl gene; nonessential, secreted); luciferase; bacterial green fluorescent protein; (human placental) secreted alkaline phosphatase (SEAP); and chloramphenicol transferase (CAT).
  • beta-galactosidase Xgal, C12FDG, Salmon-gal, Magenta-Gal (latter two from Biosynth Ag)
  • alkaline phosphatase horseradish peroxidase. exo-glucanase (product of yeast exbl gene; nonessential
  • a preferred screenable marker gene is beta-galactosidase; yeast cells expressing the enzyme convert the colorless substrate Xgal into a blue pigment.
  • the promoter may be receptor-induced or receptor-inhibited.
  • a screen can take advantage of the fact that a gpalfusl -HIS3 colony expressing wild type Gas can grow upon replica plating to media lacking histidine and containing ImM 3-aminotriazole (AT).
  • AT ImM 3-aminotriazole
  • the growth of this strain occurs due to the partially constitutive state of the pheromone pathway, which leads to partial derepression of Xhe fusl-HlS3 reporter gene.
  • AT inhibits the activity of IGP dehydratase. Cells with low, fixed level of expression of HIS3 are sensitive to the drug, while cells with higher levels are resistant.
  • the amount of AT can be selected to inhibit cells with a basal level of HIS3 expression (whatever that levej. is)_but allow growth of cells with an induced level of expression.
  • a colony to which an antagonist of a GPM has been added will presumably fail to grow on this media due to the reduced signaling via the pheromone pathway.
  • a peptide library can be expressed to test for modulators of a GPM.
  • the peptide library is expressed by the cell that also expresses the GPM, thereby creating an "autocrine" system, wherein the peptide to be tested for GPM-modulating activity is made by the same cell expressing the GPM protein.
  • yeast cell autocrine systems for screening peptide see PCT Publication WO 94/23025 by Fowlkes et.al, the contents of which are expressly incorporated herein by reference.
  • such a library can be expressed using a leader sequence for periplasmic expression, e.g., a yeast mating factor leader sequence.
  • Yeast cells are bounded by a lipid bilayer called the plasma membrane. Between this plasma membrane and the cell wall is the periplasmic space. Peptides secreted by yeast cells cross the plasma membrane through a variety of mechanisms and thereby enter the periplasmic space. The secreted peptides are then free to interact with other molecules that are present in the periplasm or displayed on the outer surface of the plasma membrane. The peptides then either undergo re-uptake into the cell, diffuse through the cell wall into the medium, or become degraded within the periplasmic space.
  • the test polypeptide library may be secreted into the periplasm by any of a number of exemplary mechanisms, depending on the nature of the expression system to which they are linked.
  • the peptide may be structurally linked to a yeast signal sequence, such as that present in the ⁇ -factor precursor, which directs secretion through the endoplasmic reticulum and Golgi app --rat _,'... Since this is the same route that the receptor protein follows in its journey to the plasma membrane, opportunity exists in cells expressing both the receptor and the peptide library for a specific peptide to interact with the receptor during transit through the secretory pathway. This has been postulated to occur in mammalian cells exhibiting autocrine activation.
  • This transport pathway and the signals that direct a protein or peptide to this pathway are not as well characterized as is the endoplasmic reticulum-based secretory pathway. Nonetheless, these transporters apparently can efficiently export certain peptides directly across the plasma membrane, without the peptides having to transit the ER/Golgi pathway. It is anticipated that at least a subset of peptides can be secreted through this pathway by expressing the library in context of the ⁇ -factor prosequence and terminal tetrapeptide. The possible advantage of this system is that the receptor and peptide do not come into contact until both are delivered to the external surface of the cell. Thus, this system strictly mimics the situation of an agonist or antagonist that is normally delivered from outside the cell. Use of either of the described pathways is within the scope of the invention.
  • the present invention does not require periplasmic secretion of peptides, or, if such secretion is provided, any particular secretion signal or transport pathway.
  • peptides expressed with a signal sequence may bind to and activate receptors prior to their transport to the cell surface.
  • a heterologous G-protein coupled receptor can be coexpressed with a heterologous GPM protein.
  • the compounds to be tested in the subject assays can be derived from libraries. While the use of libraries of peptides is well established in the art, new techniques have been developed which have allowed the production of mixtures of other compounds, such as benzodiazepines (Bunin et.al (1992) J. Am. Chem. Soc. 114:10987; DeWitt et.al. (1993) Proc. Natl. Acad. Sci. USA 90:6909) peptoids (Zuckermann. (1994) J. Med. Chem. 37:2678) oligocarbamates (Cho et.al.
  • the compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the 'one-bead one-compound' library method, and synthetic library methods using affinity chromatography selection.
  • biological libraries include biological libraries; spatially addressable parallel solid phase or solution phase libraries, synthetic library methods requiring deconvolution, the 'one-bead one-compound' library method, and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, K.S. (1997) Anticancer Drug Des. 12:145).
  • test compound is a peptide or peptidomimetic.
  • compounds are small, organic non-peptidic compounds.
  • the test compounds are exogenously added to the yeast cells expressing a recombinant receptor and compounds that modulate signal transduction via the receptor are selected.
  • the yeast cells express the compounds to be tested.
  • a culture of the subject yeast cells can be further modified to collectively express a peptide library as described in more detail in PCT Publication WO 94/23025 the contents of which is expressly incorporated herein by this reference.
  • combinatorial polypeptide libraries may also be expressed, see, for example, U.S. Patents 5,270,181 and 5,292,646; and PCT publication WO 94/02502).
  • the combinatorial polypeptides are produced from a cD ⁇ A library.
  • Exemplary compounds which can be screened for activity include, but are not limited to, peptides, nucleic acids, carbohydrates, small organic molecules, and natural product extract libraries.
  • both compounds which agonize or antagonize the receptor- or channel-mediated signaling function can be selected and identified.
  • cells can be engineered to produce the compounds to be tested.
  • This assay system has the advantage of increasing the effective concentration of the compound to be tested.
  • a method such as that described in WO 94/23025 can be utilized.
  • Other methods can also be used.
  • peptide libraries are systems which simultaneously display, in a form which permits interaction with a target, a highly diverse and numerous collection of peptides.
  • peptides may be presented in solution (Houghten (1992) Biotechniques 13:412-421), or on beads (Lam (1991) Nature 354:82-84), chips (Fodor (1993) Nature 364:555-556), bacteria (Ladner USP 5,223,409), spores (Ladner USP '409), plasmids (Cull et.al. (1992) Proc Natl Acad Sci USA 89: 1865-1869) or on phage (Scott and Smith (1990) Science 249:386-390); (Devlin (1990)Sc/ ' e- ⁇ ee 249:404-406); (Cwirla et.al. (1990) Proc. Natl. Acad.
  • the screening is for binding in vitro to an artificially presented target, not for activation or inhibition of a cellular signal transduction pathway in a living cell. While a cell surface receptor may be used as a target, the screening will not reveal whether the binding of the peptide caused an allosteric change in the conformation of the receptor.
  • the Ladner et.al. patent, USSN 5,096,815, describes a method of identifying novel proteins or polypeptides with a desired DNA binding activity.
  • Semi-random (“variegated") DNA encoding a large number of different potential binding proteins is introduced, in expressible form, into suitable yeast cells.
  • the target DNA sequence is incorporated into a genetically engineered operon such that the binding of the protein or polypeptide will prevent expression of a gene product that is deleterious to the gene under selective conditions. Cells which survive the selective conditions are thus cells which express a protein which binds the target DNA. While it is taught that yeast cells may be used for testing, bacterial cells are preferred.
  • the compounds tested are in the form of peptides from a peptide library.
  • the peptide library of the present invention takes the form of a cell culture, in which essentially each cell expresses one, and usually only one, peptide of the library. While the diversity of the library is maximized if each cell produces a peptide of a different sequence, it is usually prudent to construct the library so there is some redundancy.
  • the combinatorial peptides of the library can be expressed as is, or can be incorporated into larger fusion proteins.
  • the fusion protein can provide, for example, stability against degradation or denaturation, as well as a secretion signal if secreted.
  • the polypeptide library is expressed as thioredoxin fusion proteins (see. for example, U.S. Patents 5,270,181 and 5,292,646; and PCT publication WO94/ 02502).
  • the combinatorial peptide can be attached one the terminus of the thioredoxin protein, or, for short peptide libraries, inserted into the so-called active loop.
  • the peptide library is derived to express a combinatorial library of polypeptides which are not based on any known sequence, nor derived from cDNA. That is, the sequences of the library are largely random.
  • the combinatorial polypeptides are in the range of 3-100 amino acids in length, more preferably at least 5-50, and even more preferably at least 10, 13, 15, 20 or 25 amino acid residues in length.
  • the polypeptides of the library are of uniform length. It will be understood that the length of the --omhinatorial peptide does not reflect any extraneous sequences which may be present in order to facilitate expression, e.g., such as signal sequences or invariant portions of a fusion protein.
  • the peptide- library is a combinatorial library of polypeptides which are based at least in part on a known polypeptide sequence or a portion thereof (not a cDNA library). That is, the sequences of the library is semi- random, being derived by combinatorial mutagenesis of a known sequence. See, for example, Ladner et.al PCT publication WO 90/02909; Garrard et.al. , PCT publication WO 92/09690; Marks et.al. (1992) J. Biol Chem. 267:16007-16010: Griffths et.al. (1993) EMBO J 12:725-734; Clackson et.al.
  • polypeptide(s) which are known ligands for a target receptor can be mutagenized by standard techniques to derive a variegated library of polypeptide sequences which can further be screened_for agonists and/or antagonists.
  • the combinatorial polypeptides are produced from a cDNA library.
  • the cells collectively produce a "peptide library", preferably including at least 10 3 to 10 7 different peptides, so that diverse peptides may be simultaneously assayed for the ability to interact with the exogenous receptor.
  • a "peptide library” preferably including at least 10 3 to 10 7 different peptides, so that diverse peptides may be simultaneously assayed for the ability to interact with the exogenous receptor.
  • at least some peptides of the peptide library are secreted into the periplasm, where they may interact with the "extracellular" binding site(s) of an exogenous receptor. They thus mimic more closely the clinical interaction of drugs with cellular receptors.
  • This embodiment optionally may be further improved (in assays not requiring pheromone secretion) by preventing pheromone secretion, and thereby avoiding competition between the peptide and the pheromone for signal peptidase and other components of the secretion system.
  • the peptides of the library are encoded by a mixture of DNA molecules of different sequence.
  • Each peptide-encoding DNA molecule is ligated with a vector DNA molecule and the resulting recombinant DNA molecule is introduced into a yeast cell. Since it is a matter of chance which peptide encoding DNA molecule is introduced into a particular cell, it is not predictable which peptide that cell will produce. However, based on a knowledge of the manner in which the mixture was prepared, one may make certain statistical predictions about the mixture of peptides in the peptide library.
  • the peptides of the library can be composed of constant and variable residues. If the nth residue is the same for all peptides of the library, it is said to be constant. If the nth residue varies, depending on the peptide in question, the residue is a variable one.
  • the peptides of the library will have at least one, and usually more than one, variable residue.
  • a variable residue may vary among any of two to all twenty of the genetically encoded amino acids; the variable residues of the peptide may vary in the same or different manner.
  • the frequency of occurrence of the allowed amino acids at a particular residue position may be the same or different.
  • the peptide may also have one or more constant residues. There are two principal ways in which to prepare the required DNA mixture.
  • the DNAs are synthesized a base at a time.
  • a suitable mixture of nucleotides is reacted with the nascent DNA, rather than the pure nucleotide reagent of conventional polynucleotide synthesis.
  • the second method provides more exact control over the amino acid variation.
  • trinucleotide reagents are prepared, each trinucleotide being a codon of one (and only one) of the amino acids to be featured in the peptide library.
  • a mixture is made of the appropriate trinucleotides and reacted with the nascent DNA.
  • test polypeptide library may be secreted into the periplasm by any of a number of exemplary mechanisms, depending on the nature of the expression system to which they are linked.
  • the peptide may be structurally linked to a yeast signal sequence, such as that present in the ⁇ -factor precursor, which directs secretion through the endoplasmic reticulum and Golgi apparatus. Since this is the same route that the receptor protein follows in its journey to the plasma membrane, opportunity exists in cells expressing both the receptor and the peptide library for a specific peptide to interact with the receptor during transit through the secretory pathway. This has been postulated to occur in mammalian cells exhibiting autocrine activation.
  • Such interaction could yield activation of the response pathway during transit, which would still allow identification of those cells expressing a peptide agonist.
  • this system would still be effective, since both the peptide antagonist and receptor would be delivered to the outside of the cell in concert.
  • those cells producing an antagonist would be selectable, since the peptide antagonist would be properly and timely situated to prevent the receptor from being stimulated by the externally applied agonist.
  • An alternative mechanism for delivering peptides to the periplasmic space is to use the ATP-dependent transporters of the STE6/MDR1 class.
  • This transport pathway and the signals that direct a protein or peptide to this pathway are not as well characterized as is the endoplasmic reticulum-based secretory pathway. Nonetheless, these transporters apparently can efficiently export certain peptides directly across the plasma membrane, without the peptides having to transit the ER/Golgi pathway. It is anticipated that at least a subset of peptides can be secreted through this pathway by expressing the library in context of the a-factor prosequence and terminal tetrapeptide.
  • this system is that the receptor and peptide do not come into contact until both are delivered to the external surface of the cell. Thus, this system strictly mimics the situation of an agonist or antagonist that is normally delivered from outside the cell. Use of either of the described pathways is within the_scope of the invention.
  • the present invention does not require periplasmic secretion, or, if such secretion is provided, any particular secretion signal or transport pathway.
  • agents identified in the subject assay can be formulated in pharmaceutical preparations for in vivo administration to an animal, preferably a human.
  • the compounds selected in the subject assay, or a pharmaceutically acceptable salt thereof may accordingly be formulated for administration with a biologically acceptable medium, such as water, buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like) or suitable mixtures thereof.
  • a biologically acceptable medium such as water, buffered saline, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like) or suitable mixtures thereof.
  • a biologically acceptable medium includes any and all solvents, dispersion media, and the like which may be appropriate for the desired route of administration of the pharmaceutical preparation. The use of such media for pharmaceutically active substances is known in the art.
  • Suitable vehicles and their formulation inclusive of other proteins are described, for example, in the book Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences. Mack Publishing Company, Easton, Pa., USA 1985). These vehicles include injectable "deposit formulations". Based on the above, such pharmaceutical formulations include, although not exclusively, solutions or freeze-dried powders of the compound in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered media at a suitable pH and isosmotic with physiological fluids. In preferred embodiment, the compound can be disposed in a sterile preparation for topical and/or systemic administration.
  • the GPM proteins can be used as "bait proteins" in a two-hybrid assay (see, e.g., U.S.
  • the two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains.
  • the assay utilizes two different DNA constructs.
  • the gene that codes for GPM is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein (“prey" or "sample”) is fused to a gene that codes for the activation domain of the known transcription factor.
  • the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene which encodes the protein which interacts with GPM.
  • a reporter gene e.g., LacZ
  • Modulators of GPM protein activity and or GPM nucleic acid expression identified according to these drug screening assays can be to treat, for example, diseases or disorders characterized by excessive or insufficient G-protein mediated signal transduction.
  • diseases or disorders which can be treated using modulators of GPM protein activity and/or nucleic acid expression include inflammatory disorders/diseases, immune diseases/disorders, proliferative diseases/disorders (e.g., cancer), developmental disordesr/diseases, and or differentiative diseases/disorders.
  • These methods of treatment include the steps of administering the GPM molecules of the present invention or modulators of GPM protein activity and/or nucleic acid expression, e.g., in a pharmaceutical composition as described above, to a subject in need of such treatment.
  • the invention further provides a method for detecting the presence of GPM in a biological sample.
  • the method involves contacting the biological sample with a compound or an agent capable of detecting GPM protein or mRNA such that the presence of GPM is detected in the biological sample.
  • a preferred agent for detecting GPM mRNA is a labeled or labelable nucleic acid probe capable of hybridizing to GPM mRNA.
  • the nucleic acid probe can be, for example, the full-length GPM cDNA of SEQ ID NOT, SEQ ID NO:5, or SEQ ID NO:7, or a portion thereof, such as an oligonucieotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to GPM mRNA.
  • a preferred agent for detecting GPM protein is a labeled or labelable antibody capable of binding to GPM protein.
  • Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used.
  • labeled or labelable with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • biological sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject.
  • the detection method of the invention can be used to detect GPM mRNA or protein in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of GPM mRNA include Northern hybridizations and in situ hybridizations.
  • in vitro techniques for detection of GPM protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence.
  • GPM protein can be detected in vivo in a subject by introducing into the subject a labeled anti- GPM antibody.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • the biological sample is a cell sample, a tissue section, for example, a freeze-dried or fresh frozen section of tissue removed from a patient, or a biological fluid obtained from a subject.
  • kits for detecting the presence of GPM in a biological sample can comprise a labeled or labelable compound or agent capable of detecting GPM protein or mRNA in a biological sample; means for determining the amount of GPM in the sample; and means for comparing the amount of GPM in the sample with a standard.
  • the compound or agent can be packaged in a suitable container.
  • the kit can further comprise instructions for using the kit to detect GPM mRNA or protein.
  • Additional methods of the invention include methods for treating a subject having a disorder characterized by aberrant GPM activity or expression. These methods include administering to the subject a GPM modulator such that treatment of the subject occurs.
  • the terms "treating” or “treatment”, as used herein, refer to reduction or alleviation of at least one adverse effect or symptom of a disease or disorder, e.g., a disease or disorder characterized by or associated with abnormal or aberrant GPM protein activity or GPM nucleic acid expression.
  • a GPM modulator is a molecule which can modulate GPM nucleic acid expression and or GPM protein activity.
  • a GPM modulator can modulate, e.g., upregulate (activate) or downregulate (suppress), GPM nucleic acid expression.
  • a GPM modulator can modulate (e.g., stimulate or inhibit) GPM protein activity.
  • a GPM modulator can be an antisense molecule, e.g., a ribozyme, as described herein.
  • antisense molecules which can be used to inhibit GPM nucleic acid expression include antisense molecules which are complementary to a portion of the 5' untranslated region of SEQ ID NOT, SEQ ID NO:5, or SEQ ID NO:7, which also includes the start codon and antisense molecules which are complementary to a portion of the 3' untranslated region of SEQ ID NOT, SEQ ID NO:5, or SEQ ID NO:7.
  • a GPM modulator which inhibits GPM nucleic acid expression can also be a small molecule or other drug, e.g., a small molecule or drug identified using the screening assays described herein, which inhibits GPM nucleic acid expression.
  • a GPM modulator can be, for example, a nucleic acid molecule encoding GPM (e.g., a nucleic acid molecule comprising a nucleotide sequence homologous to the nucleotide sequence of SEQ ID NOT) or a small molecule or other drug, e.g., a small molecule (peptide) or drug identified using the screening assays described herein, which stimulates GPM nucleic acid expression.
  • a nucleic acid molecule encoding GPM e.g., a nucleic acid molecule comprising a nucleotide sequence homologous to the nucleotide sequence of SEQ ID NOT
  • a small molecule or other drug e.g., a small molecule (peptide) or drug identified using the screening assays described herein, which stimulates GPM nucleic acid expression.
  • a GPM modulator can be an anti- GPM antibody or a small molecule or other drug, e.g., a small molecule or drug identified using the screening assays described herein, which inhibits GPM protein activity.
  • a GPM modulator can be an active GPM protein or portion thereof (e.g., a GPM protein or portion thereof having an amino acid sequence which is homologous to the amino acid sequence of SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:6, SEQ ID NO:8 or SEQ ID NO: 13 or a portion thereof) or a small molecule or other drug, e.g., a small molecule or drug identified using the screening assays described herein, which stimulates GPM protein activity.
  • a cell associated activity refers to a normal or abnormal activity or function of a cell. Examples of cell associated activities include proliferation, migration, differentiation, production or secretion of molecules, such as proteins, and cell survival.
  • the cell-associated activity is mediated by a G-protein signaling pathway.
  • altered refers to a change, e.g., an increase or decrease, of a cell associated activity.
  • the agent stimulates GPM protein activity or GPM nucleic acid expression.
  • stimulatory agents include an active GPM protein, a nucleic acid molecule encoding GPM that has been introduced into the cell, and a modulatory agent which stimulates GPM protein activity or GPM nucleic acid expression and which is identified using the drug screening assays described herein.
  • the agent inhibits GPM protein activity or GPM nucleic acid expression.
  • inhibitory agents include an antisense GPM nucleic acid molecule, an anti- GPM antibody, and a modulatory agent which inhibits GPM protein activity or GPM nucleic acid expression and which is identified using the drug screening assays described herein.
  • modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject).
  • the modulatory methods are performed in vivo, i.e., the cell is present within a subject, e.g., a mammal, e.g., a human, and the subject has a disorder or disease characterized by or associated with abnormal or aberrant GPM activity or expression.
  • a nucleic acid molecule, a protein, a GPM modulator etc. used in the methods of treatment can be incorporated into an appropriate pharmaceutical composition described herein and administered to the subject through a route which allows the molecule, protein, modulator etc. to perform its intended function. Examples of routes of administration are also described herein. Particular GPM inhibitory agents and GPM stimulatory agents are described further below.
  • GPM activity is inhibited in a cell by contacting the cell with an inhibitory agent.
  • Inhibitory agents of the invention can be, for example, intracellular binding molecules that act to inhibit the expression or activity of GPM.
  • intracellular binding molecule is intended to include molecules that act in tracellularly to inhibit the expression or activity of a protein by binding to the protein itself, to a nucleic acid (e.g., an mRNA molecule) that encodes the protein or to a target with which the protein indirectly interacts.
  • intracellular binding molecules examples include antisense GPM nucleic acid molecules (e.g., to inhibit translation of GPM mRNA), intracellular anti-GPM antibodies (e.g., to inhibit the activity of GPM protein) and chemical inhibitors of the GPM protein.
  • an inhibitory agent of the invenuon is an antisense nucleic acid molecule that is complementary to a gene encoding AGSor to a portion of said gene, or a recombinant expression vector encoding said antisense nucleic acid molecule.
  • antisense nucleic acids to downregulate the expression of a particular protein in a cell is well known in the art (see e.g., Weintraub, H. et.al, Antisense RNA as a molecular tool for genetic analysis, Reviews - Trends in Genetics, Vol. 1(1) 1986; Askari, F.K. and McDonnell, W.M. (1996) N. Eng. J. Med. 334:316-318; Bennett, M.R.
  • An antisense nucleic acid molecule comprises a nucleotide sequence that is complementary to the coding strand of another nucleic acid molecule (e.g., an mRNA sequence) and accordingly is capable of hydrogen bonding to the coding strand of the other nucleic acid molecule.
  • Antisense sequences complementary to a sequence of an mRNA can be complementary to a sequence found in the coding region of the mRNA, the 5 ' or 3' untranslated region of the mRNA or a region bridging the coding region and an untranslated region (e.g., at the junction of the 5' untranslated region and the coding region).
  • an antisense nucleic acid can be complementary in sequence to a regulatory region of the gene encoding the mRNA, for instance a transcription initiation sequence or regulatory element.
  • an antisense nucleic acid is designed so as to be complementary to a region preceding or spanning the initiation codon on the coding strand or in the 3' untranslated region of an mRNA.
  • An antisense nucleic acid for inhibiting the expression of GPM protein in a cell can be designed based upon the nucleotide sequence encoding the GPM protein (e.g., SEQ ID NO: 1), constructed according to the rules of Watson and Crick base pairing (e.g., as described above in subsection I).
  • an antisense nucleic acid can exist in a variety of different forms.
  • the antisense nucleic acid can be an oligonucieotide that is complementary to only a portion of an GPM gene.
  • An antisense oligonucleotides can be constructed using chemical synthesis procedures known in the art.
  • An antisense oligonucieotide can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g. phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • one or more antisense oligonucleotides can be added to cells in culture media, typically at about 200 ⁇ g oligonucleotide/ml.
  • an antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., nucleic acid transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest).
  • Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the expression of the antisense RNA molecule in a cell of interest, for instance promoters and/or enhancers or other regulatory sequences can be chosen which direct constitutive, tissue specific or inducible expression of antisense RNA.
  • an inducible eAGSaryotic regulatory system such as the Tet system (e.g., as described in Gossen. M. and Bujard, H. (1992) Proc. Natl Acad. Sci. USA 89:5547-5551 ; Gossen, M. et.al. (1995) Science 268:1766-1769; PCT Publication No. WO 94/29442; and PCT Publication No. WO 96/01313) can be used.
  • the antisense expression vector is prepared as described above for recombinant expression vectors, except that the cDNA (or portion thereof) is cloned into the vector in the antisense orientation.
  • the antisense expression vector can be in the form of, for example, a recombinant plasmid, phagemid or attenuated virus.
  • the antisense expression vector is introduced into cells using a standard transfection technique, as described above for recombinant expression vectors.
  • an antisense nucleic acid for use as an inhibitory agent is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region (for reviews on ribozymes see e.g., Ohkawa, J. et.al. (1995) J. Biochem. 1 18:251-258; NASAdsson, ST. and Eckstein, F. (1995) Trends Biotechnol 13:286-289; Rossi, J.J. (1995) Trends Biotechnol. 0:301-306; Kiehntopf, M.
  • a ribozyme having specificity for GPM mRNA can be designed based upon the nucleotide sequence of the GPM cDNA.
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the base sequence of the active site is complementary to the base sequence to be cleaved in an GPM mRNA. See for example U.S. Patent Nos. 4,987,071 and 5,1 16,742, both by Cech et.al.
  • GPM mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See for example Bartel, D. and Szostak, J.W. (1993) Science 261 : 141 1-1418.
  • Another type of inhibitory agent that can be used to inhibit the expression and/or activity of GPM in a cell is an intracellular antibody specific for the GPM protein.
  • the use of intracellular antibodies to inhibit protein function in a cell is known in the art (see e.g., Carlson, J. R. (1988) Mol. Cell. Biol. 8:2638-2646; Biocca, S. et.al. (1990) EMBO J. 9:101-108; Werge, T.M. et.al. (1990) FEBS Letters 274:193-198; Carlson, J.R. (1993) Proc. Natl Acad. Sci. USA 90:7427-7428; Marasco, W.A. et.al.
  • a recombinant expression vector which encodes the antibody chains in a form such that, upon introduction of the vector into a cell, the antibody chains are expressed as a functional antibody in an intracellular compartment of the cell.
  • an intracellular antibody that specifically binds the GPM protein is expressed in the cytoplasm of the cell.
  • antibody light and heavy chain cDNAs encoding antibody chains specific for the target protein of interest, e.g., GPM are isolated, typically frorn a hybridoma that secretes a monoclonal antibody specific for the GPM protein.
  • Hybridomas secreting anti-GPM monoclonal antibodies, or recombinant anti-GPM monoclonal antibodies can be prepared as described above.
  • a monoclonal antibody specific for GPM protein e.g., either a hybridoma-derived monoclonal antibody or a recombinant antibody from a combinatorial library
  • DNAs encoding the light and heavy chains of the monoclonal antibody are isolated by standard molecular biology techniques.
  • light and heavy chain cDNAs can be obtained, for example, by PCR amplification or cDNA library screening.
  • cDNA encoding the light and heavy chains can be recovered from the display package (e.g., phage) isolated during the library screening process.
  • Nucleotide sequences of antibody light and heavy chain genes from which PCR primers or cDNA library probes can be prepared are known in the art. For example, many such sequences are disclosed in Kabat, E.A., et.al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242 and in the "Vbase" human germline sequence database.
  • an intracellular antibody expression vector can encode an intracellular antibody in one of several different forms. For example, in one embodiment, the vector encodes full-length antibody light and heavy chains such that a full-length antibody is expressed intracellularly. In another embodiment, the vector encodes a full-length light chain but only the VH CH1 region of the heavy chain such that a Fab fragment is expressed intracellularly.
  • the vector encodes a single chain antibody (scFv) wherein the variable regions of the light and heavy chains are linked by a flexible peptide linker (e.g., (Gly 4 Ser) 3 ) and expressed as a single chain molecule.
  • a flexible peptide linker e.g., (Gly 4 Ser) 3
  • the expression vector encoding the anti-GPM intracellular antibody is introduced into the cell by standard transfection methods, as discussed hereinbefore.
  • Other inhibitory agents that can be used to inhibit the activity of an GPM protein are chemical compounds that directly inhibit GPM activity or inhibit the interaction between GPM and target molecules. Such compounds can be identified using screening assays that select for such compounds, as described in detail above.
  • GPM activity is stimulated in a cell by contacting the cell with a stimulatory agent.
  • stimulatory agents include active GPM protein and nucleic acid molecules encoding GPM that are introduced into the cell to increase GPM activity in the cell.
  • a preferred stimulatory agent is a nucleic acid molecule encoding an GPM protein, wherein the nucleic acid molecule is introduced into the cell in a form suitable for expression of the active GPM protein in the cell.
  • an GPM-encoding DNA is first introduced into a recombinant expression vector using standard molecular biology techniques, as described herein.
  • AN GPM-encoding DNA can be obtained, for example, by amplification using the polymerase chain reaction (PCR), using primers based on the GPM nucleotide sequence. Following isolation or amplification of GPM- encoding DNA, the DNA fragment is introduced into an expression vector and transfected into target cells by standard methods, as described herein.
  • PCR polymerase chain reaction
  • Other stimulatory agents that can be used to stimulate the activity of an GPM protein are chemical compounds that stimulate GPM activity in cells, such as compounds that directly stimulate GPM protein and compounds that promote the interaction between GPM and target molecules. Such compounds can be identified using screening assays that select for such compounds, as dc cril ed in detail above.
  • the modulatory methods of the invention can be performed in vitro (e.g., by culturing the cell with the agent or by introducing the agent into cells in culture) or, alternatively, in vivo (e.g., by administering the agent to a subject or by introducing the agent into cells of a subject, such as by gene therapy).
  • cells can be obtained from a subject by standard methods and incubated (i.e., cultured) in vitro with a modulatory agent of the invention to modulate GPM activity in the cells.
  • a modulatory agent of the invention can be readministered to the subject.
  • the modulatory agent can be administered to the subject such that GPM activity in cells of the subject is modulated.
  • subject is intended to include living organisms in which an GPM-dependent cellular response can be elicited.
  • Preferred subjects are mammals. Examples of subjects include humans, monkeys, dogs, cats, mice, rats, cows, horses, goats and sheep.
  • nucleic acids including recombinant expression vectors encoding GPM protein, antisense RNA and intracellular antibodies
  • the agents can be introduced into cells of the subject using methods known in the art for introducing nucleic acid (e.g., DNA) into cells in vivo. Examples of such methods encompass both non-viral and viral methods, including:
  • Naked DNA can be introduced into cells in vivo by directly injecting the DNA into the cells (see e.g., Acsadi et.al. (1991 ) Nature 332:815-818; Wolff et.al. (1990) Science 247:1465-1468).
  • a delivery apparatus e.g., a "gene gun” for injecting DNA into cells in vivo can be used.
  • Such an apparatus is commercially available (e.g., from BioRad).
  • Cationic Lipids Naked DNA can be introduced into cells in vivo by complexing the DNA with cationic lipids or encapsulating the DNA in cationic liposomes.
  • suitable cationic lipid formulations include N-[-l-(2,3- dioleoyloxy)propyl]N,N,N-triethylammonium chloride (DOTMA) and a 1 : 1 molar ratio of l,2-dimyristyloxy-propyl-3-dimethylhydroxyethylammonium bromide (DMRIE) and dioleoyl phosphatidylethanolamine (DOPE) (see e.g., Logan, J.J. et.al. (1995) Gene Therapy 2:38-49; San, H. et.al. (1993) Human Gene Therapy 4:781-788).
  • DOTMA N-[-l-(2,3- dioleoyloxy)propyl]N,N,N-tri
  • Naked DNA can also be introduced into cells in vivo by complexing the DNA to a cation, such as polylysine, which is coupled to a ligand for a cell-surface receptor (see for example Wu, G. and Wu, CH. (1988) J. Biol Chem. 263:14621 ; Wilson et.al. (1992) J. Biol. Chem. 267:963-967; and U.S. Patent No. 5,166,320). Binding of the DNA-ligand complex to the receptor facilitates uptake of the DNA by receptor-mediated endocytosis.
  • a cation such as polylysine
  • a DNA-ligand complex linked to adenovirus capsids which naturally disrupt endosomes, thereby releasing material into the cytoplasm can be used to avoid degradation of the complex by intracellular lysosomes (see for example Curiel et.al. (1991) Proc. Natl. Acad. Sci. USA 88:8850; Cristiano et.al. (1993) Proc. Natl Acad. Sci. USA 90:2122-2126).
  • Retroviruses Defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D. (1990) Blood 76:271).
  • a recombinant retrovirus can be constructed having a nucleotide sequences of interest incorporated into the retroviral genome. Additionally, portions of the retroviral genome can be removed to render the retrovirus replication defective. The replication defective retrovirus is then packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F.M. et.al.
  • retroviruses include pLJ, pZIP, pWE and pEM which are well known to those skilled in the art.
  • suitable packaging virus lines include ⁇ Crip, ⁇ Cre, ⁇ 2 and ⁇ Am. Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial cells, endothelial cells, lymphocytes, myoblasts, hepatocytes, bone marrow cells, in vitro and/or in vivo (see for example Eglitis, et.al. (1985) Science 230:1395-1398; Danos and Mulligan (1988) Proc.
  • Retroviral vectors require target cell division in order for the retroviral genome (and foreign nucleic acid inserted into it) to be integrated into the host genome to stably introduce nucleic acid into the cell. Thus, it may be necessary to stimulate replication of the target cell.
  • Adenoviruses The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See for example Berkner et.al. (1988) BioTechniques 6:616; Rosenfeld et.al. (1991) Science 252:431-434; and Rosenfeld et.al.
  • adenoviral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus are well known to those skilled in the art.
  • Recombinant adenoviruses are advantageous in that they do not require dividing cells to be effective gene delivery vehicles and can be used to infect a wide variety of cell types, including airway epithelium (Rosenfeld et. ⁇ _(1992) cited supra), endothelial cells (Lemarchand et.al. (1992) Proc. Natl. Acad. Sci.
  • adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et.al. cited supra; Haj- Ahmand and Graham (1986) J. Virol. 57:267).
  • Most replication-defective adenoviral vectors currently in use are deleted for all or parts of the viral El and E3 genes but retain as much as 80 % of the adenoviral genetic material.
  • Adeno-associated virus is a naturally occurring defective virus that requires another virus, such as an adenovirus or a herpes virus, as a helper virus for efficient replication and a productive life cycle.
  • AAV Adeno-associated virus
  • AAV vector such as that described in Tratschin et.al. (1985) Mol. Cell. Biol. 5:3251-3260 can be used to introduce DNA into cells.
  • a variety of nucleic acids have been introduced into different cell types using AAV vectors (see for example Hermonat et.al (1984) Proc. Natl. Acad. Sci. USA 81_:6466-6470; Tratschin et.al. (1985) Mol. Cell Biol.
  • DNA introduced into a cell can be detected by a filter hybridization technique (e.g., Southern blotting) and RNA produced by transcription of introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR).
  • RNA produced by transcription of introduced DNA can be detected, for example, by Northern blotting, RNase protection or reverse transcriptase-polymerase chain reaction (RT-PCR).
  • RT-PCR reverse transcriptase-polymerase chain reaction
  • the gene product can be detected by an appropriate assay, for example by immunological detection of a produced protein, such as with a specific antibody, or by a functional assay to detect a functional activity of the gene product.
  • a retroviral expression vector encoding GPM is used to express GPM protein in cells in vivo, to thereby stimulate GPM protein activity in vivo.
  • retroviral vectors can be prepared according to standard methods known in the art (discussed further above).
  • a modulatory agent such as a chemical compound
  • Such compositions typically comprise the modulatory agent and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary' active compounds can also be incorporated into the compositions.
  • Pharmaceutical compositions can be prepared as described above.
  • yeast strains were used in the EXAMPLES described herein.
  • mRNA was prepared from 20 100 mm confluent plates of cells using the Stratagene mRNA isolation kit. The mRNA was processed for cDNA library construction using the cDNA kit from Stratagene using an oligo dT primer that contained an Xho I restriction site. The 5' end of the cDNA was modified with Eco RI adaptors. Both size fractionated and non-size fractionated cDNA was restricted with the enzymes Eco RI and Xho I and ligated into the Eco RI and Xhol restrictions sites of the yeast expression vector pYES2 to generate three independent libraries.
  • This strategy allowed the directional insert of cDNAs into the expression vector in the sense orientation downstream of a galactose-inducible promoter.
  • Aliquots of the ligated cDNA in pYES2 was used to transform ultracompetent XL 1 -Gold bacteria (Stratagene) to determine optimal conditions for large scale transformation. Typically five ul aliquots of the ligation mixture were used to transform 10 to 20 100 ul aliquots of ultracompetent XL-1 gold bacteria to generate a population of primary transformants. Analysis of individual transformants indicated cDNA inserts ranging from 400 to 3000 nt with ⁇ 70-80% of the transformants containing cDNA inserts.
  • the primary transformants were then amplified by solid phase agarose growth for 48 hrs and plasmid DNA was isolated using column fractionation using the Qaigen 7 system.
  • Each cDNA library was then used to transform the yeast strain CY1316/1183, which expresses a chimeric Gpal-G ⁇ i2 G protein ⁇ subunit and is auxotrophic for histidine (described above).
  • the screen took advantage of the inducible expression of the library cDNAs by differential plating of transformants on glucose or galactose containing plates. Based on a series of preliminary experiments using different amounts of cDNA and yeast, the primary screen was optimized to generate 100,000 to 300,000 transformants. Primary transformants were first plated on sucrose containing medium and subsequently transferred to selective medium by replica plating . The selective medium lacks histidine and thus growth is inhibited in the yeast strain.
  • Transformants exhibiting growth in the primary screen were rescreened by spot growth assays. Transformants exhibiting growth in the primary screen were re-screened on medium lacking histidine and containing either glucose or galactose as carbon source. Transformants were identified as positive in the second screen if they exhibited a galactose-dependent growth phenotype. These were then further propagated to isolate plasmid DNA. The isolated plasmid was then used to retransform yeast and verify that the cDNA conferred galactose dependent growth in the selective media. The cDNAs conferring such a response were considered positive and were further evaluated by nucleotide sequencing, RNA blot analysis and epistatic experiments in different yeast strains.
  • Replicas of each plate were then made onto galactose medium lacking uracil, tryptophan and histidine, and containing 1 mM aminotriazole (AT). Replica plates were incubated at 30° for 3 days and scored for growth. - 7o -
  • Cells from each patch were resuspended in sterile water, and equal numbers of cells were spotted onto both glucose and galactose plates lacking uracil, tryptophan and histidine, and containing 1-2 mM AT (aminotrazole). Plates incubated at 30° for 2 days prior to photographing.
  • RNA blot analysis was ; rfo ⁇ r ⁇ ed as previously described Duzic and Lanier (1992) J. Biol. Chem. 267:24045-24052.
  • the lambdaZapII TM rat brain cDNA library was screened and isolated clones processed as previously described (Lanier et.al. 1989).
  • a Multiple Tissue Northern Blot (CLONTECH ® ) was hybridized with radiolabeled random-primed probes (GPM I - coding region, GPM II - nt 2261 to 2692) and the film exposed for four days.
  • the Multiple Tissue Northern Blot includes mRNA samples form the following tissues: heart, brain, spleen, lung, liver, skeletal muscle, kidney and testis.
  • G protein modulators I and II G protein modulators I and II
  • the clones were inactive in yeast strains null for STE5, STE4 or STE20, which function just distal to activated G-protein in this system.
  • GPM1 and GPM2 were also not functional in a yeast strain expressing its native G ⁇ , Gpal (1316/1 179), both activators were functional in a yeast strain expressing a Gpal/G ⁇ i2 chimeric mutant that cannot exchange GDP for GTP (1316/5533).
  • Yeast strains 1316/1 183 (Gpal/G ⁇ -2), 1316/5533 (Gpal/G ⁇ i2-G204A), 1316/1127 (no G ⁇ ), and 1316/1179 (Gpal) were transformed with plasmids pYES, 3- 37, and 3-53, and transformants plated on sucrose medium lacking uracil and tryptophan (S-UT).
  • Equal numbers of cells from independent isolates were spotted onto S-UT, as well as glucose and galactose plates lacking uracil, tryptophan and histidine, containing 1, 2, 5, or lO mM AT. Plates were grown 2 days, 30°. Whereas transformants expressing 3-37 or 3-53 and carrying either no G ⁇ , the Gpal/G ⁇ i2 chimera or the mutant Gpal/G ⁇ i2-G204A chimera were all able to grow at all concentrations of AT tested, transformants expressing 3-37 or 3-53 and carrying yeast Gpal were unable to grow.
  • activators 3-37 and 3-53 both require the presence of an intact heterotrimeric G protein in order to function, and though this function appears to show some specificity for different G alpha subunits, neither activator appears to require GTP exchange on the G ⁇ in order to function. Thus, these activators appear to be functioning through a novel pathway, one that does not require nucleotide exchange at the G ⁇ subunit.
  • Clone #37 contained an insert of -770 nt. A polyadenylation site was found at nt 768. The first 73 nt contain GA repeats followed by an open reading frame encoding a peptide of 1 13 amino acids. The translational start methionine is found at nt 102-104 and the stop codon is at nt 441-443. Clone #37 is also referred to herein as GPM I ( Figure 3). Northern blot analysis GPM I mRNA ( ⁇ 900nt) is enriched in lung, kidney and testis. Data base analysis A blastn search of the data bases for homologous sequences indicated that GPM I is identical to mouse tctexl (M25825). The data base search also indicated significant amino acid sequence homology with the human RP3 gene (U02556, P51808).
  • the tctexl gene is a component of the mouse T complex and is implicated in the generation of sterile mouse due to chromosome 17 inversion. Lader et.al. (1989) Cell 58(5):969-79. The functional role of tctex in this phenotype is unclear but may involve a defect in sperm motility. Recent data indicate that the tctex 1 protein subserves functions in tissues other than sperm. The tctexl exhibits homology to the RP3 gene that encodes a protein of unknown function but which is implicated in X-linked retinal pigmentosa type 3. Roux et.al. (1994) Human Mol. Genetics 3(2):257-63.
  • the tctexl protein was isolated from a rat brain cDNA library in a yeast two hybrid screen in which the cytosolic tyrosine kinase fyn was used as bait indicating a potential role tctexl in signal transduction.
  • Kai et.al. (1997) J. Neurosci. Res. 48(5):407-424.
  • the tctex protein was identified as a light chain of the cytoplasmic motor protein dynein. Patelking et.al. (1997) J. Cell Biol. 137(5): 1081-1090; and King et.al. (1996) J. Biol. Chem. 271(50):32281 -32287.
  • Dynein along with kinensin and myosin represent a family of motor proteins that perform critical functions in the cell including the shuttling of vesicles within the cell, the organization of cytoskeletal components and the alignment of chromosomes during meiosis. It is of interest to note that vesicular transport and actin/tubulin dynamics are associated with GTP hydrolysis and nucleotide exchange. Indeed, the G-protein Go plays a critical role in vesicular transport in neuronal cells. Immunoblots indicate expression of tctex in rat brain, skeletal muscle and sperm cells.
  • tctexl as a stoichiometric component of cytoplasmic dynein and the bioactivity associated with GPM I suggests a regulatory role of tctexl in dynein function that involves a heterotrimeric G-protein. Indeed, both Gi ⁇ 3 and Go ⁇ are implicated in various aspects of membrane trafficking and vesicular transport. Denker et al. (1996) J. Cell. Biol. 1333:1027-1040 and Devries et al. (1998) Mol. Biol. Cell 9:1 123-1134. Tctexl also exists in the cytoplasm free of dynein where it may subserve as yet undefined roles in cellular signaling. Tai et al. ,Supra and Campbell et al. (1998) J. Immunol. 161 :1728-1737. One such role may involve a direct regulation of heterotrimeric G-protein signaling pathways. ANALYSIS OF GPM II
  • insert Clone #53 included an insert of 1.478 kb which contained a truncated open reading frame at its 5' terminus. Clone #53 is also referred to herein as GPM II (SEQ ID NOT). A potential polyadenylation site is found at ⁇ nt 1460.
  • clone #53 contains a large segment of the 3' untranslated regions of the mRNA. Indeed, truncation of the 3' end of clone #53 to nt 480 resulted in a clone that again conferred galactose dependent growth in the selective media indicating the importance of the first 480 nt of the clone in the biological activity.
  • the 84 amino acid reading frame (Frame 1, SEQ ID NO:2) began at nt 1 and terminated at nt 252.
  • the 84 amino acid peptide contained a methionine at amino acid residue #1 1 and this ATG codon is positioned with an adenine at position -3.
  • this methionine is placed in the context of Kozak's consensus sequence for translational initiation and may represent a true translational start site for Frame 1.
  • the peptide encoded by Frame 1 would be 74 amino acids beginning at nt 31 , the peptide having a predicted molecular weight of -8266 Da (SEQ ID NO:3).
  • the first methionine (residue #6 of the 119 amino acid coding sequence; nt 17-19) is positioned with a guanine at position - 3 to the ATG and thus is also in the context of a Kozak's consensus sequence for translational initiation and would lead to translation of a peptide 1 14 amino acids in length that terminated at nt 358 (stop codon at 359-361 ; calculated molecular weight -12081 (SEQ ID NO-13).
  • the first 333 nt exhibits significant homology to the 3' end of a mouse mRNA in GenBank (L23316.gb_ro), termed C10A (SEQ ID NO: 14). Only one nt differs when the sequences of GPM II and C10A are aligned for comparison purposes ⁇ The-guanine appearing at nucleotide 236 in the GPM II cDNA described herein is replaced by an adenine at the equivalent position in the C10A cDNA sequence.
  • Nt 844-1391 - exhibited significant homology to EST clones AA271904, AA036169.
  • C10A A blastp search using the peptide sequence encoded by open reading frame 1 indicated identity (with one amino acid change) to the incomplete protein encoded by L23316.gb_ro, termed C10A, which was isolated by subtractive cloning of murine erythroleukemia mRNAs.
  • the C10A cDNA sequence (SEQ ID NOT 1) encodes a predicted protein which is at least 255 amino acid residues in length and contains at least four GPR motifs from about gly66 - alal03, about ala 1 17 - prol54, about leul 66 - pro 199, and about asp200 - glu237 (amino acids 66-103, 1 17-154, 166-199, and 200-237 of SEQ ID NOT 1, respectively).
  • the one nucleotide difference in the two sequences results in a tyrosine in the L23316.gb_ro encoded protein and a cysteine in clone #53.
  • the one nucleotide difference in clone #53 was confirmed in both directions with multiple primers suggesting three possibilities 1) species difference, 2) sequencing error in the genebank entry of L23316.gb_ro or 3) a point mutation in clone #53.
  • L23316.gb_ro cDNA clone appears to be a partial clone incomplete at both the 3' and 5' end.
  • the longest open reading frame encoded by L23316.gb_ro generates a peptide of 256 amino acids.
  • the 84 amino acid peptide encoded by clone #53 frame 1 aligns to the carboxy terminus of L23316.gb_ro with the exception of the C/Y discrepancy. From this analysis, it appears that both clone #53 and L23316.gb_ro represent partial transcripts of longer entire gene sequences, with the last 84 amino acids of the L23316.gb_ro protein sharing significant homology to GPM II. With regards to the truncation of GPM II, it is possible that the truncation is due to incomplete extension of the 1 st strand synthesis during library construction. Alternatively, the GPM II gene truncated gene products may represent biologically active peptides expressed by NG108-15 cells.
  • a blastp search using the peptide sequence encoded by open reading frame 1 did not reveal any high homology with any entry in the data base.
  • the highest scoring sequence in this search was a C. elegans gene (U41544) (36% identity, 51% similarity over a 41 amino acid span) of unknown function. This degree of homology is low and of unclear significance.
  • Three sets of overlapping phage clones were isolated. Two of the clones contained inserts of -2.4 kb while the third clone contained an insert of -1 kb. The shorter clone corresponded to nt 1225 - nt 2338 of clone #53. Relative to clone #53 the larger clones exhibited 92% nt sequence identity that encompassed nt 1 to 676 of clone #53. The largest insert extended the 5' end of clone #53 by 566 in-frame amino acids. The extended amino acid sequence was identical to L23316.gb_ro with the exception of a D/E at residue 632, R/Q at residue 642, and C/Y at residue 645.
  • the 5' end of the rat cDNA was further extended by 5'RACE to yield a full length protein of 650 amino acids with a calculated molecular weight of 72,048.
  • the full-length nucleotide sequence is set forth as SEQ ID NO:5.
  • the complete rat brain GPM II sequence is shown in Figure 2.
  • RNA blot analysis using a 3' probe identified mRNAs of -2,300 and -4000 nt that were differentially enriched in heart and brain.
  • Blastn analysis of human EST databases with the GPM II sequence identified cDNAs that exhibited greater homology with GPM II than with human LGN indicating that these genes likely encode two members of a family of proteins with shared structural motifs.
  • TPR tetratricopeptide repeats
  • Non-induced/selection glucose media lacking uracil/tryptophan/histidine (UTH) and containing the histidine analog aminotriazole (AT) at 1 mM.
  • Induced/selection galactose media lacking UTH and containing 1 mM AT.
  • the carboxy terminal half of GPM II contains four repeated sequences of 18-19 amino acids that exhibit 80-85% homology: Domain I - E470 to R489, Domain II - E524 to R542, Domain III - D572 to R590, Domain IV - D606 to R624 ( Figure 4A).
  • the four repeated domains are also found in human LGN and the predicted C. elegans protein. Each of the four repeated domains regions can exist as an amphipathic helix. An amphipathic helix is also observed in the juxtamembrane portions of the 13 loop of several G-protein coupled receptors where it is involved in G-protein recognition activation. Carlson et al. (1998) Mol. Pharm. 53:451-458.
  • Repeat domains I-IV were evaluated by MAST (Motif Alignment and Search Tool) to determine their existence in other proteins in the databases.
  • MAST Motif Alignment and Search Tool
  • the complete 18-19 amino acid repeat or the carboxyl half of the repeat is also found in three proteins that influence the nucleotide binding properties or GTPase activity of G-proteins ( Figure 4B).
  • GPR G-protein regulatory
  • GPM I and GPM II do not share any apparent structural similarities that suggest common mechanisms of G-protein activation.
  • To define the mechanism of G-protein activation by GPM I and GPM II the following issues were investigated: 1) Selectivity of GPM I and GPM II for different G ⁇ subunits; 2) Specificity of interact of GPM I and GPM II with G ⁇ or G ⁇ subunits; and 3) Alteration of nucleotide binding of GPM I and GPM II by G ⁇ subunits?
  • GPM I and GPM II for different G ⁇ were determined using yeast strains that expressed Gpal (yeast G ⁇ ), Gs ⁇ , Gi ⁇ 2 Gpa , (
  • GPM I and GPM II exhibited distinct selectivity profiles for the different G ⁇ subunits.
  • GPM II was only active in the Gi ⁇ 2 and Gi ⁇ 3 genetic backgrounds, whereas GPM I was active in each of the genetic backgrounds except Gpal .
  • the G ⁇ l6 yeast strain exhibited an elevated background signaling due to its relatively lower affinity for yeast G ⁇ .
  • GPM I and the active carboxyl terminus region of GPM II were generated as glutathione-S-transferase fusion proteins, expressed in bacteria or yeast and purified using a glutathione affinity matrix.
  • the GST-GPM II construct when expressed in yeast, was functionally similar to the original GPM II isolate in terms of its ability to promote growth.
  • the GST-GPM I fusion protein did not promote growth in the yeast spot growth assay suggesting that the amino terminus region of
  • GPM I has an important functional role. GPM II specifically bound recombinant Gi ⁇ 2, transducin ⁇ and purified brain G ⁇ , but it did not interact with purified brain G ⁇ ( Figure 6A). GPM II preferred the GDP bound conformation of G ⁇ . In contrast to GPM II, GPM I selectively interacted with purified brain G ⁇ ( Figure 6A). Thus, both the functional and the protein interaction data indicated that GPM I and GPM II interact with different G-protein subunits. GPM I binding to G ⁇ may also have further functional implications unrelated to subunit interactions per se, as GPM I activated the system in the absence of any G ⁇ subunit and this action required G ⁇ .
  • GPM I and GPM II activate heterotrimeric G-protein signaling by a mechanism independent of nucleotide exchange.
  • GPM II may actively promote subunit dissociation and "stabilize" G ⁇ GDP, transiently preventing the interaction of G ⁇ GDP with G ⁇ .
  • GPM II may simply compete with G ⁇ for binding to the G ⁇ subunit. Signal duration would then depend upon the half-life of the GPM II-G ⁇ GDP complex and reassociation of G ⁇ GDP with G ⁇ .
  • GPM I interacts with the G ⁇ subunit. Again, the interaction of GPM I with G ⁇ may actively promote subunit dissociation independent of nucleotide exchange or GPM I may simply compete with G ⁇ for interaction with G ⁇ .
  • Such mechanisms of G- protein activation dramatically differ from that involving a typical G-protein coupled receptor and might lead to selective activation of G ⁇ -regulated effectors.
  • GPM proteins are indicative of a growing number of accessory proteins that influence signal propagation by heterotrimeric G-protein systems. Lanier: The Construction of Recombinant Receptor Systems and Their Use to Gain Insight Into Receptor Events at the Receptor-G-Protein and G-Protein-Effector Interface. In General Pharmacology - Handbook of Experimental Pharmacology Series. Eds. Born et al. Springer-Verlag (1998). As is the case with GPM proteins, several of the accessory proteins directly influence the activation state of the G-protein. Sato et al. (1995) J. Biol Chem. 270:15269-15276; Sato et al. (1996) J. Biol. Chem.
  • Such entities may: 1) provide a cell-specific mechanism for signal amplification by acting in concert with G-protein coupled receptors; 2) influence the population of activated G-protein effector within the cell independent of external stimuli; 3) serve as "effectors" subject to receptor regulation and/or 4) serve as a mechanism to provide signal input to heterotrimeric G-protein signaling systems that is distinct from that initiated by a seven-membrane span hormone 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)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne de nouvelles protéines polypeptidiques de modulateur de protéine G (GPM), et des molécules d'acide nucléique, qui stimulent l'activité de la protéine G indépendamment d'un récepteur. En outre, cette invention permet de fournir des protéines de fusion GPM isolées, des peptides antigènes, et des anticorps anti-GPM aux protéines GPM isolées pleine longueur. Elle fournit également des molécules GPM d'acide nucléique, des vecteurs d'expression de recombinaison contenant ladite molécule d'acide nucléique, des cellules hôtes dans lesquelles les vecteurs d'expression ont été introduits, et des animaux transgéniques non humains dans lesquels le gène GPM a été introduit ou interrompu. Elle concerne enfin un diagnostic, un criblage et des techniques utilisant lesdites compositions.
EP00930459A 1999-05-07 2000-05-05 Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations Withdrawn EP1179006A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US30710599A 1999-05-07 1999-05-07
US307105 1999-05-07
PCT/US2000/012502 WO2000068249A2 (fr) 1999-05-07 2000-05-05 Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations

Publications (2)

Publication Number Publication Date
EP1179006A2 true EP1179006A2 (fr) 2002-02-13
EP1179006A4 EP1179006A4 (fr) 2003-03-19

Family

ID=23188261

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00930459A Withdrawn EP1179006A4 (fr) 1999-05-07 2000-05-05 Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations

Country Status (3)

Country Link
EP (1) EP1179006A4 (fr)
AU (1) AU4827600A (fr)
WO (1) WO2000068249A2 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999018211A1 (fr) * 1997-10-07 1999-04-15 Cadus Pharmaceutical Corporation Cellules de levure exprimant des proteines modifiees g et procedes d'utilisation associes
WO1999058670A1 (fr) * 1998-05-08 1999-11-18 Cadus Pharmaceutical Corporation Molecules d'acides nucleiques et proteines ags, et utilisations correspondantes
WO2001012805A1 (fr) * 1999-08-16 2001-02-22 The University Of Sydney Methodes de diagnostic et de traitement de maladies humaines, y compris l'hypertension

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999018211A1 (fr) * 1997-10-07 1999-04-15 Cadus Pharmaceutical Corporation Cellules de levure exprimant des proteines modifiees g et procedes d'utilisation associes
WO1999058670A1 (fr) * 1998-05-08 1999-11-18 Cadus Pharmaceutical Corporation Molecules d'acides nucleiques et proteines ags, et utilisations correspondantes
WO2001012805A1 (fr) * 1999-08-16 2001-02-22 The University Of Sydney Methodes de diagnostic et de traitement de maladies humaines, y compris l'hypertension

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
BERNARD M ET AL: "Activators of G-protein signaling, AGS2 and AGS3, interact with different G-protein subunits." FASEB JOURNAL, vol. 13, no. 5 PART 2, 15 March 1999 (1999-03-15), page A797 XP001118172 Annual Meeting of the Professional Research Scientists on Experimental Biology 99;Washington, D.C., USA; April 17-21, 1999 ISSN: 0892-6638 *
CISMOWSKI MARY J ET AL: "Genetic screens in yeast to identify mammalian nonreceptor modulators of G-protein signaling." NATURE BIOTECHNOLOGY, vol. 17, no. 9, September 1999 (1999-09), pages 878-883, XP002218774 ISSN: 1087-0156 *
DATABASE SWISSPROT1 May 2000 (2000-05-01) TAKESONO A. ET AL.: "Activator of G-protein signaling 3, AGS3" Database accession no. Q9R080 XP002218775 -& TAKESONO AYA ET AL.: "Receptor-independent activators of heterotrimeric G-protein signaling pathways." JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 274, no. 47, 19 November 1999 (1999-11-19), pages 33202-33205, XP002218773 *
See also references of WO0068249A2 *
TAKESONO AYA ET AL: "Non-receptor dependent activators of heterotrimeric G-protein signaling pathway." JAPANESE JOURNAL OF PHARMACOLOGY, vol. 79, no. SUPPL. 1, 1999, page 152P XP001117612 72nd Annual Meeting of the Japanese Pharmacological Society;Sapporo, Japan; March 22-25, 1999 ISSN: 0021-5198 *
TAKESONO AYA ET AL: "Receptor-independent activators of Gialpha2 and Gialpha3." CIRCULATION, vol. 110, no. 18 SUPPL., 2 November 1999 (1999-11-02), page I.428 XP001118506 72nd Scientific Sessions of the American Heart Association;Atlanta, Georgia, USA; November 7-10, 1999 ISSN: 0009-7322 *

Also Published As

Publication number Publication date
AU4827600A (en) 2000-11-21
EP1179006A4 (fr) 2003-03-19
WO2000068249A2 (fr) 2000-11-16
WO2000068249B1 (fr) 2002-01-17
WO2000068249A3 (fr) 2001-10-25

Similar Documents

Publication Publication Date Title
US5948639A (en) TGF-β pathway genes
EP1109908B1 (fr) Methodes de la determination des composes pour la modulation du poids corporel
CA2491547A1 (fr) Methodes et compositions permettant de moduler la formation de fibres musculaires de type i a l'aide de pgc-1?-
WO1999058670A1 (fr) Molecules d'acides nucleiques et proteines ags, et utilisations correspondantes
JP2002523078A (ja) 新規なirap−bpポリペプチド及び核酸分子並びにこれらの利用法
US6664068B2 (en) Pablo, a polypeptide that interacts with Bcl-xL, and uses related thereto
AU3755699A (en) Novel lrsg protein and nucleic acid molecules and uses therefor
US6733991B1 (en) AGS proteins and nucleic acid molecules and uses therefor
WO2000053769A1 (fr) Nouvelles molecules du site actif de la famille de la thioredoxine et leurs utilisations
US6746852B1 (en) AGS proteins and nucleic acid molecules and uses thereof
WO2000068249A2 (fr) Nouvelles proteines de modulateur de proteine g (gpm), molecules d'acide nucleique, et leurs utilisations
AU4101599A (en) Novel secreted and membrane-associated proteins and uses therefor
US20010024808A1 (en) Leptin induced genes
JP2002528046A (ja) 新規レセプタースーパーファミリー分子およびそれについての用途
JP2002511480A (ja) T129関連タンパク質ファミリーの新規分子およびその使用
US6410261B2 (en) CIITA-interacting proteins and methods of use therefor
US6268130B1 (en) RP compositions and diagnostic uses therefor
AU6410499A (en) Slgp protein and nucleic acid molecule and uses therefor
WO1999067387A2 (fr) Nouvelles proteines et molecules d'acide nucleique de mp-7 et leurs utilisations
JP2004537277A (ja) 新規グルタミン酸レセプター調節タンパク質および核酸分子およびそれらの使用
JP2003525576A (ja) Nt2lp、ニューロテンシン−2受容体との相同性を有する新規のgタンパク質共役型受容体
WO2001096375A2 (fr) La 33358, nouveau membre de la famille humaine des ankyrines et ses utilisations
US20040157327A1 (en) Pablo, a polypeptide that interacts with BCL-XL, and uses related thereto
WO2003104261A2 (fr) Pablo, polypeptide interagissant avec bcl-xl, et utilisations correspondantes

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

Free format text: AL;LT;LV;MK;RO;SI

RIC1 Information provided on ipc code assigned before grant

Free format text: 7G 01N 33/53 A, 7G 01N 33/567 B, 7C 12P 21/06 B, 7C 12P 21/04 B, 7C 12N 1/20 B, 7C 12N 15/00 B, 7C 12N 15/09 B, 7C 12N 15/63 B, 7C 12N 15/70 B, 7C 12N 15/74 B, 7C 12N 5/00 B, 7C 12N 5/02 B, 7A 01N 37/18 B, 7A 61K 38/00 B, 7C 07K 1/00 B, 7C 07K 14/00 B, 7C 07K 16/00 B, 7C 07K 17/00 B

17P Request for examination filed

Effective date: 20020425

A4 Supplementary search report drawn up and despatched

Effective date: 20030131

17Q First examination report despatched

Effective date: 20050311

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

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

18D Application deemed to be withdrawn

Effective date: 20050722