US20020119135A1 - Inhibitors of the JNK signal transduction pathway and methods of use - Google Patents

Inhibitors of the JNK signal transduction pathway and methods of use Download PDF

Info

Publication number
US20020119135A1
US20020119135A1 US10/115,178 US11517802A US2002119135A1 US 20020119135 A1 US20020119135 A1 US 20020119135A1 US 11517802 A US11517802 A US 11517802A US 2002119135 A1 US2002119135 A1 US 2002119135A1
Authority
US
United States
Prior art keywords
jip
polypeptide
pro
ser
jnk
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.)
Abandoned
Application number
US10/115,178
Inventor
Roger Davis
Martin Dickens
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.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US10/115,178 priority Critical patent/US20020119135A1/en
Publication of US20020119135A1 publication Critical patent/US20020119135A1/en
Priority to US11/474,513 priority patent/US7838294B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy

Definitions

  • the invention relates to signal transduction inhibitors.
  • JNK c-Jun NH2-terminal kinase
  • MAP kinases Mitogen Activated Protein kinases
  • the JNK signal transduction pathway is activated in response to environmental stress and by the engagement of several classes of cell surface receptors, including cytokine receptors, serpentine receptors, and receptor tyrosine kinases. Whitmarsh et al., J. Mol. Med., 74:589 (1996).
  • genetic studies of Drosophila have demonstrated that JNK is required for early embryonic development.
  • JNK has been implicated in the immune response, oncogenic transformation, and apoptosis. JNK mediates these effects, at least in part, by increasing the expression of target genes.
  • Targets of the JNK signal transduction pathway include the transcription factors c-Jun, ATF2, and Elk-1. Whitmarsh et al., supra.
  • JNK is activated in the liver by metabolic oxidative stress. Mendelson et al., Proc. Natl. Acad. Sci. USA 93:12908-12913 (1996). Activation of JNK also occurs in the kidney during stress, for example, during Bchemic renal failure. Demari et al., Am. J. Physiol. 272:F292-F298 (1997). JNK is also activated during cardiovascular disease such as ischemia Ireperfusion and during organ transplantation. Pombo et al., J. Biol. Chem. 269:26546-26551 (1994); Force et al., Circ. Res. 78:947-53 (1996).
  • JNK is located in both the cytoplasmic and the nuclear compartments of quiescent cells, activation of the JNK signal transduction pathway is associated with accumulation of JNK in the nucleus. Mechanisms governing this sub-cellular distribution have not been previously elucidated.
  • Anchor or tethering proteins play an important role in the regulation of multiple signal transduction pathways. These anchor proteins, which include the nuclear factor kappa B (NFkB) inhibitor IkB, the A kinase anchor protein (AKAP) group of proteins that bind the type II cyclic adenosine monophosphate (AMP) dependent protein kinase, and the p190 protein that binds Ca 2+ -calmodulin-dependent protein kinase II, localize their tethered partners to specific sub-cellular compartments. Verma et al., Genes & Dev., 9:2723 (1995); McNeill et al., J. Biol.
  • NFkB nuclear factor kappa B
  • AKAP A kinase anchor protein
  • Anchor proteins also target enzymes to specific substrates, and create multi-enzyme signaling complexes, such as the Ste5 MAP kinase scaffold complex and the AKAP79 kinase/phosphatase scaffold complex. Choi et al., Cell, 78:499 (1994); Klauck et al., Science, 271:1589 (1996); Faux et al., Cell, 85:9 (1996)).
  • JIP-1 JNK-interacting protein 1
  • SEQ ID NO:1 JNK-interacting protein 1
  • JIP-1 JNK-interacting protein 1
  • JIP-1 specifically binds to and inhibits the biological effects of JNK, including the initiation of apoptosis and oncogenic transformation. JIP-1 is therefore useful as a therapeutic agent for treating pathological conditions characterized by apoptosis or transformation.
  • JIP-1 compositions can be used to treat neurodegenerative diseases characterizediby apoptosis, including Parkinson's disease and Alzheimer's disease; and blood clots, which left untreated could result in stroke and associated memory loss.
  • Other conditions that can be treated using the compositions and methods of the invention are autoimmune diseases such as arthritis; other conditions characterized by inflammation; and malignancies, such as leukemias, e.g., chronic myelogenous leukemia (CML).
  • Other conditions that can be treated with JIP-1 compositions include oxidative damage to organs such as the liver and kidney, and heart disease, particularly damage due to ischemia/reperfusion and cardiomyopathy.
  • JIP-1 compositions can also be used to treat donor organs for transplantation. These organs are exposed to substantial environmental stress, the effects of which are blocked by JNK inhibitors such as JIP-1.
  • JIP-1 polypeptide is a protein having an amino acid sequence that specifically binds JNK to the same extent, or at least 10% of the binding activity of wildtype JIP-1.
  • Such polypeptides can be from 5 to 200 amino acids in length, e.g., from 10 to 100 amino acids in length, or from 20 to 50 amino acids in length.
  • Such polypeptides include the JNK Binding Domain (JBD), or portions thereof, of JIP-1 (e.g., amino acids 148 to 174, forming the “core” of the JBD of wildtype JIP-1, shown in FIG. 2, and having the sequence SGDTYRPKRPTTLNLFPQVPRSQDTLN; SEQ ID NO:3).
  • JIP-1 polypeptides are preferably derived from a mammal, such as a mouse or a human.
  • the polypeptide is soluble, the polypeptide includes the JNK-binding domain of JIP-1 or a portion thereof, the polypeptide is at least 80%, 90%, or 100% identical to the amino acid sequence from amino acid 148 to amino acid 174 of JIP-1 (the core JNK-binding domain; SEQ ID NO:3), or the polypeptide has an amino acid sequence identical to the amino acid sequence from amino acid 148 to amino acid 174 of JIP-1 (SEQ ID NO:3), or the polypeptide is at least 80%, 90%, or 100% identical to the amino acid sequence from amino acid 127 to amino acid 281 of JIP-1 (the JNK-binding domain; SEQ ID NO:4).
  • polypeptides of the invention can be modified to enhance their uptake by cells. Such modifications increase the hydrophobicity of molecules to facilitate passage through the lipid bilayer of the cell membrane.
  • polypeptides can be complexed with myristic acid or packaged in liposomes.
  • JIP-1 polypeptides can be complexed with hydrophobic moieties (e.g., lipids) or peptides that increase the delivery of proteins into cells.
  • the invention also includes peptide mimetics of JIP-1 polypeptides.
  • a “peptide mimetic” of a known polypeptide is a compound that mimics the activity of the peptide or polypeptide, but which is composed of molecules other than, or in addition to, amino acids.
  • polypeptide is meant any chain of amino acids, regardless of length or post-translational modification (e.g., glycosylation or phosphorylation), and thus includes peptides, proteins, and fusion proteins.
  • a “substantially identical” polypeptide sequence differs from a given sequence only by conservative amino acid substitutions or by one or more nonconservative substitutions, deletions, or insertions located at positions which do not destroy the function of the polypeptide compared to wildtype JIP-1.
  • Polypeptides of the invention can be 70%, 80%, 85%, 90%, or 95% identical to wildtype JIP-1.
  • a “substantially pure” preparation is at least 60% by weight of the compound of interest, e.g., a JIP-1 polypeptide or fragment of a JIP-1 polypeptide. Preferably the preparation is at least 75%, more preferably at least 90%, and more preferably at least 95% by weight of the compound of interest. Purity can be measured by any appropriate standard method, e.g., column chromatography, polyacrylamide gel electrophoresis, or High Pressure Liquid Chromatography (HPLC) analysis.
  • HPLC High Pressure Liquid Chromatography
  • polypeptides of the invention include, but are not limited to, recombinant polypeptides, natural polypeptides, and synthetic polypeptides, as well as preproteins or proproteins and biologically active fragments.
  • a “biologically active fragment” of JIP-1 is a fragment having at least 50%, 70%, 80%, 90%, 95%, or 100% or greater, of the activity of naturally occurring or synthetic, full length JIP-1.
  • polypeptides of the invention can be physically linked to another polypeptide, e.g., a marker polypeptide.
  • a marker polypeptide e.g., the polypeptide can be fused to a hexa-histidine tag to facilitate purification of bacterially expressed proteins, or a hemagglutinin tag to facilitate purification of protein expressed in eukaryotic cells.
  • the invention features an isolated nucleic acid that includes a sequence encoding a JIP-1 polypeptide or a fragment of such a polypeptide.
  • the nucleic acid is derived from a mammal.
  • the invention also encompasses nucleic acids that hybridize under stringent conditions (as described herein) to a nucleic acid encoding a JIP-1 polypeptide.
  • Stringent conditions include hybridization at 68° C. in 5 ⁇ SSC/5 ⁇ Denhardt's solution/1.0% SDS, or in 0.5 M NaHPO 4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO 4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2 ⁇ SSC/0.1% SDS at room temperature or at 42° C., or in 0.1 ⁇ SSC/0.1% SDS at 68° C., or in 40 mM NaHPO 4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO 4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C.
  • Moderately stringent conditions include washing in 3 ⁇ SSC at 42° C.
  • the parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid. For guidance regarding such conditions see, e.g., Sambrook et al., Molecular Cloninc, A Laboratory Manual , Cold Spring Harbor Press, Cold Spring Harbor, N.Y.; and Ausubel et al., supra, at Unit 2.10.
  • the hybridizing portion of the hybridizing nucleic acid is preferably 20, 30, 50, or 70 bases long.
  • the hybridizing portion of the hybridizing nucleic acid can be 95% or even 98% or 100% identical to the sequence of a portion of a nucleic acid encoding a JIP-1 polypeptide.
  • Hybridizing nucleic acids of the type described above can be used as a cloning probe, a primer (e.g., a PCR primer), or a diagnostic probe.
  • Preferred hybridizing nucleic acids encode a polypeptide having some or all of the biological activities possessed by naturally-occurring JIP-1. Thus, they may encode a protein that is shorter or longer than the various forms of JIP-1 described herein.
  • Hybridizing nucleic acids can also encode proteins that are related to JIP-1, e.g., proteins encoded by genes that include a portion having a relatively high degree of identity to a JIP-1 gene described herein.
  • nucleic acid encompasses both RNA and DNA, including cDNA, genomic DNA, and synthetic (e.g., chemically synthesized) DNA.
  • the nucleic acid may be double-stranded or single-stranded. Where single-stranded, the nucleic acid may be the sense strand or the antisense strand.
  • An “isolated nucleic acid” is a nucleic acid that is free of the nucleic acids that normally flank it in the genome.
  • the term therefore includes, e.g., a recombinant nucleic acid incorporated into a vector, such as an autonomously replicating plasmid or virus; a cDNA or genomic DNA fragment produced by polymerase chain reaction (PCR) or restriction endonuclease treatment; and recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequences.
  • a “substantially identical” nucleic acid is a nucleic acid with a sequence that is at least 50%, preferably 70%, and more preferably 85%, 90%, or 95% homologous to a given nucleic acid sequence, e.g., SEQ ID NO:2.
  • the invention also features transformed cells harboring a nucleic acid encompassed by the invention.
  • Vectors and plasmids that include a nucleic acid properly positioned for expression are also within the invention.
  • a “transformed cell” is a cell into which (or into an ancestor of which) has been introduced, by means of recombinant DNA techniques, a DNA molecule encoding a JIP-1 polypeptide.
  • “Operably linked” means that the selected DNA molecule is positioned adjacent to one or more sequence elements that direct transcription and/or translation of the sequence such that the sequence elements can control transcription and/or translation of the selected DNA (i.e., the selected DNA is operably associated with the sequence elements). Such operably associated elements can be used to facilitate the production of a JIP-1 polypeptide.
  • the invention also features purified antibodies which specifically bind a JIP-1 protein or polypeptide.
  • a “purified antibody” is an antibody which is at least 60%, by dry weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated.
  • the preparation can be at least 75%, at least 90%, and up to 99% or more, by dry weight, antibody.
  • an antibody that “specifically binds” an antigen recognizes and binds to that antigen, e.g., a JIP-1 polypeptide.
  • antisense molecules and ribozymes for inhibiting JIP-1 expression are also within the invention.
  • the invention also features antagonists and agonists of JIP-1.
  • Antagonists can inhibit one or more of the functions of JIP-1.
  • Suitable antagonists can include large or small molecules, antibodies to JIP-1, and JIP-1 polypeptides that compete with a native form of JIP-1.
  • Such antagonists include SEQ ID NO:3, a component of an active site of JIP-1, i.e., the JNK-binding domain.
  • Agonists of JIP-1 will enhance or facilitate one or more of the functions of JIP-1.
  • Agonists and Antagonists include polyproline motifs, which bind to SH3 domains such as that found in JIP-1.
  • a “therapeutically effective amount” of a substance is an amount capable of producing a medically desirable result in a treated patient, e.g., inhibition of the expression or activity of a specific protein.
  • FIG. 1A is a schematic diagram showing the structure of a murine JIP-1 polypeptide.
  • FIG. 1B is a representation of the amino acid sequence of a murine JIP-1 polypeptide (SEQ ID NO:1), presented in single letter code.
  • FIG. 1C is a representation of the nucleotide sequence of a murine JIP-1 cDNA (SEQ ID NO:2).
  • FIG. 2 is a diagram showing an alignment of the JBDs (JNK-binding domains) of JIP-1 (SEQ ID NO:3), c-Jun (SEQ ID NO:5), and ATF2 (SEQ ID NO:6).
  • FIG. 3 is a diagram showing the amino acid sequences of wild type and mutant JIP-1 peptides (SEQ ID NOs:3 and 7 to 11), as well as a control peptide (SEQ ID NO:12).
  • FIG. 4A is a bar graph showing the effect of recombinant JBD (JIP-1 residues 127-281) on reporter gene expression mediated by the GAL4 binding domain and GAL4 fusions with the c-myc, Sp1, and VP16 activation domains.
  • FIG. 4B is a bar graph showing the effect of recombinant JBD (JIP-1 residues 127-281) on reporter gene expression mediated by wild type and mutant forms of GAL4-c-Jun, GAL4-ATF2, and GAL4-Elk-1.
  • FIG. 4C is a bar graph showing the effect of wild type and mutant JIP-1 and JED on reporter gene expression mediated by wild type and mutant GAL4-ATF2.
  • FIG. 5 is a bar graph showing the effect of the JBD of JIP-1 on Nerve Growth Factor (NGF) withdrawal-induced apoptosis.
  • NGF Nerve Growth Factor
  • FIG. 6 is a bar graph showing the effect of bicistronic retroviruses on primary mouse bone marrow cells.
  • the invention is based on the molecular cloning and characterization of JIP-1, a cytoplasmic protein that specifically binds JNK.
  • JIP-1 polypeptides cause cytoplasmic retention of JNK and inhibition of JNK-regulated gene expression.
  • JIP-1 polypeptides suppress the effects of the JNK signaling pathway, including oncogenic transformation and apoptosis.
  • autoimmune diseases such as arthritis; other conditions characterized by inflammation; and malignancies, such as leukemias, e.g., chronic myelogenous leukemia (CML).
  • JIP-1 polypeptide compositions can also be used to treat oxidative damage to organs such as the liver and kidney.
  • Heart disease can also be treated with the compositions of the invention.
  • Donor organs for transplantation can also be treated with JIP-1 compositions.
  • the 660 amino acid JIP-1 protein has an SH3 domain at its carboxy terminal end, at amino acid positions 491-540, and a JNK-binding domain (JBD) at its amino terminal end, at amino acid positions 127-281 (SEQ ID NO:4).
  • the core of the JBD is amino acids 148-174 (SEQ ID NO:3).
  • the JED of JIP-1 shares conserved residues with the JNK-binding regions of the transcription factors c-Jun and ATF2, as shown in FIG. 2.
  • JIP-1 polypeptides can be prepared for a wide range of uses including, but not limited to, generation of antibodies, preparation of reagents for diagnostic assays, identification of other molecules involved in transformation or apoptosis, preparation of reagents for use in screening assays for modulators of apoptosis or transformation, and preparation of therapeutic agents for treatment of disorders related to apoptosis or transformation.
  • the invention encompasses, but is not limited to, JIP-1 polypeptides that are functionally related to JIP-1 encoded by the nucleotide sequence of FIG. 1C (SEQ ID NO:2)
  • Functionally related polypeptides include any polypeptide sharing a functional characteristic with wildtype JIP-1 protein, e.g., the ability to bind JNK polypeptides or to affect proliferation or apoptosis.
  • Such functionally related JIP-1 polypeptides include, but are not limited to, polypeptides having additions or substitutions of amino acid residues within the amino acid sequence encoded by the JIP-1 sequences described herein which result in a silent change, thus producing a functionally equivalent gene product. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved.
  • nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid.
  • mutant JIP-1 proteins can be tested for activity
  • site-directed mutations of the JIP-1 coding sequence can be engineered (using site-directed mutagenesis techniques well known to those skilled in the art) to generate mutant JIP-1 proteins with increased function, e.g., greater JNK-1 binding and inhibition of transformation.
  • JIP-1 polypeptides it is useful to distinguish between conserved positions and variable positions.
  • conserved residues are not altered.
  • alteration of non-conserved residues are preferably conservative alterations, e.g., a basic amino acid is replaced by a different basic amino acid.
  • non-conservative changes at variable and/or conserved positions it is preferable to make non-conservative changes at variable and/or conserved positions. Deletions at conserved and variable positions can also be used to create altered function variants.
  • conserved amino acids in JIP-1 include, but are not limited to, Lys-155, Thr-159, Leu-160, Asn-161, and Leu-162.
  • JIP-1 polypeptides are those that bind JNK and inhibit transformation or apoptosis. These JIP-1 polypeptides have 20%, 40%, 50%, 75%, 80%, 90%, or even greater than 100% of the activity of the full-length JIP-1 described herein. Such comparisons are generally based on equal concentrations of the molecules being compared. The comparison can also be based on the amount of protein or polypeptide required to reach 50% of the maximal stimulation obtainable.
  • Polypeptides corresponding to one or more domains of JIP-1 are also within the scope of the invention.
  • Preferred polypeptides are those which are soluble under normal physiological conditions.
  • fusion proteins in which a portion (e.g., one or more domains) of JIP-1 is fused to an unrelated protein or polypeptide (i.e., a fusion partner) to create a fusion protein.
  • the fusion partner can be a moiety selected to facilitate purification, detection, or solubilization, or to provide some other function.
  • Fusion proteins are generally produced by expressing a hybrid gene in which a nucleotide sequence encoding all or a portion of JIP-1 is joined in-frame to a nucleotide sequence encoding the fusion partner.
  • Fusion partners include, but are not limited to, the constant region of an immunoglobulin (IgFc).
  • IgFc immunoglobulin
  • a fusion protein in which a JIP-1 polypeptide is fused to IgFc can be more stable and have a longer half-life in the body than the JIP-1 polypeptide on its own.
  • JIP-1 expressed on its own or fused to a solubilization partner, e.g., an immunoglobulin.
  • JIP-1 polypeptides can be produced by transformation (transfection, transduction, or infection) of a host cell with all or part of a JIP-1-encoding DNA fragment (e.g., the cDNA described herein) in a suitable expression vehicle.
  • suitable expression vehicles include: plasmids, viral particles, and phage.
  • baculovirus expression vectors are suitable. The entire expression vehicle, or a part thereof, can be integrated into the host cell genome.
  • an inducible expression vector e.g., the LACSWITCHTM Inducible Expression System (Stratagene; LaJolla, Calif.).
  • the JIP-1 protein can be produced in a prokaryotic host (e.g., E. coli or B. subtilis ) or in a eukaryotic host, e.g., yeast, such as Saccharomyces or Pichia; mammalian cells, such as COS, NIH 3T3, CHO, BHK, 293, or HeLa cells; or insect cells.
  • a prokaryotic host e.g., E. coli or B. subtilis
  • yeast such as Saccharomyces or Pichia
  • mammalian cells such as COS, NIH 3T3, CHO, BHK, 293, or HeLa cells
  • insect cells e.g., insect cells.
  • JIP-1 polypeptides can also be produced by plant cells.
  • Viral expression vectors e.g., cauliflower mosaic virus and tobacco mosaic virus
  • plasmid expression vectors e.g., Ti plasmid
  • Plant cells are available from a wide range of sources, e.g., the American Type Culture Collection, Rockland, Md. See also, e.g., Ausubel et al., Current Protocols in Molecular Biology , John Wiley & Sons, New York, 1995. The methods of transformation or transfection and the choice of expression vehicle will depend on the host system selected.
  • Transformation and transfection methods are described in, e.g., Ausubel et al., supra; expression vehicles may be chosen from those described in, e.g., Cloning Vectors: A Laboratory Manual , P. H. Pouwels et al., 1985, Supp. 1987.
  • the host cells harboring the expression vehicle can be cultured in conventional nutrient media adapted as needed for activation of a chosen gene, repression of a chosen gene, selection of transformants, or amplification of a chosen gene.
  • a suitable expression system is the mouse 3T3 fibroblast host cell transfected with a pMAMneo expression vector (Clontech, Palo Alto, Calif.).
  • pMAMneo provides an RSV-LTR enhancer linked to a dexamethasone-inducible MMTV-LTR promotor, an SV40 origin of replication which allows replication in mammalian systems, a selectable neomycin gene, and SV40 splicing and polyadenylation sites.
  • DNA encoding a JIP-1 in protein would be inserted into the pMAMneo vector in an orientation designed to allow expression.
  • the recombinant JIP-1 protein would be isolated as described below.
  • Other preferable host cells that can be used in conjunction with the pMAMneo expression vehicle include COS cells and CHO cells (ATCC Accession Nos. CRL 1650 and CCL 61, respectively).
  • JIP-1 polypeptides can be expressed as fusion proteins.
  • the expression vector pUR278 can be used to create lacZ fusion proteins. See Ruther et al., EMBO J. 2:1791 (1983).
  • the pGEX vectors can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can be easily purified from cell lysates by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • a JIP-1 coding sequence can be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of a AcNPV promoter, e.g., the polyhedrin promoter.
  • Successful insertion of a gene encoding a JIP-1 polypeptide or protein will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e.,, virus lacking the proteinaceous coat encoded by the polyhedrin gene).
  • Spodoptera frugiperda cells are then infected with these viruses, and the inserted gene is expressed. See, e.g., Smith et al., J. Virol. 46:584 (1983); Smith, U.S. Pat. No. 4,215,051.
  • the JIP-1 nucleic acid sequence can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence, to form a chimeric gene.
  • This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., the E1 or E3 gene) will result in a recombinant virus that is viable and capable of expressing a JIP-1 gene product in infected hosts. See, e.g., Logan, Proc. Natl. Acad. Sci. USA, 81:3655 (1984).
  • Specific initiation signals may be required for efficient translation of inserted nucleic acid sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire native JIP-1 gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. In other cases, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. Exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements. Bittner et al., Methods in Enzymol., 153:516 (1987).
  • a host cell may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in a desired fashion. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product can be used.
  • mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, and WI38 cells.
  • a JIP-1 protein can be produced by a stably-transfected mammalian cell line.
  • a number of vectors suitable for stable transfection of mammalian cells are available to the public. See, e.g., Pouwels et al., supra. Methods for constructing such cell lines are also publicly available. See, e.g., Ausubel et al., supra.
  • JIP-1 cDNA can be cloned into an expression vector that includes the dihydrofolate reductase (DHFR) gene. Methotrexate (0.01-300 ⁇ M) is present in the culture medium to select for cells which have integrated the plasmid and, therefore, the JIP-1 cDNA. See Ausubel et al., supra. This dominant selection can be accomplished in most cell types.
  • Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene.
  • Methods for selecting cell lines bearing gene amplifications are described in Ausubel et al., supra. Such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate.
  • DHFR-containing expression vectors commonly used for this purpose include pCVSEII-DHFR and pAdD26SV(A). See Ausubel et al., supra.
  • Any of the host cells described above or, preferably, a DHFR-deficient CHO cell line are among the host cells preferred for DHFR selection of a stably-transfected cell line or DHFR-mediated gene amplification.
  • a number of other selection systems can be used, including but not limited to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyl-transferase, and adenine phosphoribosyltransferase genes can be employed in tk, hgprt, or aprt cells, respectively.
  • gpt which confers resistance to mycophenolic acid (Mulligan et al., Proc. Natl. Acad. Sci. USA, 78:2072 (1981); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., J. Mol. Biol., 150:1 (1981)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene, 30:147 (1981)), can be used.
  • any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed.
  • a system described in Janknecht et al., Proc. Natl. Acad. Sci. USA, 88:8972 (1981) allows for the ready purification of non-denatured fusion proteins expressed in human cell lines.
  • the gene of interest is subcloned into a vaccinia recombination plasmid such that the gene's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni 2+ nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • JIP-1 or a portion thereof can be fused to an immunoglobulin Fc domain.
  • a fusion protein can be readily purified using a protein A column.
  • fusion proteins permit the production of a dimeric form of a JIP-1 polypeptide having increased stability in vivo.
  • JIP-1 protein After the recombinant JIP-1 protein is expressed, it is isolated. Secreted forms can be isolated from the culture media, while non-secreted forms must be isolated from the host cells. Proteins can be isolated by affinity chromatography. An anti-JIP-1 antibody (e.g., produced as described herein) is attached to a column and used to isolate the JIP-1 protein. Lysis and fractionation of JIP-1 protein-harboring cells prior to affinity chromatography can be performed by standard methods. See, e.g., Ausubel et al., supra.
  • a JIP-1 fusion protein for example, a JIP-1-maltose binding protein, a JIP-1- ⁇ -galactosidase, or a JIP-1-trpE fusion protein, can be constructed and used for JIP-1 protein isolation. See, e.g., Ausubel et al., supra; New England Biolabs, Beverly, Mass.
  • the recombinant protein can, if desired, be further purified, e.g., by high performance liquid chromatography using standard techniques. See, e.g., Fisher, Laboratory Techniques In Biochemistry And Molecular Biology , Work et al., eds., Elsevier (1980).
  • Polypeptides of the invention can also be produced by chemical synthesis, e.g., by the methods described in Solid Phase Peptide Synthesis, 2nd ed., The Pierce Chemical Co., Rockford, Ill., 1984.
  • the invention also features proteins which interact with JIP-1 and which are involved in the function of JIP-1. Also included in the invention are the genes encoding these interacting proteins. Interacting proteins can be identified using methods known to those skilled in the art. One method suitable method is the “two-hybrid system” which detects protein interactions in vivo. See, e.g., Chien et al., Proc. Natl. Acad. Sci. USA, 88:9578 (1991). A kit for practicing this method is available from Clontech (Palo Alto, Calif.).
  • JIP-1 cDNA sequences described herein, and related family members of the JIP-1 gene present in mouse, human, or other species can be identified and readily isolated without undue experimentation by well known molecular biological techniques given the specific sequences described herein. Further, genes may exist at other loci that encode proteins having extensive homology to JIP-1 polypeptides or one or more domains of JIP-1 polypeptides. These genes can be identified by known techniques using the sequences disclosed herein. For example, hybridization of JIP-1 probes to homologous nucleic acids is performed under stringent conditions. Alternatively, a labeled fragment can be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions. Such stringent conditions are well known, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived.
  • T m is the temperature at which a probe dissociates from a target DNA. This melting temperature is used to define the required stringency conditions. If sequences are to be identified that are related and substantially identical to the probe, rather than identical, then it is useful to first establish the lowest temperature at which only homologous hybridization occurs with a particular concentration of SSC or SSPE. It is then assumed that 1% mismatching results in a 1° C. decrease in the T m , and the temperature of the final wash is reduced accordingly (for example, if sequences with ⁇ 95% identity with the probe are sought, the final wash temperature is decreased by 5° C.). Note that this assumption is very approximate, and the actual change in T m can be between 0.50° and 1.5° C. per 1% mismatch.
  • stringent conditions include hybridization at 68° C. in 5 ⁇ SSC/5 ⁇ Denhardt's solution/1.0% SDS, or in 0.5 M NaHPO 4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO 4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2 ⁇ SSC/0.1% SDS at room temperature or at 42° C., or in 0.1 ⁇ SSC/0.1% SDS at 68° C., or in 40 mM NaHPO 4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO 4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C.
  • Moderately stringent conditions include washing in 3 ⁇ SSC at 42° C.
  • the parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid. For guidance regarding such conditions see, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual , Cold Spring Harbor Press, Cold Spring Harbor, N.Y.; and Ausubel et al., supra, at Unit 2.10.
  • cDNA libraries can be constructed from RNA isolated from these cell lines utilizing standard techniques. The human cDNA library can then be screened with a JIP-1 probe to isolate a human JIP-1 cDNA.
  • a human genomic DNA library can be screened using JIP-1 probes. Hybridizing clones can be sequenced and the intron and exon structure of the human JIP-1 gene can be elucidated. Once a genomic sequence is obtained, oligonucleotide primers can be designed based on the sequence for use in Reverse Transcriptase-coupled PCR, e.g., to isolate human JIP-1 cDNA.
  • An example of a suitable probe for screening a human genomic library is the coding region (nucleotides 180 to 2159) of the mouse JIP-1 cDNA (SEQ ID NO:2).
  • a previously unknown gene sequence can be isolated by performing PCR using two degenerate oligonucleotide primer pools designed on the basis of nucleotide sequences within the JIP-1 cDNA defined herein.
  • Degenerate PCR primers that can be used include: 5′GARGARTTYGARGAYGARGA 3′ (sense; SEQ ID NO:25); 5′GGNAARAARCAYAGNTGGCA 3′ (sense; SEQ ID NO:26); 5′CATRTTWTANGCYTCWTACCA 3′ (antisense; SEQ ID NO:27); and 5′AAYTGYTTKTARAAYTGYTGRAA 3′ (antisense; SEQ ID NO:28), where W is A or T, K is G or T, R is A or G, Y is C or T, and N is A, C, G or T.
  • the template for the reaction can be cDNA obtained by reverse transcription of mRNA prepared from human or non-human cell lines or tissue known to express, or suspected of expressing, JIP-1.
  • the PCR product can be subcloned and sequenced to insure that the amplified sequences represent the sequences of JIP-1 or JIP-1-like gene nucleic acid sequences.
  • the PCR fragment can then be used to isolate a full length cDNA clone by a variety of methods.
  • the amplified fragment can be labeled and used to screen a cDNA library in bacteriophage.
  • the labeled fragment can be used to screen a genomic library.
  • RNA can be isolated, following standard procedures, from an appropriate tissue or cell line.
  • a reverse transcription reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5′ end of the amplified fragment for the priming of first strand synthesis.
  • the resulting RNA/DNA hybrid can then be “tailed” with guanines using a standard terminal transferase reaction.
  • second strand synthesis can be primed with a poly-C primer.
  • cDNA sequences upstream of the amplified fragment can easily be isolated.
  • Mutant cDNAs can also be isolated using PCR techniques.
  • the first cDNA strand can be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue known to be or suspected of being expressed in an individual putatively carrying the mutant allele, and by extending the new strand with reverse transcriptase.
  • the second strand of the cDNA can then be synthesized using an oligonucleotide that hybridizes specifically to the 5′-end of the normal gene.
  • the product is amplified via PCR, cloned into a suitable vector, and subjected to DNA sequence analysis by methods well known in the art. By comparing the DNA sequence of the mutant gene to that of the normal gene, the mutation(s) responsible for the loss or alteration of function of the mutant gene product can be ascertained.
  • a genomic or cDNA library can be constructed and screened using DNA or RNA, respectively, from a tissue known to express, or suspected of expressing, the gene of interest in an individual suspected of carrying or known to carry the mutant allele.
  • the normal gene or any suitable fragment thereof can then be labeled and used as a probe to identify the corresponding mutant allele in the library.
  • the clone containing the mutant gene can then be purified through methods routinely practiced in the art, and subjected to sequence analysis using standard techniques as described herein.
  • an expression library can be constructed using DNA isolated from or cDNA synthesized from a tissue known to express or suspected of expressing the gene of interest in an individual suspected of carrying or known to carry the mutant allele.
  • gene products made by this tissue can be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against the normal gene product, as described herein.
  • screening techniques see, for example, Harlow et al., eds., Antibodies: A Laboratory Manual , Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1988).
  • JIP-1 peptide mimetics can be constructed by structure-based drug design through replacement of amino acids by organic moieties. See, e.g., Hughes, Philos. Trans. R. Soc. Lond., 290:387-394 (1980); Hodgson, Biotechnol., 9:19-21 (1991); Suckling, Sci. Prog., 75:323-359 (1991).
  • the use of peptide mimetics can be enhanced through the use of combinatorial chemistry to create drug libraries.
  • the design of peptide mimetics can be aided by identifying amino acid mutations that increase or decrease JIP-1 binding to JNK.
  • yeast two hybrid method see, e.g., Chien et al., Proc. Natl. Acad. Sci. USA, 88:9578 (1991); kit from Clontech, Palo Alto, Calif.
  • the two hybrid method detects protein-protein interactions in yeast. Fields et al., Nature, 340:245-246 (1989).
  • the phage display method detects the interaction between an immobilized protein and a protein that is expressed on the surface of phages (e.g. lambda and M13). Amberg, et al., Strategies, 6:2-4 (1993); Hogrefe et al., Gene, 128:119-126 (1993). These methods allow positive and negative selection for protein-protein interactions and the identification of the sequences that determine these interations.
  • JIP-1 polypeptides can also be expressed in transgenic animals.
  • Animals of any species including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, can be used to generate JIP-1-expressing transgenic animals.
  • JIP-1 transgene a JIP-1 transgene into animals to produce the founder lines of transgenic animals.
  • Such techniques include, but are not limited to, pronuclear microinjection (U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (van der Putten et al., Proc. Natl. Acad. Sci. USA, 82:6148 (1985)); gene targeting into embryonic stem cells (Thompson et al., Cell, 56:313 (1989)); and electroporation of embryos (Lo, Mol. Cell. Biol, 3:1803 (1983)).
  • the present invention provides for transgenic animals that carry the JIP-1 transgene in all their cells, as well as animals that carry the transgene in some, but not all of their cells, i.e., mosaic animals.
  • the transgene can be integrated as a single transgene or in concatamers, e.g., head-to-head tandems or head-to-tail tandems.
  • the transgene can also be selectively introduced into and activated in a particular cell type. Lasko et al., Proc. Natl. Acad. Sci. USA, 89:6232 (1992).
  • the regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • the JIP-1 transgene When it is desired that the JIP-1 transgene be integrated into the chromosomal site of the endocenous JIP-1 gene, gene targeting is preferred.
  • Vectors containing some nucleotide sequences homologous to an endogenous JIP-1 gene are designed for the purpose of integrating via homologous recombination into the endogenous gene and disrupting its function.
  • the transgene also can be selectively introduced into a particular cell type, thus inactivating the endogenous JIP-1 gene in only that cell type. See Gu et al., Science, 265:103 (1984).
  • the regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • the expression of the recombinant JIP-1 gene can be assayed utilizing standard techniques. Initial screening can be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals can also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and RT-PCR. Samples of tissues expressing JIP-1 can also be evaluated immunocytochemically using antibodies specific for the JIP-1 transgene product.
  • JIP-1 is an inhibitor of JNK
  • inhibition of JIP-1 increases JNK activity.
  • Increased JNK expression results in increased apoptosis, e.g., in neurons. Induction of apoptosis would be desirable in brain tumors, for example. Therefore, antibodies specific for JIP-1 can be used to inhibit JIP-1 expression.
  • Human JIP-1 proteins and polypeptides can be used to raise antibodies; such polypeptides can be produced by recombinant or peptide synthetic techniques. See, e.g., Solid Phase Peptide Synthesis , supra; Ausubel et al., supra.
  • the peptides can be coupled to a carrier protein, such as KLH, mixed with an adjuvant, and injected into a host mammal.
  • Antibodies can be purified by peptide antigen affinity chromatography.
  • various host animals can be immunized by injection with a JIP-1 protein or polypeptide.
  • Host animals include rabbits, mice, guinea pigs, and rats.
  • Various adjuvants can be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete or incomplete); mineral gels, such as aluminum hydroxide; surface active substances such as lysolecithin; pluronic polyols; polyanions; peptides; oil emulsions; keyhole limpet hemocyanin; dinitrophenol; and potentially useful human adjuvants such as BCG (bacillus Calmette-Guerin) and Corynebacterium parvum .
  • Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of the immunized animals.
  • the invention includes monoclonal antibodies, polyclonal antibodies, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′) 2 fragments, and molecules produced using a Fab expression library.
  • Monoclonal antibodies which are homogeneous populations of antibodies to a particular antigen, can be prepared using the JIP-1 polypeptides described above and standard hybridoma technology. See, e.g., Kohler e: al., Nature, 256:495 (1975); Kohler et al., Eur. J. Immunol., 6:511 (1976); Kohler et al., Eur. J. Immunol., 6:292 (1976) Hammerling et al., In Monoclonal Antibodies and T Cell Hybridomas , Elsevier, N.Y. (1981); Ausubel et al., supra.
  • monoclonal antibodies can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. Such methods include those described in Kohler et al., Nature 256:495 (1975), and U.S. Pat. No. 4,376,110. Other methods of producing monoclonal antibodies include the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72 (1983); Cole et al., Proc. Natl. Acad. Sci. USA 80:2026 (1983), and the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy , Alan R. Liss, Inc., pp. 77-96 (1983)).
  • Such antibodies can be of any immunoglobulin class, e.g., IgG, IgM, IgE, IgA, IgD, and any subclass thereof.
  • Hybridomas producing the monoclonal antibodies of the invention may be cultivated in vitro or in vivo. The ability to produce high titers of monoclonal antibodies in vivo makes this the presently preferred method of production.
  • Polyclonal or monoclonal antibodies are tested for specific JIP-1 recognition by Western blot or immunoprecipitation analysis by standard methods. See, e.g., Ausubel et al., supra. Antibodies that specifically recognize and bind to JIP-1 are useful in the invention. These antibodies can be used in immunoassays to monitor the level of JIP-1 produced by a mammal, e.g., to determine the amount or subcellular location of JIP-1).
  • antibodies of the invention are produced using JIP-1 polypeptides that correspond to regions of the JIP-1 protein that lie outside highly conserved regions and appear likely to be antigenic, by criteria such as high frequency of charged residues.
  • Such fragments can be generated by standard PCR techniques and then cloned into the pGEX expression vector. Ausubel et al., supra. Fusion proteins are expressed in E. coli and purified using a glutathione agarose affinity matrix as described in Ausubel, et al., supra.
  • each fusion protein can be generated for each protein, and each fusion protein can be injected into at least two rabbits.
  • Antisera can be raised by injections in a series, preferably including at least three booster injections.
  • Antisera is also checked for its ability to immunoprecipitate recombinant JIP-1 proteins or control proteins, such as glucocorticoid receptor, CAT, or luciferase.
  • the antibodies can be used to detect JIP-1 in a biological sample as part of a diagnostic assay. Antibodies also can be used in a screening assay to measure the effect of a candidate compound on expression or localization of JIP-1. Additionally, such antibodies can be used in conjunction with the gene therapy techniques, e.g., to evaluate the normal and/or engineered JIP-1-expressing cells prior to their introduction into the patient. Such antibodies additionally can be used in a method for inhibiting abnormal JIP-1 activity. Such abnormal activity includes altered apoptosis and proliferation, resulting in neurodegenerative diseases, autoimmune disease, cancers such as leukemias. Other abnormal JIP-1 activity includes damage caused by ischemia/reperfusion, especially in heart disease, kidney damage, and stroke.
  • murine or other monoclonal antibodies should be altered to make them less immunogenic when administered to human patients.
  • techniques have been developed for the production of “chimeric antibodies.” See Morrison et al., Proc. Natl. Acad. Sci., 81:6851 (1984); Neuberger et al., Nature, 312:604 (1984); Takeda et al., Nature, 314:452 (1984). These techniques involve splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody of appropriate biological activity.
  • Such chimeric antibodies have, e.g., a variable region derived from a murine antibody and a constant region derived from a human antibody.
  • single chain antibodies specific for JIP-1 polypeptides can be produced using known techniques. See, e.g., U.S. Pat. Nos. 4,946,778 and 4,704,692. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Antibody fragments that recognize and bind to specific epitopes of JIP-1 can also be generated by known techniques. Such fragments include, but are not limited to, F(ab′) 2 fragments, produced by pepsin digestion of antibody molecules, and Fab fragments, generated by reduction of the disulfide bonds of F(ab′) 2 fragments. Alternatively, Fab expression libraries can be constructed to allow rapid and easy identification of monoclonal Fab fragments with a desired specificity. See, e.g., Huse et al., Science 246:1275 (1989).
  • Antibodies to JIP-1 can be used to generate anti-idiotypic antibodies that resemble a portion of JIP-1, using techniques well known to those skilled in the art. See, e.g., Greenspan et al., FASEB J. 7:437 (1993); Nisonoff, J. Immunol. 147:2429 (1991).
  • antibodies that bind to JIP-1 and competitively inhibit binding of other ligands can be used to generate anti-idiotypic antibodies resembling a ligand binding domain of JIP-1.
  • These anti-idiotypic antibodies can bind and neutralize JIP-1 ligands.
  • JIP-1 ligands include proline-rich regions of proteins, since JIP-1 has an SH3 domain that binds to these regions. There are ten isoforms of JNK. Gupta et al., EMBO J. 15:2760-2770 (1996). Each of these JNK isoforms is a JIP-1 ligand. Other kinases, which may be related to JNK, may also interact with JIP-1. Neutralizing anti-idiotypic antibodies or Fab fragments of anti-idiotypic antibodies can be used in therapeutic regimens.
  • therapies can be designed to reduce the level of endogenous JIP-1 gene expression, e.g., using antisense or ribozyme approaches to inhibit or prevent translation of JIP-1 mRNA transcripts; triple helix approaches to inhibit transcription of the JIP-1 gene; or targeted homologous recombination to inactivate or “knock out” the JIP-1 gene or its endogenous promoter. Delivery techniques can be designed to allow theraputic compositions to cross the blood-brain barrier (see, e.g., PCT WO89/10134).
  • Antisense approaches involve the design of oligonucleotides (either DNA or RNA) that are complementary to JIP-1 mRNA and inhibit expression of JIP-1 protein. Absolute complementarity of the antisense oligonucleotide to JIP-1, although preferred, is not required.
  • a sequence “complementary” to a portion of an RNA, as referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may be tested, or triplex formation may be assayed.
  • the ability to hybridize will depend on both the degree of complementarily and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex.
  • Oligonucleotides that are complementary to the 5′ end of the message should work most efficiently at inhibiting translation.
  • sequences complementary to the 3′ untranslated sequences of mRNAs have been shown to be effective at inhibiting translation of mRNAs as well (Wagner, Nature, 372:333, 1994).
  • oligonucleotides complementary to either the 5′- or 3′-non-translated, non-coding regions of the JIP-1 gene can be used in an antisense approach to inhibit translation of endogenous JIP-1 mRNA.
  • Oligonucleotides complementary to the 5′ region overlapping the initiation codon can be used for this purpose.
  • Antisense oligonucleotides that are complementary to the coding region can also be used.
  • target regions are generally those lacking predicted secondary structure.
  • Antisense oligonucleotides complementary to mRNA coding regions can also be used in accordance with the invention. Whether designed to hybridize to the 5′, 3′, or coding region of JIP-1 mRNA, antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In specific aspects the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50 nucleotides in length.
  • in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to inhibit gene expression. These studies preferably utilize controls that distinguish between antisense inhibition and nonspecific biological effects of oligonucleotides. It is also preferred that these studies compare levels of the target RNA or protein with that of an internal control RNA or protein. Additionally, it is envisioned that results obtained using the antisense oligonucleotide are compared with those obtained using a control oligonucleotide.
  • control oligonucleotide is of approximately the same length as the test oligonucleotide and that the nucleotide sequence of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence.
  • the oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. Stability of the oligonucleotide can be improved by modification of the phosphodiester moieties by replacement with phosphorothioate, —CH2—, or —NH— groups. The oligonucleotides can also be stabilized by blocking the 5′ and 3′ termini with phosphorothioate, —CH2— or —NH— groups. Each of these modifications will lead to increased stability (e.g. by increasing modifications will lead to increased stability (e.g. by increasing resistance to nucleases) and therefore increased hybridization.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (as described, e.g., in Letsinger et al., Proc. Natl. Acad. Sci. USA, 86:6553 (1989); Lemaitre et al., Proc. Natl. Acad. Sci. USA, 84:648 (1987)); PCT Publication No. WO 88/09810) or the blood-brain barrier (see, e.g., PCT Publication No.
  • oligonucleotide can be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent.
  • the antisense oligonucleotide can include at least one modified base moiety which is selected from the group including, but not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethyl-aminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-is
  • the antisense oligonucleotide can also include at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2-fluoroarabinose, xylulose, and hexose.
  • the antisense oligonucleotide includes at least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal, or an analog of any of these backbones.
  • the antisense oligonucleotide is an á-anomeric oligonucleotide.
  • An á-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual á-units, the strands run parallel to each other (Gautier et al., Nucl. Acids. Res., 15:6625 (1987)).
  • the oligonucleotide is a 2′-0-methylribonucleotide (Inoue et al., Nucl. Acids Res., 15:6131 (1987)), or a chimeric RNA-DNA analog (Inoue et al., FEBS Lett., 215:327 (1987)).
  • Antisense oligonucleotides of the invention can be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (commercially available from Biosearch, Applied Biosystems, etc.).
  • an automated DNA synthesizer commercially available from Biosearch, Applied Biosystems, etc.
  • phosphorothioate oligonucleotides can be synthesized by the method of Stein et al. ( Nucl. Acids Res., 16:3209 (1988)), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., Proc. Natl. Acad. Sci. USA, 85:7448 (1988)).
  • the antisense molecules should be delivered to cells that express JIP-1 in vivo.
  • a number of methods have been developed for delivering antisense DNA or RNA to cells.
  • antisense molecules can be injected directly into specific tissue sites.
  • Modified antisense molecules, designed to target the desired cells e.g., antisense linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface
  • One approach to achieving intracellular concentrations of the antisense molecule sufficient to suppress translation of endogenous mRNAs is to use a recombinant DNA construct in which the antisense oligonucleotide is placed under the control of a strong pol III or pol II promoter.
  • the use of such a construct to transfect target cells in a patient will result in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the eldogenous JIP-1 transcripts, and thereby prevent translation of the JIP-1 mRNA.
  • a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • vectors can be constructed by recombinant DNA technology methods standard in the art.
  • Vectors can be plasmid, viral, or others known in the art, used,for replication and expression in mammalian cells.
  • Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells. Such promoters can be inducible or constitutive.
  • Such promoters include, but are not limited to: the SV40 early promoter region (Bernoist et al., Nature, 290:304 (1981)); the promoter contained in the 3′ long terminal repeat of Rous sarcoma virus (Yamamoto et al., Cell, 22:787-797 (1988)); the herpes thymidine kinase promoter (Wagner et al., Proc. Natl. Acad. Sci USA, 78:1441, 1981); or the regulatory sequences of the metallothionein gene (Brinster et al., Nature, 296:39 (1988)).
  • Any type of plasmid, cosmid, YAC, or viral vector can be used to prepare the recominant DNA construct which can be introduced directly into specific tissue sites.
  • viral vectors can be used that selectively infect the desired tissue (e.g., for brain, herpesvirus vectors may be used), in which case administration can be accomplished by another route (e.g., systemically).
  • Ribozyme molecules designed to catalytically cleave JIP-1 mRNA transcripts also can be used to prevent translation of JIP-1 mRNA and expression of JIP-1 protein (see, e.g., PCT Publication WO 90/11364; Saraver et al., Science, 247:122 (1990)). While various ribozymes that cleave mRNA at site-specific recognition sequences can be used to destroy JIP-1 mRNAs, the use of hammerhead ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA.
  • target mRNA contain the following sequence of two bases: 5′-UG-3′.
  • the construction and production of hammerhead ribozymes is well known in the art (Hasseloff et al., Nature, 334;585, 1988).
  • the ribozymes of the present invention also include RNA endoribonucleases (hereinafter “Cech-type ribozymes”), such as the one that occurs naturally in Tetrahymena Thermophila (known as the IVS or L-19 IVS RNA), which have been extensively characterized by Cech and his collaborators.
  • Cech-type ribozymes such as the one that occurs naturally in Tetrahymena Thermophila (known as the IVS or L-19 IVS RNA), which have been extensively characterized by Cech and his collaborators. See, e.g., Zaug et al., Science, 224:574 (1984); Zaug et al., Science, 231:470 (1986); Zaug et al., Nature, 324:429 (1986); PCT Application No. WO 88/04300; and Been et al., Cell, 47:207 (1986).
  • the Cech-type ribozymes have an eight base-pair sequence that hybridizes to a target RNA sequence, whereafter cleavage of the target RNA takes place.
  • the invention encompasses those Cech-type ribozymes that target eight base-pair active site sequences present in JIP-1.
  • target sites for ribozymes are regions that are predicted to lack appreciable secondary structure.
  • Target sites for ribozymes include:
  • the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.), and should be delivered to cells which express JIP-1 in vivo.
  • a preferred method of delivery involves using a DNA construct “encoding” the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the rihozyme to destroy endogenous JIP-1 messages and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency.
  • Destruction of JIP-1 mRNA would be advantageous in increasing apoptosis in tumors; in altering the cell damage occurring during ischemic reperfusion in cardiovascular disease, kidney disease, and stroke; and altering cell damage occurring in the liver in response to metabolic oxidative stress.
  • Endogenous JIP-1 gene expression can also be reduced by inactivating or “knocking out” the JIP-1 gene or its promoter using targeted homologous recomination (see, e.g., U.S. Pat. No. 5,464,764).
  • a mutant, non-functional JIP-1 (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous JIP-1 gene can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express JIP-1 in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the JIP-1 gene.
  • Such approaches are particularly suited for use in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive JIP-1.
  • this approach can be adapted for use in humans, provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors, e.g., herpes virus vectors for delivery to brain tissue.
  • endogenous JIP-1 gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to regulatory regions of the JIP-1 gene (e.g., a JIP-1 promoter and/or enhancer to form triple helical structures that prevent transcription of the JIP-1gene in target cells in the body.
  • deoxyribonucleotide sequences complementary to regulatory regions of the JIP-1 gene e.g., a JIP-1 promoter and/or enhancer to form triple helical structures that prevent transcription of the JIP-1gene in target cells in the body.
  • the invention also features polypeptides that interact with JIP-1.
  • Any method suitable for detecting protein-protein interactions can be employed for identifying intracellular or extracellular proteins that interact with JIP-1.
  • traditional methods that can be employed are co-immunoprecipitation, crosslinking, and co-purification through gradients or chromatographic columns of cell lysates, or proteins obtained from cell lysates. Purified proteins are used to identify proteins in the lysate that interact with JIP-1.
  • the JIP-1 polypeptide can be full length JIP-1, a soluble domain of JIP-1, or some other suitable JIP-1 polypeptide.
  • portions of their amino acid sequences can be ascertained using techniques well known to those of skill in the art, such as the Edman degradation technique.
  • the amino acid sequences obtained can be used to design degenerate oligonucleotide probes that can be used to screen for gene sequences encoding the interacting protein. Screening may be accomplished, for example, by standard hybridization or PCR techniques. Techniques for the generation of degenerate oligonucleotide mixtures and screening are well-known. See Ausubel, supra; and Innis et al., eds., PCR Protocols: A Guide to Methods and Applications , Academic Press, Inc., New York (1990).
  • methods can be used that result directly in the identification of genes that encode proteins that interact with JIP-1. These methods include, for example, screening expression libraries in a manner similar to the well known technique of antibody probing of ⁇ gt11 libraries using labeled JIP-1 polypeptides or a JIP-1 fusion protein, e.g., an JIP-1 polypeptide or domain fused to a marker such as an enzyme, a fluorescent dye, a luminescent protein, or to an IgFc domain.
  • a marker such as an enzyme, a fluorescent dye, a luminescent protein, or to an IgFc domain.
  • Protein interactions can also be identified in vivo using the two-hybrid system. Chien et al., Proc. Natl. Acad. Sci. USA, 88:9578 (1991). A kit for practicing this method is available from Clontech (Palo Alto, Calif.). In this system, two plasmids, each encoding a hybrid protein, are constructed. One plasmid, the “bait” plasmid, includes a nucleotide sequence encoding the DNA-binding domain of a transcription activator protein fused to a nucleotide sequence encoding a protein of interest.
  • the other plasmid includes a nucleotide sequence encoding the transcription activator protein's activation domain and an unknown protein, which may bind the protein of interest.
  • the plasmids are transformed into a strain of Saccharomyces cerevisiae that contains a reporter gene (e.g., HIS or lacZ) whose regulatory region contains the transcription activator's binding site.
  • a reporter gene e.g., HIS or lacZ
  • Neither of the hybrid proteins can activate transcription of the reporter gene alone; the bait plasmid lacks an activation function, and the other plasmid cannot localize to the transcription activator's binding sites. If the protein of interest and the unknown protein form a protein-protein complex and reconstitute the proximity of the DNA binding domain and the activation domain, this complex can bind to the regulatory region of the reporter gene, and the reporter gene will be expressed.
  • the two-hybrid system or related methodology can be used to screen libraries for proteins that interact with the “bait” gene product.
  • JIP-1 can be used as the bait gene product.
  • Total genomic or cDNA sequences are fused to the DNA encoding an activation domain of a transcriptional activator protein.
  • This library and a plasmid encoding a hybrid of bait JIP-1 gene product fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene.
  • a bait JIP-1 gene sequence such as JIP-1 or a domain of JIP-1
  • a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein, to form a bait plasmid.
  • Colonies are purified and the library plasmids responsible for reporter gene expression are isolated.
  • the library plasmids are subjected to DNA sequencing to determine the sequences of the gene encoding the JIP-1 binding proteins.
  • a cDNA library can be made by methods that are routine in the art.
  • This library can be inserted into vectors so that the sequences from the library are fused to nucleotide sequences encoding the transcriptional activation domain of the GAL4 protein.
  • These vectors can then be co-transformed, along with a bait JIP-1 gene-GAL4 fusion plasmid, into a yeast strain which contains a lacZ gene driven by a promoter that contains the GAL4 activation sequence.
  • a functional GAL4 will be formed, and the HIS3 gene will be expressed. Colonies expressing HIS3 can then be purified, and used to produce and isolate the JIP-1 interacting protein using techniques routinely practiced in the art.
  • compositions of the JNK inhibitor JIP-1 can be used to treat pathological conditions associated with apoptosis or transformation. These compositions can include JIP-1 polypeptides or peptides that specifically bind to and sequester JNK. Proteins can be purified by methods known to those skilled in the art. Ausubel, supra. Peptides can be synthesized by methods that are known to those skilled in the art. See, e.g., Merrifield, J. Am. Chem. Soc., 85:2149-2154 (1963).
  • Peptide mimetics are also included in the therapeutic compositions of the invention. These compounds mimic the activity of the peptide or polypeptide, but are composed of molecules other than, or in addition to, amino acids.
  • the design of such mimetics is described in, e.g., Hughes, Philos. Trans. R. Soc. Lond., 290:387-394 (1980); Hodgson, Biotechnol., 9:19-21 (1991); Suckling, Sci. Prog., 75:323-359 (1991).
  • compositions of the invention also include nucleic acids encoding a JIP-1 polypeptide or peptide. These nucleic acids can be administered in a manner allowing their uptake and expression by cells in vivo. Compositions containing nucleic acids can be prepared for administration by methods that are routine in the art.
  • compositions of the invention can include one or more compounds, e.g., nucleic acids, peptides, polypeptides, or peptide mimetics and a pharmaceutically acceptable carrier.
  • Pharmaceutically acceptable carriers are biologically compatible vehicles, e.g., physiological saline, which are suitable for administration to a patient.
  • Nucleic acids can be administered to the patient by standard vectors and/or gene delivery systems.
  • Suitable gene delivery systems include liposomes, receptor-mediated delivery systems, naked DNA and viral vectors such as herpes viruses, retroviruses, adenoviruses and adeno-associated viruses.
  • Peptides can be coupled to membrane permeable peptides in order to facilitate their uptake by cells. This can be done by colinear synthesis of a membrane permeable peptide with a peptide sequence of interest. The two peptides could also be crosslinked together. Methods of coupling are described in, e.g., Lin et al., J. Biol. Chem., 269:12320-12324 (1996); Rojas et al., J. Biol. Chem., 271:27456-127461 (1997). Peptides can also be coupled to lipids to provide membrane permeability. See, e.g., Vijayaraghavan, J. Biol. Chem., 272:4747-4752 (1997). As an example, a fatty acid can be coupled to the ⁇ -NH 2 group of the peptide as an amide.
  • compositions can be delivered in encapsulated cell implants (e.g., those produced by CytoTherapeutics, Inc., Buffalo R.I.; see Bioworld Today 7:6 (Monday, Dec. 2, 1996)). Delivery of drugs to the brain can also be accomplished using RMP-7TM technology (Alkermes, Inc., Cambridge, Mass.; see Business Wire , “Third Major Agreement for Prolease Sustained Release Drug Delivery System,” Dec. 2, 1996) or implantable wafers containing the drug (see PR Newswire , “Implantable Wafer is First Treatment to Deliver Chemotherapy Directly to Tumor Site,” Sep. 24, 1996).
  • RMP-7TM technology Alkermes, Inc., Cambridge, Mass.; see Business Wire , “Third Major Agreement for Prolease Sustained Release Drug Delivery System,” Dec. 2, 1996)
  • implantable wafers containing the drug see PR Newswire , “Implantable Wafer is First Treatment to Deliver Chemotherapy Directly to Tumor Site,” Sep. 24, 1996).
  • compositions can also be administered using an implantable pump for direct administration into intrathecal fluid (e.g., that made by Medtronic, Minneapolis, Minn.; see Genetic Engineering News , “Neurobiotechnology Companies Focus Programs on Pain and Neuroprotection,” Nov. 1, 1996).
  • an implantable pump for direct administration into intrathecal fluid (e.g., that made by Medtronic, Minneapolis, Minn.; see Genetic Engineering News , “Neurobiotechnology Companies Focus Programs on Pain and Neuroprotection,” Nov. 1, 1996).
  • Parenteral administration such as intravenous, subcutaneous, intramuscular, or intraperitoneal delivery routes can be used to deliver the therapeutic compositions of the invention. Dosages for particular patients depend upon many factors, including the patient's size, body surface area, age, the particular substance to be administered, time and route of administration, general health and other drugs being administered concurrently.
  • the amount of therapeutic composition to be administered to a patient can be in the range of 1 to 1000 ⁇ g/kg of body weight, e.g., 10 to 500 ⁇ g/kg, or 20 to 200 ⁇ g/kg of body weight.
  • a typical dose of peptide or nucleic acid to be administered to a patient is 100 ⁇ g per kilogram of body weight.
  • the yeast two hybrid method was used to screen a mouse embryo cDNA library to identify proteins that interact with JNK. The method is described in detail in Fields et al., Nature, 340:245 (1989).
  • the yeast strain used was L40 (MATa his ⁇ 200 trp1-901 leu2-3, 112 ade2 LYS:: (lexAop) 4 -HIS URA3:: (lexAop) 8 -lacZ.
  • the bait plasmid, pLexA-JNK1 was constructed by blunt-end ligation of a XbaI/HindIII fragment of pCMV-Flag-JNK1 (Derijard et al., Cell, 76:1025 (1994); Sluss et al., Mol. Cell Biol., 14:8376 (1994); Gupta et al., EMBO. J., 15:2760 (1996)) into the vector pBTM116 at the SmaI and SalI sites.
  • Yeast transformants (5.2 ⁇ 10 6 ) were examined for growth on media with 25 mM 3-aminotriazole in the absence of histidine. Positive clones were confirmed by measurement of lacZ expression.
  • the two-hybrid screen yielded a cDNA fragment encoding a portion of a JNK binding protein.
  • Full-length cDNA clones were obtained by screening a mouse brain ⁇ ZAPII cDNA library (Stratagene Inc.) with a random-primed cDNA fragment corresponding to base pairs 560-1020 of the full length cDNA. Isolated cDNA clones were sequenced using an Applied Biosystems model 373A machine. The sequence of the 5′ GC-rich non-coding region of the largest cDNA clone was confirmed using the Maxam-Gilbert method.
  • JIP-1 JNK interacting protein-1
  • JNK binding domain JBD; residues 127-281, SEQ ID NO:4
  • putative SH3 domain residues 491-540, SEQ ID NO:13
  • the putative SH3 domain of JIP-1 is highly related to the SH3 domains located in the tyrosine kinase c-fyn and the p85 subunit of PI-3′ kinase.
  • Human homologs of the murine JIP-1 gene can be isolated using the murine JIP-1 cDNA clones or fragments of those clones as probes, by methods that are routine in the art of molecular biology. For example, a library of human cDNA can be screened with a fragment of murine JIP-1 cDNA, and human cDNA hybridizing to the murine JIP-1 cDNA can be isolated, cloned, sequenced and analyzed for structural and functional similarity to murine JIP-1. For general methods, of isolating and characterizing homologous genes from libraries, see Sambrook et al., Molecular Cloning, A Laboratory Manual , Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2d ed. 1989).
  • JIP-1 is expressed in many different tissues, including brain, heart, spleen, lung, liver, muscle, kidney and testis. Highest amounts of JIP-1 mRNA were detected in brain, kidney, and testis.
  • JIP-1 Specifically Binds JNK in vivo
  • DNA encoding the Flag epitope was inserted into JIP-1 cDNA between the DNA encoding the first two codons of JIP-1 using insertional overlapping polymerase chain reaction (PCR).
  • PCR insertional overlapping polymerase chain reaction
  • Bacterial JIP-1 expression vectors were constructed by subcloning PCR fragments of the JIP-1 cDNA into the EcoRI and XbaI sites of pGEX-3 ⁇ (Pharmacia LKB Biotechnology Inc.). GST fusion proteins were purified by affinity chromatography on GSH-agarose as described previously. Smith et al., Gene, 67:31 (1988). Sequences of the constructs were confirmed using an Applied Biosystems model 373A machine.
  • transfected or mock-transfected cells were irradiated with or without UV-C (40 J/m 2 ) and incubated for one hour. Lysates prepared from the cells were examined by protein immunoblot analysis using a mixture of antibodies specific for Flag and HA to detect Flag-JIP-1 and HA-JNK1, respectively.
  • TLB Tris (pH 7.5), 1% Triton X-100, 10% glycerol, 0.137 M NaCl, 25 mM sodium ⁇ -glycerophosphate, 1 mm sodium orthovanadate, 2 mM sodium pyrophosphate, 2 mM EDTA, 10 ⁇ g/ml leupeptin, 1 mM phenylmethylsulfonylfluoride). Soluble extracts were prepared by centrifugation at 100,000 ⁇ g for 30 minutes at 4° C.
  • the extracts were pre-cleared using protein G-Sepharose (Pharmacia-LKB Biotechnologies Inc.) and incubated for one hour with monoclonal antibody to the Flag epitope (M2; IBI-Kodak) or the HA epitope (12CA5; Boehringer-Mannheim) pre-bound to protein G-Sepharose.
  • the immunoprecipitates were washed three times with TLB and once with 25 mM Hepes (pH 7.5), 0.2% (w/v) Triton X-100, 1 mM EDTA. Proteins were fractionated by SDS-PAGE and transferred electrophoretically to Immobilon-P membranes (Millipore).
  • the membranes were blocked with 10% gamma globulin-free horse serum (Gibco-BRL) and probed with the M2 monoclonal antibody, to detect Flag-JIP-1, and either the 12CA5 monoclonal antibody or a sheep anti-JNK polyclonal antibody, to detect HA-JNK1. Immune complexes were detected with a second antibody coupled to horseradish peroxidase and enhanced chemiluminescence (Amersham International PLC)
  • JIP-1 was detected in JNK1 immunoprecipitates by protein immunoblot analysis, and JNK1 was detected in JIP-1 immunoprecipitates. Co-immunoprecipitation of JIP-1 with JNK2, was also observed. These data indicate that JIP-1 specifically binds JNK in vivo. Exposure to UV radiation caused no significant change in the amount of the JNK/JIP-1 complex detected by co-immunoprecipitation analysis. Control experiments performed to examine the specificity of the interaction of JIP-1 with JNK demonstrated that the related MAP kinases ERK2 and p38 did not co-immunoprecipitate with JIP-1. The absence of co-immunoprecipitation of JIP-1 with these MAP kinases demonstrates that JIP-1 forms a specific complex with JNK.
  • This same assay can be used to determine whether a specific polypeptide is a JIP-1 polypeptide.
  • JIP-1 Interacts Directly with JNK
  • JIP-1 interacts directly with JNK
  • JBD putative JNK binding domain
  • GST-fusion proteins were purified by affinity chromatography on GSH-agarose as described previously. Smith et al., Gene, 67:31 (1988).
  • JNK Recombinant JNK, prepared by in vitro translation in the presence of [ 35 S]methionine (Dérijard et al., supra; Sluss et al., supra; Gupta et al., supra), was incubated with GST-JIP-1 immobilized on GSH-agarose. Control experiments revealed no detectable binding of JNK isoforms to GST alone. In contrast, in assays using GST-JNK fusion proteins representing ten different human JNK isoforms, each of these proteins exhibited similar amounts of binding to JIP-1. JIP-1 therefore interacts with multiple JNK isoforms.
  • JNK binds to the transcription factors c-Jun and ATF2, and that the binding of JNK to these transcription factors is isoform-dependent.
  • a binding assay was utilized. Cell lysates (1 ml in TLE) containing JNK1, JNK2, or p38 MAP kinase, each tagged with the Flag epitope, were incubated with 5 ⁇ g GST-fusion proteins pre-bound to 10 ⁇ l glutathione-Sepharose.
  • the GST fusion proteins used contain residues 127-202 (SEQ ID NO:15), 203-281 (SEQ ID NO:16), 164-240 (SEQ ID NO:17), or 127-281 (SEQ ID NO:4) of JIP-1.
  • the GST-ATF2 fusion protein contains residues 1-109 of ATF2, and the GST-Jun fusion protein contains residues 1-79 of c-Jun. After incubation for one hour at 4° C. and three washes with TLB, bound proteins were detected by protein immunoblot analysis with the M2 monoclonal antibody, which is specific for the Flag epitope.
  • the GST-JIP-1 fusion proteins containing various portions of the JIP-1 gene were used to define regions of JIP-1 that are required for JNK interaction.
  • a GST-JIP-1 fusion protein containing residues 127-281 of JIP-1 bound both JNK1 and JNK2.
  • No JNK binding was detected in experiments using the central region (residues 164-240) or the carboxy terminal region (residues 203-281) of JIP-1.
  • JNK binding activity was observed in experiments using the amino terminal region (residues 127-202 of JIP-1).
  • JIP-1 sequence required for JNK binding activity additional GST-JNK fusion proteins; were constructed. These fusion proteins contained residues 135-202 (SEQ ID NO:18), 144-202 (SEQ ID NO:19), 154-202 (SEQ ID NO:20), 164-202 (SEQ ID NO:21), 127-143 (SEQ ID NO:22), 127-153 (SEQ ID NO:23), or 127-163 (SEQ ID NO:24, of JIP-1. Proteins containing residues 127-202, 135-202, 144-202, 154-202, and 127-163 all bound both JNK1 and JNK2. Thus, JIP-1 residues 144-163 are important for the interaction of JIP-1 with JNK. As shown in FIG. 2, this region of JIP-1 shares sequence similarity with the JNK binding domains of ATF2 and c-Jun.
  • This assay can also be used to analyze a given polypeptide to determine whether it is, or is not, a JIP-1 polypeptide.
  • the JNK binding domain of JIP-1 contains three amino eacids, Lys-155, Thr-159, and Leu-160, that are conserved in the JNK binding domains of ATF2 and c-Jun (FIG. 2).
  • a hydrophobic amino acid, Leu is found at residue 162 of JIP-1.
  • ATF2 and c-Jun also contain hydrophobic amino acids (ATF2, Phe; c-Jun, Leu) in the position corresponding to residue 162 of JIP-1.
  • the wild type JIP-1 JBD was substituted with glycines in these positions to produce the peptides shown in FIG.
  • the mutant peptides are identical to the wild type peptide except for the indicated glycine substitutions.
  • a peptide with a scrambled sequence (SEQ ID NO:12) was used as a control.
  • the peptide representing the wild-type JIP-1 sequence caused a dose-dependent inhibition of JIP-1 binding to JNK.
  • the control peptide caused no change in JNK binding.
  • JIP-1 is a Selective Inhibitor of JNK Activity
  • JIP-1 To test whether JIP-1 competes with Jun and ATF2 for interaction with JNK, the effect of JIP-1 on transcription factor-mediated phosphorylation of exogenous substrates was analyzed using an in vitro protein kinase assay.
  • Chinese hamster ovary (CHO) cells were serum-starved for one hour.
  • the cells were treated with 10 ng/ml mouse interleukin 1 or 100 nM phorbol myristic acetate.
  • JNK, p38, and ERK protein kinase activity was measured in an immune complex kinase assay using 3 ⁇ g of the substrates GST-Jun, GST-ATF2, and myelin basic protein (MBP), respectively.
  • Protein kinase assays were performed using 40 ⁇ l 20 mM Hepes (pH 7.4), 20 mM MgCl 2, 20 mM ⁇ -glycerophosphate, 2 mM dithiothreitol, 0.1 mM sodium orthovanadate, 50 ⁇ M [ ⁇ - 32 P]ATP (10 Ci/mmol). After incubation for 30 minutes at 30° C., phosphorylation of substrates was analyzed by polyacrylamide gel electrophoresis and autoradiography.
  • JIP-1 markedly inhibited the phosphorylation of c-Jun by JNK.
  • JIP-1 caused no significant change in the phosphorylation of substrates by the related MAP kinases p38 and ERK2. JIP-1 is thus a selective inhibitor of JNK.
  • This same assay can be used to determine whether a specific polypeptide has the same phosphorylation function of wildtype JIP-1.
  • JIP-1 The effect of JIP-1 on targets of the JNK signal ransduction pathway, including the transcription factors c-Jun, ATF2 and Elk-1, was examined to determine whether JIP-1 inhibits signal transduction by JNK.
  • CHO cells were cotransfected with constructs encoding the transcription factors and JIP-1 using previously described methods.
  • a luciferase reporter plasmid was used to monitor the expression of the transcription factors. Transfection efficiency was determined using a ⁇ -galactosidase expression vector.
  • Constructs used in these experiments included GAL4 fusions with the c-myc, Sp1 and VP16 activation domains (described in Whitmarsh et al., Science, 269:403 (1995); Davis, Science, 269:403 (1995); Gille et al., Curr.
  • GAL4-c-Jun in which GAL4 is fused to c-Jun
  • GAL4-c-Jun S63A/S73A
  • GAL4-ATF2 wild type ATF2
  • GAL4-ATF2 T69A/T71A
  • ATF2(Ala-69/71) in which GAL4 has been fused to a mutant ATF2 in which the tyrosines at positions 69 and 71 have been changed to alanines
  • GAL4-Elk-1 in which GAL4 has been fused to Elk-1
  • GAL4-Elk-1 S383A
  • This assay can be used to determine whether specific polypeptides have the same effect on signal transduction as full length, wildtype JIP-1.
  • JIP-1 The subcellular distribution of JIP-1 was analyzed by performing indirect immunofluorescence on JIP-1-expressing cells. In these experiments, the cells were grown on coverslips, fixed with 4% paraformaldehyde, permeabilized with 0.25% Triton X-100, and processed for immunofluorescence microscopy. JIP-1 was detected in the cytoplasm, but not the nucleus, of control and UV-irradiated cells. In contrast, JNK is detected in both the cytoplasmic and nuclear compartments.
  • JNK JNK in the nucleus which is involved in the regulation of gene expression. Since JIP-1 is cytoplasmic, it was unclear how it could inhibit the nuclear function of JNK. To investigate the mechanism of JIP-1 action, the distribution of JNK was examined in cells that had been transfected with either hemagglutinin (HA)-tagged JNK1 (HA-JNK1) alone, or HA-JNK1 and Flag-tagged JIP-1 (Flag-JIP-1). In these experiments, the cells were exposed to a potent JNK activator (40 J/m 2 UV-C) for one hour prior to processing for immunofluorescence.
  • HA hemagglutinin
  • Flag-JIP-1 Flag-tagged JIP-1
  • the primary antibodies used were rabbit anti-HA (12CA5; Boehringer Mannheim), which recognizes HA-tagged JNK1; and mouse monoclonal anti-Flag (M2; IBI-Kodak), which recognizes Flag-tagged JIP-1.
  • the secondary antibodies were Texas Red-goat anti-mouse Ig and fluoroscein isothiocyanate-conjugated donkey anti-rabbit Ig (Jackson Immunoresearch). Procedures for digital imaging microscopy and image restoration using the exhaustive photon reassignment algorithm are described in Carrington et al., Science, 268:1483 (1995). Individual optical sections were inspected using computer graphics software on a Silicon Graphics workstation.
  • JIP-1 reduces the amount of nuclear JNK detected in control and UV-irradiated cells.
  • JIP-1 expression has no significant effect on the subcellular distribution of p38 MAP kinase, which is also located in both nuclear and cytoplasmic compartments of cultured cells.
  • JIP-1 Inhibits Nerve Growth Factor Withdrawal-Induced Apoptosis
  • JIP-1 Inhibits Pre-B Cell Transformation by BCR-ABL
  • the BCR-ABL oncogene which is associated with chronic myelogenous leukemia (CML), causes JNK activation in the absence of increased ERK activity. Raitano et al., Proc. Natl. Acad. Sci. USA, 92:11746 (1995). Oncogenic transformation by BCR-ABL may be mediated in part by the JNK signaling pathway. Since JIP-1 can inhibit JNK activity, the effect of JIP-1 expression on oncogenic transformation was examined. Plasmid vectors expressing v-ABL or BCR-ABL and Flag-tagged JBD (residues 127-281) of JIP-1 were used to transfect 293 cells.
  • CML chronic myelogenous leukemia
  • JNK activity was then measured in an immune complex kinase assay of lysates of the transfected 293 cells, using a polyclonal JNK antibody and GST-Jun as the substrate.
  • Control experiments demonstrated that v-ABL and BCR-ABL caused constitutive JNK activation (approximately 5-fold), which was blocked by co-expression of the JBD of JIP-1. JIP-1 can therefore inhibit BCR-ABL-mediated activation of JNK.
  • FIG. 6 shows the mean density ( ⁇ SE) of non-adherent pre-B cells on day 10 post infection. The data shown are derived from three independent experiments plated in triplicate. As expected, BCR-ABL caused pre-B cell transformation. The JBD of JIP-1 caused marked inhibition of transformation when present in the sense, but not the anti-sense, orientation.
  • JNK JNK signal transduction pathway
  • JIP-1 an inhibitor of the JNK signal transduction pathway
  • JIP-1 The physiological function of JIP-1 may be to suppress signal transduction by the JNK pathway.
  • JIP-1 may compete with substrates that bind JNK.
  • JIP-1 may have a more direct role in targeting JNK to specific regions of the cell or to specific substrates. Since JIP-1 causes redistribution of JNK within the cell, JIP-1 may function as a cytoplasmic anchor for JNK. The tethering of JNK in the cytoplasm by interactions with JIP-1 provides a mechanism for controlling signal transduction by the JNK pathway, and the related phenomena of apoptosis and transformation.
  • This assay can be used to determine if specific polypeptides have the same effect on cellular transformation as full length, wildtype JIP-1.
  • Peptides suspected of having JIP-1 activity can be tested in the JNK binding assay described supra.
  • Peptides are synthesized by methods that are well known to those skilled in the art; for example, using an Applied Biosystems synthesizer.
  • Cell lysates containing Flag epitope-tagged JNK are incubated with GST-JIP-1 (residues 127-281 bound to glutathione-Sepharose, either with or without synthetic peptides (0, 4, 8, 16, 32 or 64 ⁇ g/ml), for one hour at 4° C.
  • the beads are washed in TLB, and the bound proteins are detected by protein immunoblot analysis with a Flag-specific antibody, e.g., M2.
  • Synthetic peptides with JIP-1 activity are those which inhibit the interaction of JIP-1 with JNK, as detected in this assay. These synthetic peptides should possess at least 60% of the binding activity of JIP-1.
  • JIP-1 is shown herein to be capable of inhibiting apoptosis and transformation.
  • Compositions containing JIP-1 polypeptides or nucleic acids can be administered to treat conditions characterized by these phenomena.
  • Nucleic acids can be administered by, methods described in, e.g., Ausubel, et al., supra.
  • a standard dosage would be from 1 to 1000 ⁇ g/kg of body weight.
  • Polypeptides, peptides and peptide mimetics of the invention can be formulated according to procedures which are well known to those skilled in the art.
  • a standard dosage of polypeptide also would be from 1 to 1000, e.g., 10 to 500, or 20 to 200 ⁇ g/kg of body weight.
  • Animal models for testing the effect of JIP-1 therapeutic compositions include the bcr-abl leukemia model. Daley et al., Science, 247:824-830 (1990). Other animal models that can be used include the myc/ras transformation model. Sinn et al., Cell, 49:465-475 (1987). Animal models for testing the effect of JIP-1 therapeutic compositions on conditions associated with apoptosis include a model of excitotoxic stress in the hippocampus, and a model of E2F-1 induced apoptosis. Ben-Ari et al., Neuroscience, 14:375-403 (1985); Field et al., Cell, 85:549-561 (1996).
  • JIP-1 compositions include liver damage (Mendelson et al., Proc. Natl. Acad. Sci. USA, 93:12908-12913 (1996)); kidney disease and organ transplantation (DeMari et al., Am. J. Physiol., 272:F292-F298 (1997)); and heart disease (Pombo et al., J. Biol. Chem., 269:26546-26551 (1994); Force et al., Circ. Res., 78:947-953 (1996).
  • the polypeptides and antibodies of the invention can be used to detect or monitor JIP-1 expression.
  • Levels of JIP-1 protein in a sample can be assayed by any standard technique.
  • JIP-1 protein expression can be monitored by standard immunological or immunohistochemical procedures using the antibodies described herein. See, e.g., Ausubel et al., supra; Bancroft et al., Theory and Practice of Histological Techniques , Churchill Livingstone (1982).
  • JIP-1 expression can be assayed by standard Northern blot analysis, or can be aided by PCR. See Ausubel, supra; Ehrlich, ed., PCR Technology: Principles and Applications for DNA Amplification , Stockton Press, New York.
  • Point mutations in the JIP-1 sequence can be detected using well known nucleic acid mismatch detection techniques. Lower than normal levels of JIP-1 would have the effect of altering apoptosis, while higher than normal levels would cause immune suppression, altered inflammatory responses, and alterations in tumor growth.
  • the invention also includes naturally occurring and non-naturally-occurring allelic variants of JIP-1.
  • the nucleic acid sequence encoding allelic variants may have a substitution, deletion, or addiction of one or more nucleotides.
  • the preferred allelic variants are functionally equivalent to naturally occurring JIP-1.

Abstract

JNK-interacting protein 1 (JIP-1), an inhibitor of the JNK1 protein, and methods of treating a pathological condition or of preventing the occurrence of a pathological condition in a patient by the administration of a therapeutically effective amount of JIP-1 polypeptides, peptides, peptide mimetics, or nucleic acids are described.

Description

    STATEMENT AS TO FEDERALLY SPONSORED RESEARCH
  • [0001] This invention was made, in part, with Government support under grants CA58396 and CA65831 awarded by the National Cancer Institute. The government may have certain rights in the invention.
  • FIELD OF THE INVENTION
  • The invention relates to signal transduction inhibitors. [0002]
  • BACKGROUND OF THE INVENTION
  • The c-Jun NH2-terminal kinase (JNK) is a member of the stress-activated group of Mitogen Activated Protein kinases (MAP kinases) implicated in the control of cell growth. The JNK signal transduction pathway is activated in response to environmental stress and by the engagement of several classes of cell surface receptors, including cytokine receptors, serpentine receptors, and receptor tyrosine kinases. Whitmarsh et al., [0003] J. Mol. Med., 74:589 (1996). In addition, genetic studies of Drosophila have demonstrated that JNK is required for early embryonic development. Sluss et al., Genes & Dev., 10:2745 (1996); Riesgo-Escovar et al., Genes & Dev. 10:2759 (1996). In mammalian cells, JNK has been implicated in the immune response, oncogenic transformation, and apoptosis. JNK mediates these effects, at least in part, by increasing the expression of target genes. Targets of the JNK signal transduction pathway include the transcription factors c-Jun, ATF2, and Elk-1. Whitmarsh et al., supra.
  • JNK is activated in the liver by metabolic oxidative stress. Mendelson et al., [0004] Proc. Natl. Acad. Sci. USA 93:12908-12913 (1996). Activation of JNK also occurs in the kidney during stress, for example, during Bchemic renal failure. Demari et al., Am. J. Physiol. 272:F292-F298 (1997). JNK is also activated during cardiovascular disease such as ischemia Ireperfusion and during organ transplantation. Pombo et al., J. Biol. Chem. 269:26546-26551 (1994); Force et al., Circ. Res. 78:947-53 (1996).
  • While JNK is located in both the cytoplasmic and the nuclear compartments of quiescent cells, activation of the JNK signal transduction pathway is associated with accumulation of JNK in the nucleus. Mechanisms governing this sub-cellular distribution have not been previously elucidated. [0005]
  • Anchor or tethering proteins play an important role in the regulation of multiple signal transduction pathways. These anchor proteins, which include the nuclear factor kappa B (NFkB) inhibitor IkB, the A kinase anchor protein (AKAP) group of proteins that bind the type II cyclic adenosine monophosphate (AMP) dependent protein kinase, and the p190 protein that binds Ca[0006] 2+-calmodulin-dependent protein kinase II, localize their tethered partners to specific sub-cellular compartments. Verma et al., Genes & Dev., 9:2723 (1995); McNeill et al., J. Biol. Chem., 270:10043 (1995); Faux et al., Trends Biochem. Sci., 21:312 (1996)). Anchor proteins also target enzymes to specific substrates, and create multi-enzyme signaling complexes, such as the Ste5 MAP kinase scaffold complex and the AKAP79 kinase/phosphatase scaffold complex. Choi et al., Cell, 78:499 (1994); Klauck et al., Science, 271:1589 (1996); Faux et al., Cell, 85:9 (1996)).
  • SUMMARY OF THE INVENTION
  • The invention, which is based on the discovery of a cytoplasmic anchor protein, JNK-interacting protein 1 (JIP-1; SEQ ID NO:1), features JIP-1 polypeptides and nucleic acids, therapeutic compositions containing these polypeptides and nucleic acids, and methods of administering these compositions. JIP-1 specifically binds to and inhibits the biological effects of JNK, including the initiation of apoptosis and oncogenic transformation. JIP-1 is therefore useful as a therapeutic agent for treating pathological conditions characterized by apoptosis or transformation. For example, JIP-1 compositions can be used to treat neurodegenerative diseases characterizediby apoptosis, including Parkinson's disease and Alzheimer's disease; and blood clots, which left untreated could result in stroke and associated memory loss. Other conditions that can be treated using the compositions and methods of the invention are autoimmune diseases such as arthritis; other conditions characterized by inflammation; and malignancies, such as leukemias, e.g., chronic myelogenous leukemia (CML). Other conditions that can be treated with JIP-1 compositions include oxidative damage to organs such as the liver and kidney, and heart disease, particularly damage due to ischemia/reperfusion and cardiomyopathy. JIP-1 compositions can also be used to treat donor organs for transplantation. These organs are exposed to substantial environmental stress, the effects of which are blocked by JNK inhibitors such as JIP-1. [0007]
  • The invention features a substantially pure JIP-1 polypeptide. A “JIP-1 polypeptide” is a protein having an amino acid sequence that specifically binds JNK to the same extent, or at least 10% of the binding activity of wildtype JIP-1. Such polypeptides can be from 5 to 200 amino acids in length, e.g., from 10 to 100 amino acids in length, or from 20 to 50 amino acids in length. Such polypeptides include the JNK Binding Domain (JBD), or portions thereof, of JIP-1 (e.g., amino acids 148 to 174, forming the “core” of the JBD of wildtype JIP-1, shown in FIG. 2, and having the sequence SGDTYRPKRPTTLNLFPQVPRSQDTLN; SEQ ID NO:3). JIP-1 polypeptides are preferably derived from a mammal, such as a mouse or a human. [0008]
  • In various embodiments, the polypeptide is soluble, the polypeptide includes the JNK-binding domain of JIP-1 or a portion thereof, the polypeptide is at least 80%, 90%, or 100% identical to the amino acid sequence from amino acid 148 to amino acid 174 of JIP-1 (the core JNK-binding domain; SEQ ID NO:3), or the polypeptide has an amino acid sequence identical to the amino acid sequence from amino acid 148 to amino acid 174 of JIP-1 (SEQ ID NO:3), or the polypeptide is at least 80%, 90%, or 100% identical to the amino acid sequence from [0009] amino acid 127 to amino acid 281 of JIP-1 (the JNK-binding domain; SEQ ID NO:4).
  • The polypeptides of the invention can be modified to enhance their uptake by cells. Such modifications increase the hydrophobicity of molecules to facilitate passage through the lipid bilayer of the cell membrane. For example, polypeptides can be complexed with myristic acid or packaged in liposomes. Alternatively, JIP-1 polypeptides can be complexed with hydrophobic moieties (e.g., lipids) or peptides that increase the delivery of proteins into cells. [0010]
  • The invention also includes peptide mimetics of JIP-1 polypeptides. A “peptide mimetic” of a known polypeptide is a compound that mimics the activity of the peptide or polypeptide, but which is composed of molecules other than, or in addition to, amino acids. [0011]
  • By “polypeptide” is meant any chain of amino acids, regardless of length or post-translational modification (e.g., glycosylation or phosphorylation), and thus includes peptides, proteins, and fusion proteins. [0012]
  • A “substantially identical” polypeptide sequence differs from a given sequence only by conservative amino acid substitutions or by one or more nonconservative substitutions, deletions, or insertions located at positions which do not destroy the function of the polypeptide compared to wildtype JIP-1. Polypeptides of the invention can be 70%, 80%, 85%, 90%, or 95% identical to wildtype JIP-1. [0013]
  • A “substantially pure” preparation is at least 60% by weight of the compound of interest, e.g., a JIP-1 polypeptide or fragment of a JIP-1 polypeptide. Preferably the preparation is at least 75%, more preferably at least 90%, and more preferably at least 95% by weight of the compound of interest. Purity can be measured by any appropriate standard method, e.g., column chromatography, polyacrylamide gel electrophoresis, or High Pressure Liquid Chromatography (HPLC) analysis. [0014]
  • The polypeptides of the invention include, but are not limited to, recombinant polypeptides, natural polypeptides, and synthetic polypeptides, as well as preproteins or proproteins and biologically active fragments. A “biologically active fragment” of JIP-1 is a fragment having at least 50%, 70%, 80%, 90%, 95%, or 100% or greater, of the activity of naturally occurring or synthetic, full length JIP-1. [0015]
  • The polypeptides of the invention can be physically linked to another polypeptide, e.g., a marker polypeptide. For example, the polypeptide can be fused to a hexa-histidine tag to facilitate purification of bacterially expressed proteins, or a hemagglutinin tag to facilitate purification of protein expressed in eukaryotic cells. [0016]
  • In another aspect, the invention features an isolated nucleic acid that includes a sequence encoding a JIP-1 polypeptide or a fragment of such a polypeptide. Preferably, the nucleic acid is derived from a mammal. [0017]
  • The invention also encompasses nucleic acids that hybridize under stringent conditions (as described herein) to a nucleic acid encoding a JIP-1 polypeptide. Stringent conditions include hybridization at 68° C. in 5×SSC/5×Denhardt's solution/1.0% SDS, or in 0.5 M NaHPO[0018] 4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2×SSC/0.1% SDS at room temperature or at 42° C., or in 0.1×SSC/0.1% SDS at 68° C., or in 40 mM NaHPO4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C. Moderately stringent conditions include washing in 3×SSC at 42° C. The parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid. For guidance regarding such conditions see, e.g., Sambrook et al., Molecular Cloninc, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y.; and Ausubel et al., supra, at Unit 2.10.
  • The hybridizing portion of the hybridizing nucleic acid is preferably 20, 30, 50, or 70 bases long. The hybridizing portion of the hybridizing nucleic acid can be 95% or even 98% or 100% identical to the sequence of a portion of a nucleic acid encoding a JIP-1 polypeptide. Hybridizing nucleic acids of the type described above can be used as a cloning probe, a primer (e.g., a PCR primer), or a diagnostic probe. Preferred hybridizing nucleic acids encode a polypeptide having some or all of the biological activities possessed by naturally-occurring JIP-1. Thus, they may encode a protein that is shorter or longer than the various forms of JIP-1 described herein. Hybridizing nucleic acids can also encode proteins that are related to JIP-1, e.g., proteins encoded by genes that include a portion having a relatively high degree of identity to a JIP-1 gene described herein. [0019]
  • The term “nucleic acid” encompasses both RNA and DNA, including cDNA, genomic DNA, and synthetic (e.g., chemically synthesized) DNA. The nucleic acid may be double-stranded or single-stranded. Where single-stranded, the nucleic acid may be the sense strand or the antisense strand. [0020]
  • An “isolated nucleic acid” is a nucleic acid that is free of the nucleic acids that normally flank it in the genome. The term therefore includes, e.g., a recombinant nucleic acid incorporated into a vector, such as an autonomously replicating plasmid or virus; a cDNA or genomic DNA fragment produced by polymerase chain reaction (PCR) or restriction endonuclease treatment; and recombinant DNA which is part of a hybrid gene encoding additional polypeptide sequences. [0021]
  • A “substantially identical” nucleic acid is a nucleic acid with a sequence that is at least 50%, preferably 70%, and more preferably 85%, 90%, or 95% homologous to a given nucleic acid sequence, e.g., SEQ ID NO:2. [0022]
  • The invention also features transformed cells harboring a nucleic acid encompassed by the invention. Vectors and plasmids that include a nucleic acid properly positioned for expression are also within the invention. A “transformed cell” is a cell into which (or into an ancestor of which) has been introduced, by means of recombinant DNA techniques, a DNA molecule encoding a JIP-1 polypeptide. [0023]
  • “Operably linked” means that the selected DNA molecule is positioned adjacent to one or more sequence elements that direct transcription and/or translation of the sequence such that the sequence elements can control transcription and/or translation of the selected DNA (i.e., the selected DNA is operably associated with the sequence elements). Such operably associated elements can be used to facilitate the production of a JIP-1 polypeptide. [0024]
  • The invention also features purified antibodies which specifically bind a JIP-1 protein or polypeptide. A “purified antibody” is an antibody which is at least 60%, by dry weight, free from the proteins and naturally-occurring organic molecules with which it is naturally associated. The preparation can be at least 75%, at least 90%, and up to 99% or more, by dry weight, antibody. [0025]
  • An antibody that “specifically binds” an antigen recognizes and binds to that antigen, e.g., a JIP-1 polypeptide. [0026]
  • Also within the invention are antisense molecules and ribozymes for inhibiting JIP-1 expression. [0027]
  • The invention also features antagonists and agonists of JIP-1. Antagonists can inhibit one or more of the functions of JIP-1. Suitable antagonists can include large or small molecules, antibodies to JIP-1, and JIP-1 polypeptides that compete with a native form of JIP-1. Such antagonists include SEQ ID NO:3, a component of an active site of JIP-1, i.e., the JNK-binding domain. Agonists of JIP-1 will enhance or facilitate one or more of the functions of JIP-1. Agonists and Antagonists include polyproline motifs, which bind to SH3 domains such as that found in JIP-1. [0028]
  • A “therapeutically effective amount” of a substance is an amount capable of producing a medically desirable result in a treated patient, e.g., inhibition of the expression or activity of a specific protein. [0029]
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the arts of protein chemistry or molecular biology. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described infra. All publications, patent applications, patents and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be limiting. [0030]
  • Other features and advantages of the invention will be apparent from the detailed description and the claims.[0031]
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A is a schematic diagram showing the structure of a murine JIP-1 polypeptide. [0032]
  • FIG. 1B is a representation of the amino acid sequence of a murine JIP-1 polypeptide (SEQ ID NO:1), presented in single letter code. [0033]
  • FIG. 1C is a representation of the nucleotide sequence of a murine JIP-1 cDNA (SEQ ID NO:2). [0034]
  • FIG. 2 is a diagram showing an alignment of the JBDs (JNK-binding domains) of JIP-1 (SEQ ID NO:3), c-Jun (SEQ ID NO:5), and ATF2 (SEQ ID NO:6). [0035]
  • FIG. 3 is a diagram showing the amino acid sequences of wild type and mutant JIP-1 peptides (SEQ ID NOs:3 and 7 to 11), as well as a control peptide (SEQ ID NO:12). [0036]
  • FIG. 4A is a bar graph showing the effect of recombinant JBD (JIP-1 residues 127-281) on reporter gene expression mediated by the GAL4 binding domain and GAL4 fusions with the c-myc, Sp1, and VP16 activation domains. [0037]
  • FIG. 4B is a bar graph showing the effect of recombinant JBD (JIP-1 residues 127-281) on reporter gene expression mediated by wild type and mutant forms of GAL4-c-Jun, GAL4-ATF2, and GAL4-Elk-1. [0038]
  • FIG. 4C is a bar graph showing the effect of wild type and mutant JIP-1 and JED on reporter gene expression mediated by wild type and mutant GAL4-ATF2. [0039]
  • FIG. 5 is a bar graph showing the effect of the JBD of JIP-1 on Nerve Growth Factor (NGF) withdrawal-induced apoptosis. [0040]
  • FIG. 6 is a bar graph showing the effect of bicistronic retroviruses on primary mouse bone marrow cells.[0041]
  • DETAILED DESCRIPTION
  • The invention is based on the molecular cloning and characterization of JIP-1, a cytoplasmic protein that specifically binds JNK. JIP-1 polypeptides cause cytoplasmic retention of JNK and inhibition of JNK-regulated gene expression. In addition, JIP-1 polypeptides suppress the effects of the JNK signaling pathway, including oncogenic transformation and apoptosis. These findings have important implications for the treatment or prevention of pathological conditions and diseases, many of which are characterized by transformation or apoptosis. Conditions associated with apoptosis include neurodegenerative conditions, such as Parkinson's disease or Alzheimer's disease; and blood clots, which left untreated could result in stroke and associated memory loss. Other conditions that can be treated using the compositions and methods of the invention are autoimmune diseases such as arthritis; other conditions characterized by inflammation; and malignancies, such as leukemias, e.g., chronic myelogenous leukemia (CML). JIP-1 polypeptide compositions can also be used to treat oxidative damage to organs such as the liver and kidney. Heart disease can also be treated with the compositions of the invention. Donor organs for transplantation can also be treated with JIP-1 compositions. [0042]
  • JIP-1 Proteins and Polypeptides [0043]
  • As shown in FIG. 1, the 660 amino acid JIP-1 protein has an SH3 domain at its carboxy terminal end, at amino acid positions 491-540, and a JNK-binding domain (JBD) at its amino terminal end, at amino acid positions 127-281 (SEQ ID NO:4). The core of the JBD is amino acids 148-174 (SEQ ID NO:3). The JED of JIP-1 shares conserved residues with the JNK-binding regions of the transcription factors c-Jun and ATF2, as shown in FIG. 2. [0044]
  • JIP-1 polypeptides can be prepared for a wide range of uses including, but not limited to, generation of antibodies, preparation of reagents for diagnostic assays, identification of other molecules involved in transformation or apoptosis, preparation of reagents for use in screening assays for modulators of apoptosis or transformation, and preparation of therapeutic agents for treatment of disorders related to apoptosis or transformation. [0045]
  • The invention encompasses, but is not limited to, JIP-1 polypeptides that are functionally related to JIP-1 encoded by the nucleotide sequence of FIG. 1C (SEQ ID NO:2) Functionally related polypeptides include any polypeptide sharing a functional characteristic with wildtype JIP-1 protein, e.g., the ability to bind JNK polypeptides or to affect proliferation or apoptosis. Such functionally related JIP-1 polypeptides include, but are not limited to, polypeptides having additions or substitutions of amino acid residues within the amino acid sequence encoded by the JIP-1 sequences described herein which result in a silent change, thus producing a functionally equivalent gene product. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. [0046]
  • For example, nonpolar (hydrophobic) amino acids include alanine, leucine, isoleucine, valine, proline, phenylalanine, tryptophan, and methionine; polar neutral amino acids include glycine, serine, threonine, cysteine, tyrosine, asparagine, and glutamine; positively charged (basic) amino acids include arginine, lysine, and histidine; and negatively charged (acidic) amino acids include aspartic acid and glutamic acid. [0047]
  • While random mutations can be made to JIP-1 DNA (using random mutagenesis techniques well known to those skilled in the art) and the resulting mutant JIP-1 proteins can be tested for activity, site-directed mutations of the JIP-1 coding sequence can be engineered (using site-directed mutagenesis techniques well known to those skilled in the art) to generate mutant JIP-1 proteins with increased function, e.g., greater JNK-1 binding and inhibition of transformation. [0048]
  • To design functionally related and functionally variant JIP-1 polypeptides, it is useful to distinguish between conserved positions and variable positions. To preserve JIP-1 function, it is preferable that conserved residues are not altered. Moreover, alteration of non-conserved residues are preferably conservative alterations, e.g., a basic amino acid is replaced by a different basic amino acid. To produce altered function variants, it is preferable to make non-conservative changes at variable and/or conserved positions. Deletions at conserved and variable positions can also be used to create altered function variants. Conserved amino acids in JIP-1 include, but are not limited to, Lys-155, Thr-159, Leu-160, Asn-161, and Leu-162. [0049]
  • Preferred JIP-1 polypeptides are those that bind JNK and inhibit transformation or apoptosis. These JIP-1 polypeptides have 20%, 40%, 50%, 75%, 80%, 90%, or even greater than 100% of the activity of the full-length JIP-1 described herein. Such comparisons are generally based on equal concentrations of the molecules being compared. The comparison can also be based on the amount of protein or polypeptide required to reach 50% of the maximal stimulation obtainable. [0050]
  • Polypeptides corresponding to one or more domains of JIP-1, e.g., the JNK Binding Domain (JBD), are also within the scope of the invention. Preferred polypeptides are those which are soluble under normal physiological conditions. Also within the invention are fusion proteins in which a portion (e.g., one or more domains) of JIP-1 is fused to an unrelated protein or polypeptide (i.e., a fusion partner) to create a fusion protein. The fusion partner can be a moiety selected to facilitate purification, detection, or solubilization, or to provide some other function. Fusion proteins are generally produced by expressing a hybrid gene in which a nucleotide sequence encoding all or a portion of JIP-1 is joined in-frame to a nucleotide sequence encoding the fusion partner. Fusion partners include, but are not limited to, the constant region of an immunoglobulin (IgFc). A fusion protein in which a JIP-1 polypeptide is fused to IgFc can be more stable and have a longer half-life in the body than the JIP-1 polypeptide on its own. [0051]
  • Also within the scope of the invention are various soluble forms of JIP-1, including JIP-1 expressed on its own or fused to a solubilization partner, e.g., an immunoglobulin. [0052]
  • In general, JIP-1 polypeptides can be produced by transformation (transfection, transduction, or infection) of a host cell with all or part of a JIP-1-encoding DNA fragment (e.g., the cDNA described herein) in a suitable expression vehicle. Suitable expression vehicles include: plasmids, viral particles, and phage. For insect cells, baculovirus expression vectors are suitable. The entire expression vehicle, or a part thereof, can be integrated into the host cell genome. In some circumstances, it is desirable to employ an inducible expression vector, e.g., the LACSWITCH™ Inducible Expression System (Stratagene; LaJolla, Calif.). [0053]
  • Any of a wide variety of expression systems can be used to provide the recombinant proteins. The precise host cell used is not critical to the invention. The JIP-1 protein can be produced in a prokaryotic host (e.g., [0054] E. coli or B. subtilis) or in a eukaryotic host, e.g., yeast, such as Saccharomyces or Pichia; mammalian cells, such as COS, NIH 3T3, CHO, BHK, 293, or HeLa cells; or insect cells.
  • JIP-1 polypeptides can also be produced by plant cells. Viral expression vectors (e.g., cauliflower mosaic virus and tobacco mosaic virus) and plasmid expression vectors (e.g., Ti plasmid) are suitable for use in plant cells. Plant cells are available from a wide range of sources, e.g., the American Type Culture Collection, Rockland, Md. See also, e.g., Ausubel et al., [0055] Current Protocols in Molecular Biology, John Wiley & Sons, New York, 1995. The methods of transformation or transfection and the choice of expression vehicle will depend on the host system selected. Transformation and transfection methods are described in, e.g., Ausubel et al., supra; expression vehicles may be chosen from those described in, e.g., Cloning Vectors: A Laboratory Manual, P. H. Pouwels et al., 1985, Supp. 1987.
  • The host cells harboring the expression vehicle can be cultured in conventional nutrient media adapted as needed for activation of a chosen gene, repression of a chosen gene, selection of transformants, or amplification of a chosen gene. [0056]
  • A suitable expression system is the mouse 3T3 fibroblast host cell transfected with a pMAMneo expression vector (Clontech, Palo Alto, Calif.). pMAMneo provides an RSV-LTR enhancer linked to a dexamethasone-inducible MMTV-LTR promotor, an SV40 origin of replication which allows replication in mammalian systems, a selectable neomycin gene, and SV40 splicing and polyadenylation sites. DNA encoding a JIP-1 in protein would be inserted into the pMAMneo vector in an orientation designed to allow expression. The recombinant JIP-1 protein would be isolated as described below. Other preferable host cells that can be used in conjunction with the pMAMneo expression vehicle include COS cells and CHO cells (ATCC Accession Nos. CRL 1650 and CCL 61, respectively). [0057]
  • JIP-1 polypeptides can be expressed as fusion proteins. For example, the expression vector pUR278 can be used to create lacZ fusion proteins. See Ruther et al., [0058] EMBO J. 2:1791 (1983). The pGEX vectors can be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can be easily purified from cell lysates by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • In an insect cell expression system, [0059] Autographa californica nuclear polyhidrosis virus (AcNPV), which grows in Spodoptera frugiperda cells, is used as a vector to express foreign genes. A JIP-1 coding sequence can be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of a AcNPV promoter, e.g., the polyhedrin promoter. Successful insertion of a gene encoding a JIP-1 polypeptide or protein will result in inactivation of the polyhedrin gene and production of non-occluded recombinant virus (i.e.,, virus lacking the proteinaceous coat encoded by the polyhedrin gene). Spodoptera frugiperda cells are then infected with these viruses, and the inserted gene is expressed. See, e.g., Smith et al., J. Virol. 46:584 (1983); Smith, U.S. Pat. No. 4,215,051.
  • In mammalian host cells, a number of viral-based expression systems can be utilized. In cases where an adenovirus is used as an expression vector, the JIP-1 nucleic acid sequence can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence, to form a chimeric gene. This chimeric gene can then be inserted into the adenovirus genome by in vitro or in vivo recombination. Insertion into a non-essential region of the viral genome (e.g., the E1 or E3 gene) will result in a recombinant virus that is viable and capable of expressing a JIP-1 gene product in infected hosts. See, e.g., Logan, [0060] Proc. Natl. Acad. Sci. USA, 81:3655 (1984).
  • Specific initiation signals may be required for efficient translation of inserted nucleic acid sequences. These signals include the ATG initiation codon and adjacent sequences. In cases where an entire native JIP-1 gene or cDNA, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. In other cases, exogenous translational control signals, including, perhaps, the ATG initiation codon, must be provided. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. Exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements. Bittner et al., [0061] Methods in Enzymol., 153:516 (1987).
  • In addition, a host cell may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in a desired fashion. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product can be used. Such mammalian host cells include, but are not limited to, CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, and WI38 cells. [0062]
  • Alternatively, a JIP-1 protein can be produced by a stably-transfected mammalian cell line. A number of vectors suitable for stable transfection of mammalian cells are available to the public. See, e.g., Pouwels et al., supra. Methods for constructing such cell lines are also publicly available. See, e.g., Ausubel et al., supra. JIP-1 cDNA can be cloned into an expression vector that includes the dihydrofolate reductase (DHFR) gene. Methotrexate (0.01-300 μM) is present in the culture medium to select for cells which have integrated the plasmid and, therefore, the JIP-1 cDNA. See Ausubel et al., supra. This dominant selection can be accomplished in most cell types. [0063]
  • Recombinant protein expression can be increased by DHFR-mediated amplification of the transfected gene. Methods for selecting cell lines bearing gene amplifications are described in Ausubel et al., supra. Such methods generally involve extended culture in medium containing gradually increasing levels of methotrexate. DHFR-containing expression vectors commonly used for this purpose include pCVSEII-DHFR and pAdD26SV(A). See Ausubel et al., supra. Any of the host cells described above or, preferably, a DHFR-deficient CHO cell line (e.g., [0064] CHO DHFR 0 cells, ATCC Accession No. CRL 9096) are among the host cells preferred for DHFR selection of a stably-transfected cell line or DHFR-mediated gene amplification.
  • A number of other selection systems can be used, including but not limited to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyl-transferase, and adenine phosphoribosyltransferase genes can be employed in tk, hgprt, or aprt cells, respectively. In addition, gpt, which confers resistance to mycophenolic acid (Mulligan et al., [0065] Proc. Natl. Acad. Sci. USA, 78:2072 (1981); neo, which confers resistance to the aminoglycoside G-418 (Colberre-Garapin et al., J. Mol. Biol., 150:1 (1981)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene, 30:147 (1981)), can be used.
  • Alternatively, any fusion protein can be readily purified by utilizing an antibody specific for the fusion protein being expressed. For example, a system described in Janknecht et al., [0066] Proc. Natl. Acad. Sci. USA, 88:8972 (1981), allows for the ready purification of non-denatured fusion proteins expressed in human cell lines. In this system, the gene of interest is subcloned into a vaccinia recombination plasmid such that the gene's open reading frame is translationally fused to an amino-terminal tag consisting of six histidine residues. Extracts from cells infected with recombinant vaccinia virus are loaded onto Ni2+ nitriloacetic acid-agarose columns, and histidine-tagged proteins are selectively eluted with imidazole-containing buffers.
  • Alternatively, JIP-1 or a portion thereof can be fused to an immunoglobulin Fc domain. Such a fusion protein can be readily purified using a protein A column. Moreover, such fusion proteins permit the production of a dimeric form of a JIP-1 polypeptide having increased stability in vivo. [0067]
  • After the recombinant JIP-1 protein is expressed, it is isolated. Secreted forms can be isolated from the culture media, while non-secreted forms must be isolated from the host cells. Proteins can be isolated by affinity chromatography. An anti-JIP-1 antibody (e.g., produced as described herein) is attached to a column and used to isolate the JIP-1 protein. Lysis and fractionation of JIP-1 protein-harboring cells prior to affinity chromatography can be performed by standard methods. See, e.g., Ausubel et al., supra. Alternatively, a JIP-1 fusion protein, for example, a JIP-1-maltose binding protein, a JIP-1-β-galactosidase, or a JIP-1-trpE fusion protein, can be constructed and used for JIP-1 protein isolation. See, e.g., Ausubel et al., supra; New England Biolabs, Beverly, Mass. [0068]
  • Once isolated, the recombinant protein can, if desired, be further purified, e.g., by high performance liquid chromatography using standard techniques. See, e.g., Fisher, [0069] Laboratory Techniques In Biochemistry And Molecular Biology, Work et al., eds., Elsevier (1980).
  • Polypeptides of the invention, particularly short JIP-1 fragments, can also be produced by chemical synthesis, e.g., by the methods described in [0070] Solid Phase Peptide Synthesis, 2nd ed., The Pierce Chemical Co., Rockford, Ill., 1984.
  • The invention also features proteins which interact with JIP-1 and which are involved in the function of JIP-1. Also included in the invention are the genes encoding these interacting proteins. Interacting proteins can be identified using methods known to those skilled in the art. One method suitable method is the “two-hybrid system” which detects protein interactions in vivo. See, e.g., Chien et al., [0071] Proc. Natl. Acad. Sci. USA, 88:9578 (1991). A kit for practicing this method is available from Clontech (Palo Alto, Calif.).
  • JIP-1 Nucleic Acids [0072]
  • The JIP-1 cDNA sequences described herein, and related family members of the JIP-1 gene present in mouse, human, or other species can be identified and readily isolated without undue experimentation by well known molecular biological techniques given the specific sequences described herein. Further, genes may exist at other loci that encode proteins having extensive homology to JIP-1 polypeptides or one or more domains of JIP-1 polypeptides. These genes can be identified by known techniques using the sequences disclosed herein. For example, hybridization of JIP-1 probes to homologous nucleic acids is performed under stringent conditions. Alternatively, a labeled fragment can be used to screen a genomic library derived from the organism of interest, again, using appropriately stringent conditions. Such stringent conditions are well known, and will vary predictably depending on the specific organisms from which the library and the labeled sequences are derived. [0073]
  • Nucleic acid duplex or hybrid stability is expressed as the melting temperature, or T[0074] m, which is the temperature at which a probe dissociates from a target DNA. This melting temperature is used to define the required stringency conditions. If sequences are to be identified that are related and substantially identical to the probe, rather than identical, then it is useful to first establish the lowest temperature at which only homologous hybridization occurs with a particular concentration of SSC or SSPE. It is then assumed that 1% mismatching results in a 1° C. decrease in the Tm, and the temperature of the final wash is reduced accordingly (for example, if sequences with ≧95% identity with the probe are sought, the final wash temperature is decreased by 5° C.). Note that this assumption is very approximate, and the actual change in Tm can be between 0.50° and 1.5° C. per 1% mismatch.
  • As used herein, stringent conditions include hybridization at 68° C. in 5×SSC/5×Denhardt's solution/1.0% SDS, or in 0.5 M NaHPO[0075] 4 (pH 7.2)/1 mM EDTA/7% SDS, or in 50% formamide/0.25 M NaHPO4 (pH 7.2)/0.25 M NaCl/1 mM EDTA/7% SDS; and washing in 0.2×SSC/0.1% SDS at room temperature or at 42° C., or in 0.1×SSC/0.1% SDS at 68° C., or in 40 mM NaHPO4 (pH 7.2)/1 mM EDTA/5% SDS at 50° C., or in 40 mM NaHPO4 (pH 7.2) 1 mM EDTA/1% SDS at 50° C. Moderately stringent conditions include washing in 3×SSC at 42° C. The parameters of salt concentration and temperature can be varied to achieve the desired level of identity between the probe and the target nucleic acid. For guidance regarding such conditions see, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y.; and Ausubel et al., supra, at Unit 2.10.
  • Upon detection of JIP-1 transcript in human cell lines by Northern blot analysis, cDNA libraries can be constructed from RNA isolated from these cell lines utilizing standard techniques. The human cDNA library can then be screened with a JIP-1 probe to isolate a human JIP-1 cDNA. [0076]
  • Alternatively, a human genomic DNA library can be screened using JIP-1 probes. Hybridizing clones can be sequenced and the intron and exon structure of the human JIP-1 gene can be elucidated. Once a genomic sequence is obtained, oligonucleotide primers can be designed based on the sequence for use in Reverse Transcriptase-coupled PCR, e.g., to isolate human JIP-1 cDNA. An example of a suitable probe for screening a human genomic library is the coding region (nucleotides 180 to 2159) of the mouse JIP-1 cDNA (SEQ ID NO:2). [0077]
  • Further, a previously unknown gene sequence can be isolated by performing PCR using two degenerate oligonucleotide primer pools designed on the basis of nucleotide sequences within the JIP-1 cDNA defined herein. Degenerate PCR primers that can be used include: 5′[0078] GARGARTTYGARGAYGARGA 3′ (sense; SEQ ID NO:25); 5′GGNAARAARCAYAGNTGGCA 3′ (sense; SEQ ID NO:26); 5′CATRTTWTANGCYTCWTACCA 3′ (antisense; SEQ ID NO:27); and 5′AAYTGYTTKTARAAYTGYTGRAA 3′ (antisense; SEQ ID NO:28), where W is A or T, K is G or T, R is A or G, Y is C or T, and N is A, C, G or T. The template for the reaction can be cDNA obtained by reverse transcription of mRNA prepared from human or non-human cell lines or tissue known to express, or suspected of expressing, JIP-1. The PCR product can be subcloned and sequenced to insure that the amplified sequences represent the sequences of JIP-1 or JIP-1-like gene nucleic acid sequences.
  • The PCR fragment can then be used to isolate a full length cDNA clone by a variety of methods. For example, the amplified fragment can be labeled and used to screen a cDNA library in bacteriophage. Alternatively, the labeled fragment can be used to screen a genomic library. [0079]
  • PCR technology also can be used to isolate full length cDNA sequences. For example, RNA can be isolated, following standard procedures, from an appropriate tissue or cell line. A reverse transcription reaction can be performed on the RNA using an oligonucleotide primer specific for the most 5′ end of the amplified fragment for the priming of first strand synthesis. The resulting RNA/DNA hybrid can then be “tailed” with guanines using a standard terminal transferase reaction. After digestion with RNAase H, and second strand synthesis can be primed with a poly-C primer. Thus, cDNA sequences upstream of the amplified fragment can easily be isolated. For a review of useful cloning strategies, see e.g., Sambrook et al., supra; Ausubel et al., supra. [0080]
  • Mutant cDNAs can also be isolated using PCR techniques. The first cDNA strand can be synthesized by hybridizing an oligo-dT oligonucleotide to mRNA isolated from tissue known to be or suspected of being expressed in an individual putatively carrying the mutant allele, and by extending the new strand with reverse transcriptase. The second strand of the cDNA can then be synthesized using an oligonucleotide that hybridizes specifically to the 5′-end of the normal gene. Using these two oligonucleotides as primers, the product is amplified via PCR, cloned into a suitable vector, and subjected to DNA sequence analysis by methods well known in the art. By comparing the DNA sequence of the mutant gene to that of the normal gene, the mutation(s) responsible for the loss or alteration of function of the mutant gene product can be ascertained. [0081]
  • Alternatively, a genomic or cDNA library can be constructed and screened using DNA or RNA, respectively, from a tissue known to express, or suspected of expressing, the gene of interest in an individual suspected of carrying or known to carry the mutant allele. The normal gene or any suitable fragment thereof can then be labeled and used as a probe to identify the corresponding mutant allele in the library. The clone containing the mutant gene can then be purified through methods routinely practiced in the art, and subjected to sequence analysis using standard techniques as described herein. [0082]
  • Additionally, an expression library can be constructed using DNA isolated from or cDNA synthesized from a tissue known to express or suspected of expressing the gene of interest in an individual suspected of carrying or known to carry the mutant allele. In this manner, gene products made by this tissue can be expressed and screened using standard antibody screening techniques in conjunction with antibodies raised against the normal gene product, as described herein. For screening techniques, see, for example, Harlow et al., eds., [0083] Antibodies: A Laboratory Manual, Cold Spring Harbor Press, Cold Spring Harbor, N.Y. (1988).
  • In cases where the mutation results in an expressed gene product with altered function (e.g., as a result of a missense mutation), a polyclonal set of antibodies is likely to cross-react with the mutant gene product. Library clones, detected via their reaction with such labeled antibodies can be purified and subjected to sequence analysis as described herein. [0084]
  • JIP-1 Peptide Mimetics [0085]
  • JIP-1 peptide mimetics can be constructed by structure-based drug design through replacement of amino acids by organic moieties. See, e.g., Hughes, [0086] Philos. Trans. R. Soc. Lond., 290:387-394 (1980); Hodgson, Biotechnol., 9:19-21 (1991); Suckling, Sci. Prog., 75:323-359 (1991). The use of peptide mimetics can be enhanced through the use of combinatorial chemistry to create drug libraries. The design of peptide mimetics can be aided by identifying amino acid mutations that increase or decrease JIP-1 binding to JNK. Approaches that can be used include the yeast two hybrid method (see, e.g., Chien et al., Proc. Natl. Acad. Sci. USA, 88:9578 (1991); kit from Clontech, Palo Alto, Calif.), and the phage display method. The two hybrid method detects protein-protein interactions in yeast. Fields et al., Nature, 340:245-246 (1989). The phage display method detects the interaction between an immobilized protein and a protein that is expressed on the surface of phages (e.g. lambda and M13). Amberg, et al., Strategies, 6:2-4 (1993); Hogrefe et al., Gene, 128:119-126 (1993). These methods allow positive and negative selection for protein-protein interactions and the identification of the sequences that determine these interations.
  • Transgenic Animals [0087]
  • JIP-1 polypeptides can also be expressed in transgenic animals. Animals of any species, including, but not limited to, mice, rats, rabbits, guinea pigs, pigs, micro-pigs, goats, and non-human primates, e.g., baboons, monkeys, and chimpanzees, can be used to generate JIP-1-expressing transgenic animals. [0088]
  • Various techniques known in the art can be used to introduce a JIP-1 transgene into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (U.S. Pat. No. 4,873,191); retrovirus mediated gene transfer into germ lines (van der Putten et al., [0089] Proc. Natl. Acad. Sci. USA, 82:6148 (1985)); gene targeting into embryonic stem cells (Thompson et al., Cell, 56:313 (1989)); and electroporation of embryos (Lo, Mol. Cell. Biol, 3:1803 (1983)).
  • The present invention provides for transgenic animals that carry the JIP-1 transgene in all their cells, as well as animals that carry the transgene in some, but not all of their cells, i.e., mosaic animals. The transgene can be integrated as a single transgene or in concatamers, e.g., head-to-head tandems or head-to-tail tandems. The transgene can also be selectively introduced into and activated in a particular cell type. Lasko et al., [0090] Proc. Natl. Acad. Sci. USA, 89:6232 (1992). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • When it is desired that the JIP-1 transgene be integrated into the chromosomal site of the endocenous JIP-1 gene, gene targeting is preferred. Vectors containing some nucleotide sequences homologous to an endogenous JIP-1 gene are designed for the purpose of integrating via homologous recombination into the endogenous gene and disrupting its function. The transgene also can be selectively introduced into a particular cell type, thus inactivating the endogenous JIP-1 gene in only that cell type. See Gu et al., [0091] Science, 265:103 (1984). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art.
  • Once transgenic animals have been generated, the expression of the recombinant JIP-1 gene can be assayed utilizing standard techniques. Initial screening can be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to assay whether integration of the transgene has taken place. The level of mRNA expression of the transgene in the tissues of the transgenic animals can also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and RT-PCR. Samples of tissues expressing JIP-1 can also be evaluated immunocytochemically using antibodies specific for the JIP-1 transgene product. [0092]
  • Anti-JIP-1 Antibodies [0093]
  • Since JIP-1 is an inhibitor of JNK, inhibition of JIP-1 increases JNK activity. Increased JNK expression results in increased apoptosis, e.g., in neurons. Induction of apoptosis would be desirable in brain tumors, for example. Therefore, antibodies specific for JIP-1 can be used to inhibit JIP-1 expression. [0094]
  • Human JIP-1 proteins and polypeptides (or immunogenic fragments or analogs) can be used to raise antibodies; such polypeptides can be produced by recombinant or peptide synthetic techniques. See, e.g., [0095] Solid Phase Peptide Synthesis, supra; Ausubel et al., supra. In general, the peptides can be coupled to a carrier protein, such as KLH, mixed with an adjuvant, and injected into a host mammal. Antibodies can be purified by peptide antigen affinity chromatography.
  • In particular, various host animals can be immunized by injection with a JIP-1 protein or polypeptide. Host animals include rabbits, mice, guinea pigs, and rats. Various adjuvants can be used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete or incomplete); mineral gels, such as aluminum hydroxide; surface active substances such as lysolecithin; pluronic polyols; polyanions; peptides; oil emulsions; keyhole limpet hemocyanin; dinitrophenol; and potentially useful human adjuvants such as BCG (bacillus Calmette-Guerin) and [0096] Corynebacterium parvum. Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of the immunized animals.
  • The invention includes monoclonal antibodies, polyclonal antibodies, humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab′)[0097] 2 fragments, and molecules produced using a Fab expression library.
  • Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, can be prepared using the JIP-1 polypeptides described above and standard hybridoma technology. See, e.g., Kohler e: al., [0098] Nature, 256:495 (1975); Kohler et al., Eur. J. Immunol., 6:511 (1976); Kohler et al., Eur. J. Immunol., 6:292 (1976) Hammerling et al., In Monoclonal Antibodies and T Cell Hybridomas, Elsevier, N.Y. (1981); Ausubel et al., supra.
  • In particular, monoclonal antibodies can be obtained by any technique that provides for the production of antibody molecules by continuous cell lines in culture. Such methods include those described in Kohler et al., [0099] Nature 256:495 (1975), and U.S. Pat. No. 4,376,110. Other methods of producing monoclonal antibodies include the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4:72 (1983); Cole et al., Proc. Natl. Acad. Sci. USA 80:2026 (1983), and the EBV-hybridoma technique (Cole et al., Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 (1983)). Such antibodies can be of any immunoglobulin class, e.g., IgG, IgM, IgE, IgA, IgD, and any subclass thereof. Hybridomas producing the monoclonal antibodies of the invention may be cultivated in vitro or in vivo. The ability to produce high titers of monoclonal antibodies in vivo makes this the presently preferred method of production.
  • Polyclonal or monoclonal antibodies are tested for specific JIP-1 recognition by Western blot or immunoprecipitation analysis by standard methods. See, e.g., Ausubel et al., supra. Antibodies that specifically recognize and bind to JIP-1 are useful in the invention. These antibodies can be used in immunoassays to monitor the level of JIP-1 produced by a mammal, e.g., to determine the amount or subcellular location of JIP-1). [0100]
  • Preferably, antibodies of the invention are produced using JIP-1 polypeptides that correspond to regions of the JIP-1 protein that lie outside highly conserved regions and appear likely to be antigenic, by criteria such as high frequency of charged residues. Such fragments can be generated by standard PCR techniques and then cloned into the pGEX expression vector. Ausubel et al., supra. Fusion proteins are expressed in [0101] E. coli and purified using a glutathione agarose affinity matrix as described in Ausubel, et al., supra.
  • In some cases it may be desirable to minimize the potential problems of low affinity or specificity of antisera. In such circumstances, two or three fusion proteins can be generated for each protein, and each fusion protein can be injected into at least two rabbits. Antisera can be raised by injections in a series, preferably including at least three booster injections. [0102]
  • Antisera is also checked for its ability to immunoprecipitate recombinant JIP-1 proteins or control proteins, such as glucocorticoid receptor, CAT, or luciferase. [0103]
  • The antibodies can be used to detect JIP-1 in a biological sample as part of a diagnostic assay. Antibodies also can be used in a screening assay to measure the effect of a candidate compound on expression or localization of JIP-1. Additionally, such antibodies can be used in conjunction with the gene therapy techniques, e.g., to evaluate the normal and/or engineered JIP-1-expressing cells prior to their introduction into the patient. Such antibodies additionally can be used in a method for inhibiting abnormal JIP-1 activity. Such abnormal activity includes altered apoptosis and proliferation, resulting in neurodegenerative diseases, autoimmune disease, cancers such as leukemias. Other abnormal JIP-1 activity includes damage caused by ischemia/reperfusion, especially in heart disease, kidney damage, and stroke. [0104]
  • For theraputic uses, murine or other monoclonal antibodies should be altered to make them less immunogenic when administered to human patients. For example, techniques have been developed for the production of “chimeric antibodies.” See Morrison et al., [0105] Proc. Natl. Acad. Sci., 81:6851 (1984); Neuberger et al., Nature, 312:604 (1984); Takeda et al., Nature, 314:452 (1984). These techniques involve splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody of appropriate biological activity. Such chimeric antibodies have, e.g., a variable region derived from a murine antibody and a constant region derived from a human antibody.
  • Alternatively, single chain antibodies specific for JIP-1 polypeptides can be produced using known techniques. See, e.g., U.S. Pat. Nos. 4,946,778 and 4,704,692. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. [0106]
  • Antibody fragments that recognize and bind to specific epitopes of JIP-1 can also be generated by known techniques. Such fragments include, but are not limited to, F(ab′)[0107] 2 fragments, produced by pepsin digestion of antibody molecules, and Fab fragments, generated by reduction of the disulfide bonds of F(ab′)2 fragments. Alternatively, Fab expression libraries can be constructed to allow rapid and easy identification of monoclonal Fab fragments with a desired specificity. See, e.g., Huse et al., Science 246:1275 (1989).
  • Antibodies to JIP-1 can be used to generate anti-idiotypic antibodies that resemble a portion of JIP-1, using techniques well known to those skilled in the art. See, e.g., Greenspan et al., [0108] FASEB J. 7:437 (1993); Nisonoff, J. Immunol. 147:2429 (1991). For example, antibodies that bind to JIP-1 and competitively inhibit binding of other ligands can be used to generate anti-idiotypic antibodies resembling a ligand binding domain of JIP-1. These anti-idiotypic antibodies can bind and neutralize JIP-1 ligands. JIP-1 ligands include proline-rich regions of proteins, since JIP-1 has an SH3 domain that binds to these regions. There are ten isoforms of JNK. Gupta et al., EMBO J. 15:2760-2770 (1996). Each of these JNK isoforms is a JIP-1 ligand. Other kinases, which may be related to JNK, may also interact with JIP-1. Neutralizing anti-idiotypic antibodies or Fab fragments of anti-idiotypic antibodies can be used in therapeutic regimens.
  • Antisense Nucleic Acids [0109]
  • In alternate embodiments, therapies can be designed to reduce the level of endogenous JIP-1 gene expression, e.g., using antisense or ribozyme approaches to inhibit or prevent translation of JIP-1 mRNA transcripts; triple helix approaches to inhibit transcription of the JIP-1 gene; or targeted homologous recombination to inactivate or “knock out” the JIP-1 gene or its endogenous promoter. Delivery techniques can be designed to allow theraputic compositions to cross the blood-brain barrier (see, e.g., PCT WO89/10134). [0110]
  • Antisense approaches involve the design of oligonucleotides (either DNA or RNA) that are complementary to JIP-1 mRNA and inhibit expression of JIP-1 protein. Absolute complementarity of the antisense oligonucleotide to JIP-1, although preferred, is not required. A sequence “complementary” to a portion of an RNA, as referred to herein, means a sequence having sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may be tested, or triplex formation may be assayed. The ability to hybridize will depend on both the degree of complementarily and the length of the antisense nucleic acid. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures to determine the melting point of the hybridized complex. [0111]
  • Oligonucleotides that are complementary to the 5′ end of the message, e.g., the 5′ untranslated sequence up to and including the AUG initiation condon, should work most efficiently at inhibiting translation. However, sequences complementary to the 3′ untranslated sequences of mRNAs have been shown to be effective at inhibiting translation of mRNAs as well (Wagner, [0112] Nature, 372:333, 1994). Thus, oligonucleotides complementary to either the 5′- or 3′-non-translated, non-coding regions of the JIP-1 gene can be used in an antisense approach to inhibit translation of endogenous JIP-1 mRNA. Oligonucleotides complementary to the 5′ region overlapping the initiation codon can be used for this purpose. Antisense oligonucleotides that are complementary to the coding region can also be used. In designing suitable oligonucleotides, target regions are generally those lacking predicted secondary structure.
  • Antisense oligonucleotides complementary to mRNA coding regions can also be used in accordance with the invention. Whether designed to hybridize to the 5′, 3′, or coding region of JIP-1 mRNA, antisense nucleic acids should be at least six nucleotides in length, and are preferably oligonucleotides ranging from 6 to about 50 nucleotides in length. In specific aspects the oligonucleotide is at least 10 nucleotides, at least 17 nucleotides, at least 25 nucleotides or at least 50 nucleotides in length. [0113]
  • It is preferred that in vitro studies are first performed to quantitate the ability of the antisense oligonucleotide to inhibit gene expression. These studies preferably utilize controls that distinguish between antisense inhibition and nonspecific biological effects of oligonucleotides. It is also preferred that these studies compare levels of the target RNA or protein with that of an internal control RNA or protein. Additionally, it is envisioned that results obtained using the antisense oligonucleotide are compared with those obtained using a control oligonucleotide. It is preferred that the control oligonucleotide is of approximately the same length as the test oligonucleotide and that the nucleotide sequence of the oligonucleotide differs from the antisense sequence no more than is necessary to prevent specific hybridization to the target sequence. [0114]
  • The oligonucleotides can be DNA or RNA or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. Stability of the oligonucleotide can be improved by modification of the phosphodiester moieties by replacement with phosphorothioate, —CH2—, or —NH— groups. The oligonucleotides can also be stabilized by blocking the 5′ and 3′ termini with phosphorothioate, —CH2— or —NH— groups. Each of these modifications will lead to increased stability (e.g. by increasing modifications will lead to increased stability (e.g. by increasing resistance to nucleases) and therefore increased hybridization. The oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone. The oligonucleotide may include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (as described, e.g., in Letsinger et al., [0115] Proc. Natl. Acad. Sci. USA, 86:6553 (1989); Lemaitre et al., Proc. Natl. Acad. Sci. USA, 84:648 (1987)); PCT Publication No. WO 88/09810) or the blood-brain barrier (see, e.g., PCT Publication No. WO 89/10134), or hybridization-triggered cleavage agents (see, e.g., Krol et al., BioTechniques 6:958 (1988)), or intercalating agents (see e.g., Zon, Pharm. Res. 5:539 (1988)). To this end, the oligonucleotide can be conjugated to another molecule, e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent.
  • The antisense oligonucleotide can include at least one modified base moiety which is selected from the group including, but not limited to, 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethyl-aminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1-methylcytosine, N6-adenine, 7-methylguanine, 5-methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D-mannosylqueosine, 5′-methoxycarboxymethyluracil, 5-methoxyuracil, 2-methylthio-N6-isopentenyladenine, uracil-5-oxyacetic acid (v), wybutoxosine, pseudouracil, queosine, 2-thiocytosine, 5-methyl-2-theouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5-oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 2-(3-amino-3-N-2-carboxypropl) uracil, (acp3)w, and 2,6-diaminopurine. [0116]
  • The antisense oligonucleotide can also include at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2-fluoroarabinose, xylulose, and hexose. [0117]
  • In yet another embodiment, the antisense oligonucleotide includes at least one modified phosphate backbone selected from the group consisting of a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal, or an analog of any of these backbones. [0118]
  • In yet another embodiment, the antisense oligonucleotide is an á-anomeric oligonucleotide. An á-anomeric oligonucleotide forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual á-units, the strands run parallel to each other (Gautier et al., [0119] Nucl. Acids. Res., 15:6625 (1987)). The oligonucleotide is a 2′-0-methylribonucleotide (Inoue et al., Nucl. Acids Res., 15:6131 (1987)), or a chimeric RNA-DNA analog (Inoue et al., FEBS Lett., 215:327 (1987)).
  • Antisense oligonucleotides of the invention can be synthesized by standard methods known in the art, e.g., by use of an automated DNA synthesizer (commercially available from Biosearch, Applied Biosystems, etc.). As examples, phosphorothioate oligonucleotides can be synthesized by the method of Stein et al. ([0120] Nucl. Acids Res., 16:3209 (1988)), and methylphosphonate oligonucleotides can be prepared by use of controlled pore glass polymer supports (Sarin et al., Proc. Natl. Acad. Sci. USA, 85:7448 (1988)).
  • The antisense molecules should be delivered to cells that express JIP-1 in vivo. A number of methods have been developed for delivering antisense DNA or RNA to cells. For example, antisense molecules can be injected directly into specific tissue sites. Modified antisense molecules, designed to target the desired cells (e.g., antisense linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface) can be administered systemically. [0121]
  • One approach to achieving intracellular concentrations of the antisense molecule sufficient to suppress translation of endogenous mRNAs is to use a recombinant DNA construct in which the antisense oligonucleotide is placed under the control of a strong pol III or pol II promoter. The use of such a construct to transfect target cells in a patient will result in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the eldogenous JIP-1 transcripts, and thereby prevent translation of the JIP-1 mRNA. For example, a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA. [0122]
  • Such vectors can be constructed by recombinant DNA technology methods standard in the art. Vectors can be plasmid, viral, or others known in the art, used,for replication and expression in mammalian cells. Expression of the sequence encoding the antisense RNA can be by any promoter known in the art to act in mammalian, preferably human cells. Such promoters can be inducible or constitutive. Such promoters include, but are not limited to: the SV40 early promoter region (Bernoist et al., [0123] Nature, 290:304 (1981)); the promoter contained in the 3′ long terminal repeat of Rous sarcoma virus (Yamamoto et al., Cell, 22:787-797 (1988)); the herpes thymidine kinase promoter (Wagner et al., Proc. Natl. Acad. Sci USA, 78:1441, 1981); or the regulatory sequences of the metallothionein gene (Brinster et al., Nature, 296:39 (1988)).
  • Any type of plasmid, cosmid, YAC, or viral vector can be used to prepare the recominant DNA construct which can be introduced directly into specific tissue sites. Alternatively, viral vectors can be used that selectively infect the desired tissue (e.g., for brain, herpesvirus vectors may be used), in which case administration can be accomplished by another route (e.g., systemically). [0124]
  • Ribozymes [0125]
  • Ribozyme molecules designed to catalytically cleave JIP-1 mRNA transcripts also can be used to prevent translation of JIP-1 mRNA and expression of JIP-1 protein (see, e.g., PCT Publication WO 90/11364; Saraver et al., [0126] Science, 247:122 (1990)). While various ribozymes that cleave mRNA at site-specific recognition sequences can be used to destroy JIP-1 mRNAs, the use of hammerhead ribozymes is preferred. Hammerhead ribozymes cleave mRNAs at locations dictated by flanking regions that form complementary base pairs with the target mRNA. The sole requirement is that the target mRNA contain the following sequence of two bases: 5′-UG-3′. The construction and production of hammerhead ribozymes is well known in the art (Hasseloff et al., Nature, 334;585, 1988).
  • The ribozymes of the present invention also include RNA endoribonucleases (hereinafter “Cech-type ribozymes”), such as the one that occurs naturally in [0127] Tetrahymena Thermophila (known as the IVS or L-19 IVS RNA), which have been extensively characterized by Cech and his collaborators. See, e.g., Zaug et al., Science, 224:574 (1984); Zaug et al., Science, 231:470 (1986); Zaug et al., Nature, 324:429 (1986); PCT Application No. WO 88/04300; and Been et al., Cell, 47:207 (1986). The Cech-type ribozymes have an eight base-pair sequence that hybridizes to a target RNA sequence, whereafter cleavage of the target RNA takes place. The invention encompasses those Cech-type ribozymes that target eight base-pair active site sequences present in JIP-1. In general, target sites for ribozymes are regions that are predicted to lack appreciable secondary structure. Target sites for ribozymes include:
  • 5′[0128] GGUAUCGAUAAGCUUGAUAUCGCUGUCCGGAGC 3′ (SEQ ID NO:29) and
  • 5′[0129] AGAGGCACUGUCCCAUCCUGGGCCUGUUUCAUG 3′ (SEQ ID NO:30).
  • As in the antisense approach, the ribozymes can be composed of modified oligonucleotides (e.g., for improved stability, targeting, etc.), and should be delivered to cells which express JIP-1 in vivo. A preferred method of delivery involves using a DNA construct “encoding” the ribozyme under the control of a strong constitutive pol III or pol II promoter, so that transfected cells will produce sufficient quantities of the rihozyme to destroy endogenous JIP-1 messages and inhibit translation. Because ribozymes, unlike antisense molecules, are catalytic, a lower intracellular concentration is required for efficiency. Destruction of JIP-1 mRNA would be advantageous in increasing apoptosis in tumors; in altering the cell damage occurring during ischemic reperfusion in cardiovascular disease, kidney disease, and stroke; and altering cell damage occurring in the liver in response to metabolic oxidative stress. [0130]
  • Other Methods for Reducina JIP-1 Expression [0131]
  • Endogenous JIP-1 gene expression can also be reduced by inactivating or “knocking out” the JIP-1 gene or its promoter using targeted homologous recomination (see, e.g., U.S. Pat. No. 5,464,764). For example, a mutant, non-functional JIP-1 (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous JIP-1 gene (either the coding regions or regulatory regions of the JIP-1 gene) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express JIP-1 in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the JIP-1 gene. Such approaches are particularly suited for use in the agricultural field where modifications to ES (embryonic stem) cells can be used to generate animal offspring with an inactive JIP-1. However, this approach can be adapted for use in humans, provided the recombinant DNA constructs are directly administered or targeted to the required site in vivo using appropriate viral vectors, e.g., herpes virus vectors for delivery to brain tissue. [0132]
  • Alternatively, endogenous JIP-1 gene expression can be reduced by targeting deoxyribonucleotide sequences complementary to regulatory regions of the JIP-1 gene (e.g., a JIP-1 promoter and/or enhancer to form triple helical structures that prevent transcription of the JIP-1gene in target cells in the body. See, e.g., Helene, [0133] Anticancer Drug Des. 6:569 (1981); Helene et al., Ann N.Y. Acad. Sci. 660:27 (1992); Maher, Bioassays 14:807 (1992).
  • Identification of Proteins that Interact with JIP-1 [0134]
  • The invention also features polypeptides that interact with JIP-1. Any method suitable for detecting protein-protein interactions can be employed for identifying intracellular or extracellular proteins that interact with JIP-1. Among the traditional methods that can be employed are co-immunoprecipitation, crosslinking, and co-purification through gradients or chromatographic columns of cell lysates, or proteins obtained from cell lysates. Purified proteins are used to identify proteins in the lysate that interact with JIP-1. For these assays, the JIP-1 polypeptide can be full length JIP-1, a soluble domain of JIP-1, or some other suitable JIP-1 polypeptide. [0135]
  • To characterize JIP-1 interacting proteins, portions of their amino acid sequences can be ascertained using techniques well known to those of skill in the art, such as the Edman degradation technique. The amino acid sequences obtained can be used to design degenerate oligonucleotide probes that can be used to screen for gene sequences encoding the interacting protein. Screening may be accomplished, for example, by standard hybridization or PCR techniques. Techniques for the generation of degenerate oligonucleotide mixtures and screening are well-known. See Ausubel, supra; and Innis et al., eds., [0136] PCR Protocols: A Guide to Methods and Applications, Academic Press, Inc., New York (1990).
  • Additionally, methods can be used that result directly in the identification of genes that encode proteins that interact with JIP-1. These methods include, for example, screening expression libraries in a manner similar to the well known technique of antibody probing of λgt11 libraries using labeled JIP-1 polypeptides or a JIP-1 fusion protein, e.g., an JIP-1 polypeptide or domain fused to a marker such as an enzyme, a fluorescent dye, a luminescent protein, or to an IgFc domain. [0137]
  • Protein interactions can also be identified in vivo using the two-hybrid system. Chien et al., [0138] Proc. Natl. Acad. Sci. USA, 88:9578 (1991). A kit for practicing this method is available from Clontech (Palo Alto, Calif.). In this system, two plasmids, each encoding a hybrid protein, are constructed. One plasmid, the “bait” plasmid, includes a nucleotide sequence encoding the DNA-binding domain of a transcription activator protein fused to a nucleotide sequence encoding a protein of interest. The other plasmid includes a nucleotide sequence encoding the transcription activator protein's activation domain and an unknown protein, which may bind the protein of interest. The plasmids are transformed into a strain of Saccharomyces cerevisiae that contains a reporter gene (e.g., HIS or lacZ) whose regulatory region contains the transcription activator's binding site. Neither of the hybrid proteins can activate transcription of the reporter gene alone; the bait plasmid lacks an activation function, and the other plasmid cannot localize to the transcription activator's binding sites. If the protein of interest and the unknown protein form a protein-protein complex and reconstitute the proximity of the DNA binding domain and the activation domain, this complex can bind to the regulatory region of the reporter gene, and the reporter gene will be expressed.
  • The two-hybrid system or related methodology can be used to screen libraries for proteins that interact with the “bait” gene product. By way of example, JIP-1 can be used as the bait gene product. Total genomic or cDNA sequences are fused to the DNA encoding an activation domain of a transcriptional activator protein. This library and a plasmid encoding a hybrid of bait JIP-1 gene product fused to the DNA-binding domain are cotransformed into a yeast reporter strain, and the resulting transformants are screened for those that express the reporter gene. For example, a bait JIP-1 gene sequence, such as JIP-1 or a domain of JIP-1, can be cloned into a vector such that it is translationally fused to the DNA encoding the DNA-binding domain of the GAL4 protein, to form a bait plasmid. Colonies are purified and the library plasmids responsible for reporter gene expression are isolated. The library plasmids are subjected to DNA sequencing to determine the sequences of the gene encoding the JIP-1 binding proteins. [0139]
  • As an example, a cDNA library can be made by methods that are routine in the art. This library can be inserted into vectors so that the sequences from the library are fused to nucleotide sequences encoding the transcriptional activation domain of the GAL4 protein. These vectors can then be co-transformed, along with a bait JIP-1 gene-GAL4 fusion plasmid, into a yeast strain which contains a lacZ gene driven by a promoter that contains the GAL4 activation sequence. If any of the library vectors encode hybrid proteins that bind the hybrid JIP-1 protein encoded by the bait plasmid, a functional GAL4 will be formed, and the HIS3 gene will be expressed. Colonies expressing HIS3 can then be purified, and used to produce and isolate the JIP-1 interacting protein using techniques routinely practiced in the art. [0140]
  • Therapeutic Compositions of JIP-1 Nucleic Acids, Peptides, and Polypeptides [0141]
  • Therapeutic compositions of the JNK inhibitor JIP-1 can be used to treat pathological conditions associated with apoptosis or transformation. These compositions can include JIP-1 polypeptides or peptides that specifically bind to and sequester JNK. Proteins can be purified by methods known to those skilled in the art. Ausubel, supra. Peptides can be synthesized by methods that are known to those skilled in the art. See, e.g., Merrifield, [0142] J. Am. Chem. Soc., 85:2149-2154 (1963).
  • Peptide mimetics are also included in the therapeutic compositions of the invention. These compounds mimic the activity of the peptide or polypeptide, but are composed of molecules other than, or in addition to, amino acids. The design of such mimetics is described in, e.g., Hughes, [0143] Philos. Trans. R. Soc. Lond., 290:387-394 (1980); Hodgson, Biotechnol., 9:19-21 (1991); Suckling, Sci. Prog., 75:323-359 (1991).
  • Therapeutic compositions of the invention also include nucleic acids encoding a JIP-1 polypeptide or peptide. These nucleic acids can be administered in a manner allowing their uptake and expression by cells in vivo. Compositions containing nucleic acids can be prepared for administration by methods that are routine in the art. [0144]
  • Therapeutic compositions of the invention can include one or more compounds, e.g., nucleic acids, peptides, polypeptides, or peptide mimetics and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are biologically compatible vehicles, e.g., physiological saline, which are suitable for administration to a patient. [0145]
  • Nucleic acids can be administered to the patient by standard vectors and/or gene delivery systems. Suitable gene delivery systems include liposomes, receptor-mediated delivery systems, naked DNA and viral vectors such as herpes viruses, retroviruses, adenoviruses and adeno-associated viruses. [0146]
  • Peptides can be coupled to membrane permeable peptides in order to facilitate their uptake by cells. This can be done by colinear synthesis of a membrane permeable peptide with a peptide sequence of interest. The two peptides could also be crosslinked together. Methods of coupling are described in, e.g., Lin et al., [0147] J. Biol. Chem., 269:12320-12324 (1996); Rojas et al., J. Biol. Chem., 271:27456-127461 (1997). Peptides can also be coupled to lipids to provide membrane permeability. See, e.g., Vijayaraghavan, J. Biol. Chem., 272:4747-4752 (1997). As an example, a fatty acid can be coupled to the α-NH2 group of the peptide as an amide.
  • To enable the compositions to penetrate the blood-brain barrier, they can be delivered in encapsulated cell implants (e.g., those produced by CytoTherapeutics, Inc., Providence R.I.; see [0148] Bioworld Today 7:6 (Monday, Dec. 2, 1996)). Delivery of drugs to the brain can also be accomplished using RMP-7™ technology (Alkermes, Inc., Cambridge, Mass.; see Business Wire, “Third Major Agreement for Prolease Sustained Release Drug Delivery System,” Dec. 2, 1996) or implantable wafers containing the drug (see PR Newswire, “Implantable Wafer is First Treatment to Deliver Chemotherapy Directly to Tumor Site,” Sep. 24, 1996). The compositions can also be administered using an implantable pump for direct administration into intrathecal fluid (e.g., that made by Medtronic, Minneapolis, Minn.; see Genetic Engineering News, “Neurobiotechnology Companies Focus Programs on Pain and Neuroprotection,” Nov. 1, 1996).
  • Administration of Therapeutic Compositions [0149]
  • Parenteral administration, such as intravenous, subcutaneous, intramuscular, or intraperitoneal delivery routes can be used to deliver the therapeutic compositions of the invention. Dosages for particular patients depend upon many factors, including the patient's size, body surface area, age, the particular substance to be administered, time and route of administration, general health and other drugs being administered concurrently. The amount of therapeutic composition to be administered to a patient can be in the range of 1 to 1000 μg/kg of body weight, e.g., 10 to 500 μg/kg, or 20 to 200 μg/kg of body weight. A typical dose of peptide or nucleic acid to be administered to a patient is 100 μg per kilogram of body weight. [0150]
  • EXAMPLES Example 1 Identification of JIP-1
  • The yeast two hybrid method was used to screen a mouse embryo cDNA library to identify proteins that interact with JNK. The method is described in detail in Fields et al., [0151] Nature, 340:245 (1989). The yeast strain used was L40 (MATa hisΔ200 trp1-901 leu2-3, 112 ade2 LYS:: (lexAop)4-HIS URA3:: (lexAop)8-lacZ. Vojtek, et al., Cell, 74:205 (1993). Human JNK1 fused to the LexA DNA binding domain was used as the bait. The bait plasmid, pLexA-JNK1, was constructed by blunt-end ligation of a XbaI/HindIII fragment of pCMV-Flag-JNK1 (Derijard et al., Cell, 76:1025 (1994); Sluss et al., Mol. Cell Biol., 14:8376 (1994); Gupta et al., EMBO. J., 15:2760 (1996)) into the vector pBTM116 at the SmaI and SalI sites. Yeast transformants (5.2×106) were examined for growth on media with 25 mM 3-aminotriazole in the absence of histidine. Positive clones were confirmed by measurement of lacZ expression.
  • The two-hybrid screen yielded a cDNA fragment encoding a portion of a JNK binding protein. A group of 7 independent clones, corresponding to overlapping fragments of this cDNA, was identified by sequencing. Full-length cDNA clones were obtained by screening a mouse brain λZAPII cDNA library (Stratagene Inc.) with a random-primed cDNA fragment corresponding to base pairs 560-1020 of the full length cDNA. Isolated cDNA clones were sequenced using an Applied Biosystems model 373A machine. The sequence of the 5′ GC-rich non-coding region of the largest cDNA clone was confirmed using the Maxam-Gilbert method. Maxam et al., [0152] Proc. Natl. Acad. Sci. USA, 74:560-564 (1977). The amino acid sequence of the encoded protein was deduced from the cDNA sequence. Single letter abbreviations for the amino acid residues are as follows: A, Ala; C, Cys; D, Asp; E, Glu; F, Phe; G, Gly; H, His; I, Ile; K, Lys; L, Leu; M, Met; N, Asn; P, Pro; Q, Gln; R, Arg; S, Ser; T, Thr; V, Val; W, Trp; and Y, Tyr.
  • The largest cDNA clone isolated from the mouse brain library was 2790 base pairs in length, and contained a predicted coding region of 660 amino acids in the same reading frame as the partial clones obtained in the two-hybrid screen. The protein encoded by this cDNA, designated JNK interacting protein-1 (JIP-1), contains an amino terminal JNK binding domain (defined by the overlapping 2-hybrid clones) and a putative SH3 domain in the carboxy terminus. The structure and amino acid and nucleotide sequences of JIP-1 are shown in FIGS. [0153] 1A-1C. The JNK binding domain (JBD; residues 127-281, SEQ ID NO:4) and the putative SH3 domain (residues 491-540, SEQ ID NO:13) are indicated by boxes. The putative SH3 domain of JIP-1 is highly related to the SH3 domains located in the tyrosine kinase c-fyn and the p85 subunit of PI-3′ kinase.
  • Human homologs of the murine JIP-1 gene can be isolated using the murine JIP-1 cDNA clones or fragments of those clones as probes, by methods that are routine in the art of molecular biology. For example, a library of human cDNA can be screened with a fragment of murine JIP-1 cDNA, and human cDNA hybridizing to the murine JIP-1 cDNA can be isolated, cloned, sequenced and analyzed for structural and functional similarity to murine JIP-1. For general methods, of isolating and characterizing homologous genes from libraries, see Sambrook et al., [0154] Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (2d ed. 1989).
  • The tissue distribution of Jip-1 mRNA was examined by Northern blot analysis of poly (A) RNA isolated from different murine tissues. Northern blots were performed using 2 μg of polyA[0155] + RNA isolated from various murine tissues (Clontech). The blots were hybridized to a probe that was prepared by labeling JIP-1 cDNA (base pairs 515-970) with [α-32P] dCTP (Amersham International PLC) by the random priming method (Stratagene Inc.) according to the manufacturer's instructions. The integrity of the mRNA samples was confirmed by hybridization to an actin probe. The blots were washed three times with 1×SSC, 0.05% SDS, and 1 mM EDTA prior to autoradiography. The results indicate that JIP-1 is expressed in many different tissues, including brain, heart, spleen, lung, liver, muscle, kidney and testis. Highest amounts of JIP-1 mRNA were detected in brain, kidney, and testis.
  • Example 2 JIP-1 Specifically Binds JNK in vivo
  • To test whether JIP-1 and JNK interact in vivo, co-immunoprecipitation analysis was performed. COS-′1 cells were mock-transfected or transfected with JIP-1 and JNK1 expression vectors. Constructs expressing epitope (HA) tagged JNK1 have been described previously. Dérijard et al., supra; Sluss et al., supra; Gupta et al., supra. Mammalian JIP-1 expression vectors were constructed by subcloning the JIP-1 cDNA into the XbaI and HindIII sites of pCMV5 and the HindIII and EcoRI sites of pcDNA3 (Invitrogen Inc.). DNA encoding the Flag epitope, -Asp-Tyr-Lys-Asp-Asp-Asp-Asp-Lys-(SEQ ID NO:14) (Immunex Corp.), was inserted into JIP-1 cDNA between the DNA encoding the first two codons of JIP-1 using insertional overlapping polymerase chain reaction (PCR). Ho et al., [0156] Gene, 77:51 (1989). Bacterial JIP-1 expression vectors were constructed by subcloning PCR fragments of the JIP-1 cDNA into the EcoRI and XbaI sites of pGEX-3×(Pharmacia LKB Biotechnology Inc.). GST fusion proteins were purified by affinity chromatography on GSH-agarose as described previously. Smith et al., Gene, 67:31 (1988). Sequences of the constructs were confirmed using an Applied Biosystems model 373A machine.
  • For the co-immunoprecipitation experiments, transfected or mock-transfected cells were irradiated with or without UV-C (40 J/m[0157] 2) and incubated for one hour. Lysates prepared from the cells were examined by protein immunoblot analysis using a mixture of antibodies specific for Flag and HA to detect Flag-JIP-1 and HA-JNK1, respectively. Cells were lysed in TLB (20 mM Tris (pH 7.5), 1% Triton X-100, 10% glycerol, 0.137 M NaCl, 25 mM sodium β-glycerophosphate, 1 mm sodium orthovanadate, 2 mM sodium pyrophosphate, 2 mM EDTA, 10 μg/ml leupeptin, 1 mM phenylmethylsulfonylfluoride). Soluble extracts were prepared by centrifugation at 100,000×g for 30 minutes at 4° C. The extracts were pre-cleared using protein G-Sepharose (Pharmacia-LKB Biotechnologies Inc.) and incubated for one hour with monoclonal antibody to the Flag epitope (M2; IBI-Kodak) or the HA epitope (12CA5; Boehringer-Mannheim) pre-bound to protein G-Sepharose. The immunoprecipitates were washed three times with TLB and once with 25 mM Hepes (pH 7.5), 0.2% (w/v) Triton X-100, 1 mM EDTA. Proteins were fractionated by SDS-PAGE and transferred electrophoretically to Immobilon-P membranes (Millipore). The membranes were blocked with 10% gamma globulin-free horse serum (Gibco-BRL) and probed with the M2 monoclonal antibody, to detect Flag-JIP-1, and either the 12CA5 monoclonal antibody or a sheep anti-JNK polyclonal antibody, to detect HA-JNK1. Immune complexes were detected with a second antibody coupled to horseradish peroxidase and enhanced chemiluminescence (Amersham International PLC)
  • JIP-1 was detected in JNK1 immunoprecipitates by protein immunoblot analysis, and JNK1 was detected in JIP-1 immunoprecipitates. Co-immunoprecipitation of JIP-1 with JNK2, was also observed. These data indicate that JIP-1 specifically binds JNK in vivo. Exposure to UV radiation caused no significant change in the amount of the JNK/JIP-1 complex detected by co-immunoprecipitation analysis. Control experiments performed to examine the specificity of the interaction of JIP-1 with JNK demonstrated that the related MAP kinases ERK2 and p38 did not co-immunoprecipitate with JIP-1. The absence of co-immunoprecipitation of JIP-1 with these MAP kinases demonstrates that JIP-1 forms a specific complex with JNK. [0158]
  • This same assay can be used to determine whether a specific polypeptide is a JIP-1 polypeptide. [0159]
  • Example 3 JIP-1 Interacts Directly with JNK
  • To test whether JIP-1 interacts directly with JNK, in vitro binding assays were performed. The putative JNK binding domain (JBD; amino acid residues 127-281 of JIP-1), defined by the clones obtained in the two-hybrid screen, was expressed as a glutathione-S-transferase (GST) fusion protein. GST-fusion proteins were purified by affinity chromatography on GSH-agarose as described previously. Smith et al., [0160] Gene, 67:31 (1988). Recombinant JNK, prepared by in vitro translation in the presence of [35S]methionine (Dérijard et al., supra; Sluss et al., supra; Gupta et al., supra), was incubated with GST-JIP-1 immobilized on GSH-agarose. Control experiments revealed no detectable binding of JNK isoforms to GST alone. In contrast, in assays using GST-JNK fusion proteins representing ten different human JNK isoforms, each of these proteins exhibited similar amounts of binding to JIP-1. JIP-1 therefore interacts with multiple JNK isoforms.
  • Previous studies have demonstrated that JNK binds to the transcription factors c-Jun and ATF2, and that the binding of JNK to these transcription factors is isoform-dependent. To test whether JIP-1 binding to JNK is also isoform-dependent, a binding assay was utilized. Cell lysates (1 ml in TLE) containing JNK1, JNK2, or p38 MAP kinase, each tagged with the Flag epitope, were incubated with 5 μg GST-fusion proteins pre-bound to 10 μl glutathione-Sepharose. The GST fusion proteins used contain residues 127-202 (SEQ ID NO:15), 203-281 (SEQ ID NO:16), 164-240 (SEQ ID NO:17), or 127-281 (SEQ ID NO:4) of JIP-1. The GST-ATF2 fusion protein contains residues 1-109 of ATF2, and the GST-Jun fusion protein contains residues 1-79 of c-Jun. After incubation for one hour at 4° C. and three washes with TLB, bound proteins were detected by protein immunoblot analysis with the M2 monoclonal antibody, which is specific for the Flag epitope. Dérijard, et al., supra; Sluss et al., supra; Gupta et al., supra. When ATF2 or c-jun GST fusion proteins were used in this assay, binding to JNK1 was greater than binding to JNK2. This finding is consistent with the results of previous studies. In contrast, when GST-JIP-1 fusion proteins containing residues 127-202 or 127-281 of JIP-1 were tested in the assay, these proteins bound both JNK1 and JNK2. The level of binding of these fusion proteins to JNK1 was similar to the level of binding to JNK2. The binding of JNK to JIP-1 was significantly greater than the binding of JNK to ATF2 or c-Jun. Control experiments demonstrated that JIP-1 did not bind to p38 MAP kinase. These data establish that JIP-1 binding to JNK1 is quantitatively similar to JIP-1 binding to JNK2, and that JNK binding to JIP-1 is significantly greater than JNK binding to the transcription factors ATF2 and c-Jun. [0161]
  • The GST-JIP-1 fusion proteins containing various portions of the JIP-1 gene were used to define regions of JIP-1 that are required for JNK interaction. A GST-JIP-1 fusion protein containing residues 127-281 of JIP-1 bound both JNK1 and JNK2. No JNK binding was detected in experiments using the central region (residues 164-240) or the carboxy terminal region (residues 203-281) of JIP-1. However, JNK binding activity was observed in experiments using the amino terminal region (residues 127-202 of JIP-1). These data indicate that residues 127-164 of JIP-1 are required for JNK binding activity. [0162]
  • To more precisely define the JIP-1 sequence required for JNK binding activity, additional GST-JNK fusion proteins; were constructed. These fusion proteins contained residues 135-202 (SEQ ID NO:18), 144-202 (SEQ ID NO:19), 154-202 (SEQ ID NO:20), 164-202 (SEQ ID NO:21), 127-143 (SEQ ID NO:22), 127-153 (SEQ ID NO:23), or 127-163 (SEQ ID NO:24, of JIP-1. Proteins containing residues 127-202, 135-202, 144-202, 154-202, and 127-163 all bound both JNK1 and JNK2. Thus, JIP-1 residues 144-163 are important for the interaction of JIP-1 with JNK. As shown in FIG. 2, this region of JIP-1 shares sequence similarity with the JNK binding domains of ATF2 and c-Jun. [0163]
  • This assay can also be used to analyze a given polypeptide to determine whether it is, or is not, a JIP-1 polypeptide. [0164]
  • Example 4 A Small NH2-terminal Region of JIP-1 is Sufficient for Interaction with JNK
  • The effect of increasing concentrations (0, 4, 8, 16, 32, and 64 μg/ml) of a synthetic peptide corresponding to JIP-1 residues 148-174 (SEQ ID NO:3) or a control peptide having a scrambled sequence (SEQ ID NO:12) on JIP-1-JNK interaction was examined. Peptides were synthesized using an Applied Biosystems machine. Cell lysates containing Flag epitope-tagged JNK were incubated with GST-JIP-1 (residues 127-281) prebound to glutathione-Sepharose. The beads were washed, and bound proteins were detected by protein immunoblot analysis with a Flag-specific antibody. While the control peptide had no effect, incubation with the synthetic peptide corresponding to residues 148-174 of JIP-1 resulted in a dose-dependent decrease in JIP-1 binding to JNK1, indicating that this peptide competes with JIP-1 for binding to JNK. [0165]
  • The JNK binding domain of JIP-1 contains three amino eacids, Lys-155, Thr-159, and Leu-160, that are conserved in the JNK binding domains of ATF2 and c-Jun (FIG. 2). A hydrophobic amino acid, Leu, is found at residue 162 of JIP-1. ATF2 and c-Jun also contain hydrophobic amino acids (ATF2, Phe; c-Jun, Leu) in the position corresponding to residue 162 of JIP-1. To test if these conserved residues are involved in JNK binding, the wild type JIP-1 JBD (residues 148-174) was substituted with glycines in these positions to produce the peptides shown in FIG. 3 (SEQ ID NOs:7-11). The mutant peptides are identical to the wild type peptide except for the indicated glycine substitutions. The binding of JNK1 to GST, as well as a GST fusion protein containing JIP-1 residues 127-281, was examined in the absence and presence of the synthetic peptides (64 μg/ml). A peptide with a scrambled sequence (SEQ ID NO:12) was used as a control. The peptide representing the wild-type JIP-1 sequence caused a dose-dependent inhibition of JIP-1 binding to JNK. In contrast, the control peptide caused no change in JNK binding. When any of the glycine-substituted peptides was used in the assay, the inhibition of JNK binding was greatly reduced compared to that observed with the wild type peptide. These data indicate that Lys-155, Thr-159, Leu-160, and Leu-162 are involved in JIP-1 binding to JNK. [0166]
  • Example 5 JIP-1 is a Selective Inhibitor of JNK Activity
  • To test whether JIP-1 competes with Jun and ATF2 for interaction with JNK, the effect of JIP-1 on transcription factor-mediated phosphorylation of exogenous substrates was analyzed using an in vitro protein kinase assay. In these experiments, Chinese hamster ovary (CHO) cells were serum-starved for one hour. In some experiments, the cells were treated with 10 ng/[0167] ml mouse interleukin 1 or 100 nM phorbol myristic acetate. JNK, p38, and ERK protein kinase activity was measured in an immune complex kinase assay using 3 μg of the substrates GST-Jun, GST-ATF2, and myelin basic protein (MBP), respectively. Protein kinase assays were performed using 40 μl 20 mM Hepes (pH 7.4), 20 mM MgCl2, 20 mM β-glycerophosphate, 2 mM dithiothreitol, 0.1 mM sodium orthovanadate, 50 μM [γ-32P]ATP (10 Ci/mmol). After incubation for 30 minutes at 30° C., phosphorylation of substrates was analyzed by polyacrylamide gel electrophoresis and autoradiography.
  • The results show that JIP-1 markedly inhibited the phosphorylation of c-Jun by JNK. However, JIP-1 caused no significant change in the phosphorylation of substrates by the related MAP kinases p38 and ERK2. JIP-1 is thus a selective inhibitor of JNK. [0168]
  • This same assay can be used to determine whether a specific polypeptide has the same phosphorylation function of wildtype JIP-1. [0169]
  • Example 6 Expression of JIP-1 Inhibits Targets of the JNK-regulated Signal Transduction Pathway
  • The effect of JIP-1 on targets of the JNK signal ransduction pathway, including the transcription factors c-Jun, ATF2 and Elk-1, was examined to determine whether JIP-1 inhibits signal transduction by JNK. [0170]
  • In these experiments, CHO cells were cotransfected with constructs encoding the transcription factors and JIP-1 using previously described methods. Dérijard et al., supra; Sluss et al., supra; Gupta et al., supra. A luciferase reporter plasmid was used to monitor the expression of the transcription factors. Transfection efficiency was determined using a β-galactosidase expression vector. Constructs used in these experiments included GAL4 fusions with the c-myc, Sp1 and VP16 activation domains (described in Whitmarsh et al., [0171] Science, 269:403 (1995); Davis, Science, 269:403 (1995); Gille et al., Curr. Biol., 5:1191 (1995)). Other constructs used were GAL4-c-Jun, in which GAL4 is fused to c-Jun; GAL4-c-Jun (S63A/S73A), in which GAL4 is fused to a mutant c-Jun in which the serines at positions 63 and 73 have been changed to alanines; GAL4-ATF2, in which GAL4 is fused to wild type ATF2 (ATF2(Thr-69, 71)); GAL4-ATF2 (T69A/T71A) (ATF2(Ala-69/71)), in which GAL4 has been fused to a mutant ATF2 in which the tyrosines at positions 69 and 71 have been changed to alanines; GAL4-Elk-1, in which GAL4 has been fused to Elk-1; and GAL4-Elk-1 (S383A), in which GAL4 has been fused to a mutant Elk-1 in which the serine at position 383 has been changed to an alanine. Gupta et al., EMBO J, 15:2760-2770 (1996); Raingeaud et al., Mol. Cell. Biol., 16:1247-1255 (1996); Whitmarsh et al., Science, 269:403-407 (1995).
  • Insertional overlapping PCR, described in detail in Ho, supra, was used to construct expression vectors for the JNK binding domain (JBD; amino acid residues 127-281) of JIP-1 and for a mutant JIP-1 lacking the SH3 (amino acid residues 491-540) domain. [0172]
  • Transfected cells were activated by treatment with 10% (v/v) fetal calf serum, and luciferase and β-galactosidase activities were measured in cell lysates at 48 hours post-transfection. The results are shown in FIGS. [0173] 4A-4C. The data are presented as the ratio of luciferase activity (light units) to β-galactosidase activity (OD units) measured in the cell extracts (mean±SEM (n=3)), and are normalized to the luciferase activity detected in the absence of JBD (FIGS. 4A and 4B) or ATF2 (FIG. 4C).
  • Control experiments demonstrated that the JNK binding domain (JBD; residues 127-281 of JIP-1) did not inhibit reporter gene expression mediated by the activation domains of c-Myc, E1a, Sp1, and VP16 (FIG. 4A). Significant inhibition of c-Jun and ATF2 transcriptional activity by JIP-1 was observed, however (FIG. 4B). The partial inhibition of Elk-1 transcriptional activity shown in FIG. 4B may reflect an association of both ERK and JNK MAP kinases with Elk-1 regulation. Mutation of the JNK phosphorylation sites in ATF2, c-Jun, and Elk-1 caused lower basal transcriptional activity that was not markedly inhibited by JIP-1. [0174]
  • As shown in FIG. 4C, inhibition of JNK-regulated gene expression was observed in experiments using wild-type JIP-1, the JNK binding domain (JBP), and ΔSH3, a JIP-1 deletion mutant lacking the SH3 domain. Together, these data indicate that JIP-1 suppresses JNK-regulated gene expression, and that the JNK binding domain is sufficient for this activity. [0175]
  • This assay can be used to determine whether specific polypeptides have the same effect on signal transduction as full length, wildtype JIP-1. [0176]
  • Example 7 Subcellular Distribution of JIP-1
  • The subcellular distribution of JIP-1 was analyzed by performing indirect immunofluorescence on JIP-1-expressing cells. In these experiments, the cells were grown on coverslips, fixed with 4% paraformaldehyde, permeabilized with 0.25% Triton X-100, and processed for immunofluorescence microscopy. JIP-1 was detected in the cytoplasm, but not the nucleus, of control and UV-irradiated cells. In contrast, JNK is detected in both the cytoplasmic and nuclear compartments. [0177]
  • It is likely that it is JNK in the nucleus which is involved in the regulation of gene expression. Since JIP-1 is cytoplasmic, it was unclear how it could inhibit the nuclear function of JNK. To investigate the mechanism of JIP-1 action, the distribution of JNK was examined in cells that had been transfected with either hemagglutinin (HA)-tagged JNK1 (HA-JNK1) alone, or HA-JNK1 and Flag-tagged JIP-1 (Flag-JIP-1). In these experiments, the cells were exposed to a potent JNK activator (40 J/m[0178] 2 UV-C) for one hour prior to processing for immunofluorescence. The primary antibodies used were rabbit anti-HA (12CA5; Boehringer Mannheim), which recognizes HA-tagged JNK1; and mouse monoclonal anti-Flag (M2; IBI-Kodak), which recognizes Flag-tagged JIP-1. The secondary antibodies were Texas Red-goat anti-mouse Ig and fluoroscein isothiocyanate-conjugated donkey anti-rabbit Ig (Jackson Immunoresearch). Procedures for digital imaging microscopy and image restoration using the exhaustive photon reassignment algorithm are described in Carrington et al., Science, 268:1483 (1995). Individual optical sections were inspected using computer graphics software on a Silicon Graphics workstation.
  • The results demonstrate that expression of JIP-1 reduces the amount of nuclear JNK detected in control and UV-irradiated cells. In contrast, JIP-1 expression has no significant effect on the subcellular distribution of p38 MAP kinase, which is also located in both nuclear and cytoplasmic compartments of cultured cells. These data indicate that JIP-1 expression results in selective cytoplasmic retention of JNK. It is likely that this cytoplasmic retention contributes to the ability of JIP-1 to inhibit JNK-mediated signal transduction. [0179]
  • Example 8 JIP-1 Inhibits Nerve Growth Factor Withdrawal-Induced Apoptosis
  • The effect of JIP-1 on this biological response was investigated to determine whether JIP-1 inhibits the biological effects of the JNK signaling pathway. NGF withdrawal-induced apoptosis of differentiated PC12 cells was examined following transfection with the expression vectors pcDNA3 (control) and pcDNA3-Flag-JIP-1 (containing residues 127-281 of JIP-1) using methods described previously. Xia et al., [0180] Science, 270:1326 (1995). The percentage of apoptotic cells in the total transfected cell population was blind-scored and quantitated. This assay scores adherent cells with an apoptotic morphology at 17 hours following NGF withdrawal. Cells that have completed apoptosis are nonadherent and are not scored. Thus, while the cumulative extent of apoptosis is large (100%), lower numbers of apoptotic cells are detected at a single time point.
  • The results are shown in FIG. 5. The data shown are representative of three independent experiments. The error bars in the graph indicate the SEM and the numbers within each bar are the total number of transfected cells counted. Expression of the JNK binding domain of JIP-1 (JBD; residues 127-281) in differentiated PC12 cells markedly reduced apoptosis following NGF-withdrawal. These data demonstrate that JIP-1 suppresses JNK-mediated signal transduction. This assay can be used to test new JIP-1 polypeptides. [0181]
  • Example 9 JIP-1 Inhibits Pre-B Cell Transformation by BCR-ABL
  • The BCR-ABL oncogene, which is associated with chronic myelogenous leukemia (CML), causes JNK activation in the absence of increased ERK activity. Raitano et al., [0182] Proc. Natl. Acad. Sci. USA, 92:11746 (1995). Oncogenic transformation by BCR-ABL may be mediated in part by the JNK signaling pathway. Since JIP-1 can inhibit JNK activity, the effect of JIP-1 expression on oncogenic transformation was examined. Plasmid vectors expressing v-ABL or BCR-ABL and Flag-tagged JBD (residues 127-281) of JIP-1 were used to transfect 293 cells. JNK activity was then measured in an immune complex kinase assay of lysates of the transfected 293 cells, using a polyclonal JNK antibody and GST-Jun as the substrate. Control experiments demonstrated that v-ABL and BCR-ABL caused constitutive JNK activation (approximately 5-fold), which was blocked by co-expression of the JBD of JIP-1. JIP-1 can therefore inhibit BCR-ABL-mediated activation of JNK.
  • To examine the effect of JIP-1 on BCR-ABL-mediated cellular transformation, bone marrow transformation assays were performed using recombinant retroviruses packaged with 293T cells as described in Sawyers et al., [0183] J. Exp. Med., 181:307 (1995). Bicistronic retroviruses expressing BCR-ABL, alone, or in combination with the Flag-tagged JBD (residues 127-281) of JIP-1, were prepared by subcloning p185BCR-ABL into the ClaI site of pSRαTK, downstream of the internal TK promoter, to create pSRαMSVTKp185. The JBD was subcloned into the upstream EcoRI site in the sense and antisense orientations. The structures of the retroviral constructs are shown in FIG. 6.
  • Transfection of 293 cells with these retroviral constructs resulted in expression of the appropriate proteins, as demonstrated by immunoblot analysis of whole cell lysates using antibodies specific for ABL to detect BCR-ABL, and antibodies to Flag to detect the JBD of JIP-1. The recombinant retroviruses were then used to infect primary mouse marrow cells, and the transformation of pre-B cells was monitored in culture. FIG. 6 shows the mean density (±SE) of non-adherent pre-B cells on [0184] day 10 post infection. The data shown are derived from three independent experiments plated in triplicate. As expected, BCR-ABL caused pre-B cell transformation. The JBD of JIP-1 caused marked inhibition of transformation when present in the sense, but not the anti-sense, orientation. In some cultures infected with BCR-ABL and JBD, pre-B cell outgrowths were detected after 3-4 weeks, but these clones demonstrated no expression of JBD by protein immunoblot analysis. Since JIP-1 inhibits JNK signaling, these results indicate that the JNK pathway is required for pre-B cell transformation by BCR-ABL.
  • The demonstration that JNK is involved in both apoptosis and oncogenic transformation indicates that the biological actions of the JNK signal transduction pathway depend on the specific cellular context. The integration of JNK with other signal transduction pathways may be an important determinant of the functional consequences of JNK activation. The ability of JIP-1, an inhibitor of the JNK signal transduction pathway, to block both transformation and apoptosis is consistent with this hypothesis. [0185]
  • The physiological function of JIP-1 may be to suppress signal transduction by the JNK pathway. For example, JIP-1 may compete with substrates that bind JNK. Alternatively, JIP-1 may have a more direct role in targeting JNK to specific regions of the cell or to specific substrates. Since JIP-1 causes redistribution of JNK within the cell, JIP-1 may function as a cytoplasmic anchor for JNK. The tethering of JNK in the cytoplasm by interactions with JIP-1 provides a mechanism for controlling signal transduction by the JNK pathway, and the related phenomena of apoptosis and transformation. [0186]
  • This assay can be used to determine if specific polypeptides have the same effect on cellular transformation as full length, wildtype JIP-1. [0187]
  • Example 10 Screening for Peptides with JIP-1 Activity
  • Peptides suspected of having JIP-1 activity can be tested in the JNK binding assay described supra. Peptides are synthesized by methods that are well known to those skilled in the art; for example, using an Applied Biosystems synthesizer. Cell lysates containing Flag epitope-tagged JNK are incubated with GST-JIP-1 (residues 127-281 bound to glutathione-Sepharose, either with or without synthetic peptides (0, 4, 8, 16, 32 or 64 μg/ml), for one hour at 4° C. The beads are washed in TLB, and the bound proteins are detected by protein immunoblot analysis with a Flag-specific antibody, e.g., M2. Synthetic peptides with JIP-1 activity are those which inhibit the interaction of JIP-1 with JNK, as detected in this assay. These synthetic peptides should possess at least 60% of the binding activity of JIP-1. [0188]
  • Example 11 Therapeutic Applications
  • JIP-1 is shown herein to be capable of inhibiting apoptosis and transformation. Compositions containing JIP-1 polypeptides or nucleic acids can be administered to treat conditions characterized by these phenomena. Nucleic acids can be administered by, methods described in, e.g., Ausubel, et al., supra. A standard dosage would be from 1 to 1000 μg/kg of body weight. Polypeptides, peptides and peptide mimetics of the invention can be formulated according to procedures which are well known to those skilled in the art. A standard dosage of polypeptide also would be from 1 to 1000, e.g., 10 to 500, or 20 to 200 μg/kg of body weight. [0189]
  • Animal models for testing the effect of JIP-1 therapeutic compositions include the bcr-abl leukemia model. Daley et al., [0190] Science, 247:824-830 (1990). Other animal models that can be used include the myc/ras transformation model. Sinn et al., Cell, 49:465-475 (1987). Animal models for testing the effect of JIP-1 therapeutic compositions on conditions associated with apoptosis include a model of excitotoxic stress in the hippocampus, and a model of E2F-1 induced apoptosis. Ben-Ari et al., Neuroscience, 14:375-403 (1985); Field et al., Cell, 85:549-561 (1996). Other conditions that can be treated with JIP-1 compositions include liver damage (Mendelson et al., Proc. Natl. Acad. Sci. USA, 93:12908-12913 (1996)); kidney disease and organ transplantation (DeMari et al., Am. J. Physiol., 272:F292-F298 (1997)); and heart disease (Pombo et al., J. Biol. Chem., 269:26546-26551 (1994); Force et al., Circ. Res., 78:947-953 (1996).
  • Example 12 Diagnostic Applications
  • The polypeptides and antibodies of the invention can be used to detect or monitor JIP-1 expression. Levels of JIP-1 protein in a sample can be assayed by any standard technique. For example, JIP-1 protein expression can be monitored by standard immunological or immunohistochemical procedures using the antibodies described herein. See, e.g., Ausubel et al., supra; Bancroft et al., [0191] Theory and Practice of Histological Techniques, Churchill Livingstone (1982). Alternatively, JIP-1 expression can be assayed by standard Northern blot analysis, or can be aided by PCR. See Ausubel, supra; Ehrlich, ed., PCR Technology: Principles and Applications for DNA Amplification, Stockton Press, New York. Point mutations in the JIP-1 sequence can be detected using well known nucleic acid mismatch detection techniques. Lower than normal levels of JIP-1 would have the effect of altering apoptosis, while higher than normal levels would cause immune suppression, altered inflammatory responses, and alterations in tumor growth.
  • Other Embodiments
  • The invention also includes naturally occurring and non-naturally-occurring allelic variants of JIP-1. Compared to the most common naturally occurring nucleotide sequence encoding JIP-1, the nucleic acid sequence encoding allelic variants may have a substitution, deletion, or addiction of one or more nucleotides. The preferred allelic variants are functionally equivalent to naturally occurring JIP-1. [0192]
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, that the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages and modifications are within the scope of the following claims. [0193]
  • 1 30 660 amino acids amino acid linear protein internal JIP-1 protein 1 Met Ala Glu Arg Glu Ser Gly Leu Gly Gly Gly Ala Ala Ser Pro Pro 1 5 10 15 Ala Ala Ser Pro Phe Leu Gly Leu His Ile Ala Ser Pro Pro Asn Phe 20 25 30 Arg Leu Thr His Asp Ile Ser Leu Glu Glu Phe Glu Asp Glu Asp Leu 35 40 45 Ser Glu Ile Thr Asp Glu Cys Gly Ile Ser Leu Gln Cys Lys Asp Thr 50 55 60 Leu Ser Leu Arg Pro Pro Arg Ala Gly Leu Leu Ser Ala Gly Ser Ser 65 70 75 80 Gly Ser Ala Gly Ser Arg Leu Gln Ala Glu Met Leu Gln Met Asp Leu 85 90 95 Ile Asp Ala Ala Gly Asp Thr Pro Gly Ala Glu Asp Asp Glu Glu Glu 100 105 110 Glu Asp Asp Glu Leu Ala Ala Gln Arg Pro Gly Val Gly Pro Pro Lys 115 120 125 Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln Gly Pro 130 135 140 Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr Leu 145 150 155 160 Asn Leu Phe Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn Asn Asn 165 170 175 Ser Leu Gly Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg Ser Ser 180 185 190 Ser Pro Leu Lys Thr Gly Glu Gln Thr Pro Pro His Glu His Ile Cys 195 200 205 Leu Ser Asp Glu Leu Pro Pro Gln Gly Ser Pro Val Pro Thr Gln Asp 210 215 220 Arg Gly Thr Ser Thr Asp Ser Pro Cys Arg Arg Ser Ala Ala Thr Gln 225 230 235 240 Met Ala Pro Pro Ser Gly Pro Pro Ala Thr Ala Pro Gly Gly Arg Gly 245 250 255 His Ser His Arg Asp Arg Ile His Tyr Gln Ala Asp Val Arg Leu Glu 260 265 270 Ala Thr Glu Glu Ile Tyr Leu Thr Pro Val Gln Arg Pro Pro Asp Pro 275 280 285 Ala Glu Pro Thr Ser Thr Phe Met Pro Pro Thr Glu Ser Arg Met Ser 290 295 300 Val Ser Ser Asp Pro Asp Pro Ala Ala Tyr Ser Val Thr Ala Gly Arg 305 310 315 320 Pro His Pro Ser Ile Ser Glu Glu Asp Glu Gly Phe Asp Cys Leu Ser 325 330 335 Ser Pro Glu Arg Ala Glu Pro Pro Gly Gly Gly Trp Arg Gly Ser Leu 340 345 350 Gly Glu Pro Pro Pro Pro Pro Arg Ala Ser Leu Ser Ser Asp Thr Ser 355 360 365 Ala Leu Ser Tyr Asp Ser Val Lys Tyr Thr Leu Val Val Asp Glu His 370 375 380 Ala Gln Leu Glu Leu Val Ser Leu Arg Pro Cys Phe Gly Asp Tyr Ser 385 390 395 400 Asp Glu Ser Asp Ser Ala Thr Val Tyr Asp Asn Cys Ala Ser Ala Ser 405 410 415 Ser Pro Tyr Glu Ser Ala Ile Gly Glu Glu Tyr Glu Glu Ala Pro Gln 420 425 430 Pro Arg Pro Pro Thr Cys Leu Ser Glu Asp Ser Thr Pro Asp Glu Pro 435 440 445 Asp Val His Phe Ser Lys Lys Phe Leu Asn Val Phe Met Ser Gly Arg 450 455 460 Ser Arg Ser Ser Ser Ala Glu Ser Phe Gly Leu Phe Ser Cys Val Ile 465 470 475 480 Asn Gly Glu Glu His Glu Gln Thr His Arg Ala Ile Phe Arg Phe Val 485 490 495 Pro Arg His Glu Asp Glu Leu Glu Leu Glu Val Asp Asp Pro Leu Leu 500 505 510 Val Glu Leu Gln Ala Glu Asp Tyr Trp Tyr Glu Ala Tyr Asn Met Arg 515 520 525 Thr Gly Ala Arg Gly Val Phe Pro Ala Tyr Tyr Ala Ile Glu Val Thr 530 535 540 Lys Glu Pro Glu His Met Ala Ala Leu Ala Lys Asn Ser Cys Val Leu 545 550 555 560 Glu Ile Ser Val Arg Gly Val Lys Ile Gly Val Lys Ala Asp Asp Ala 565 570 575 Leu Glu Ala Lys Gly Asn Lys Cys Ser His Phe Phe Gln Leu Lys Asn 580 585 590 Ile Ser Phe Cys Gly Tyr His Pro Lys Asn Asn Lys Tyr Phe Gly Phe 595 600 605 Ile Thr Lys His Pro Ala Asp His Arg Phe Ala Cys His Val Phe Val 610 615 620 Ser Glu Asp Ser Thr Lys Ala Leu Ala Glu Ser Val Gly Arg Ala Phe 625 630 635 640 Gln Gln Phe Tyr Lys Gln Phe Val Glu Tyr Thr Cys Pro Thr Glu Asp 645 650 655 Ile Tyr Leu Glu 660 2832 base pairs nucleic acid double linear cDNA Coding Sequence 180...2159 JIP-1 cDNA 2 CTCGAGGTCG ACGGTATCGA TAAGCTTGAT ATCGCTGTCC GGAGCCGCGC CCGCCCCAGC 60 TCAGTCCGAA CCCCGCGGCG GCGGCGGCCT CCTCCACGCG CCTCCGCTGC TGCCGCCGCC 120 GCCGCCGCCG CCGCCTCCCG CGCCGCTCTC CGCCCGGATG GCCAGGGCTG CACCCCGGA 179 ATG GCG GAG CGA GAG AGC GGC CTG GGC GGG GGC GCC GCG TCC CCA CCG 227 Met Ala Glu Arg Glu Ser Gly Leu Gly Gly Gly Ala Ala Ser Pro Pro 1 5 10 15 GCC GCT TCC CCA TTC CTG GGA CTG CAC ATC GCG TCG CCT CCC AAT TTC 275 Ala Ala Ser Pro Phe Leu Gly Leu His Ile Ala Ser Pro Pro Asn Phe 20 25 30 AGG CTC ACC CAT GAC ATC AGC CTG GAG GAG TTT GAG GAT GAA GAC CTT 323 Arg Leu Thr His Asp Ile Ser Leu Glu Glu Phe Glu Asp Glu Asp Leu 35 40 45 TCG GAG ATC ACT GAC GAG TGT GGC ATC AGC CTG CAG TGC AAA GAC ACC 371 Ser Glu Ile Thr Asp Glu Cys Gly Ile Ser Leu Gln Cys Lys Asp Thr 50 55 60 CTG TCT CTC CGG CCC CCG CGC GCC GGG CTG CTG TCT GCG GGT AGC AGC 419 Leu Ser Leu Arg Pro Pro Arg Ala Gly Leu Leu Ser Ala Gly Ser Ser 65 70 75 80 GGC AGC GCG GGG AGC CGG CTG CAG GCG GAG ATG CTG CAG ATG GAC CTG 467 Gly Ser Ala Gly Ser Arg Leu Gln Ala Glu Met Leu Gln Met Asp Leu 85 90 95 GTC GAC GCG GCA GGT GAC ACT CCG GGC GCC GAG GAC GAC GAG GAG GAG 515 Ile Asp Ala Ala Gly Asp Thr Pro Gly Ala Glu Asp Asp Glu Glu Glu 100 105 110 GAG GAC GAC GAG CTC GCT GCC CAA CGA CCA GGA GTG GGG CCT CCC AAA 563 Glu Asp Asp Glu Leu Ala Ala Gln Arg Pro Gly Val Gly Pro Pro Lys 115 120 125 GCG GAG TCC AAC CAG GAT CCG GCG CCT CGC AGC CAG GGC CAG GGC CCG 611 Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln Gly Pro 130 135 140 GGC ACA GGC AGC GGA GAC ACC TAC CGA CCC AAG AGG CCT ACC ACG CTC 659 Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr Leu 145 150 155 160 AAC CTT TTC CCG CAG GTG CCG CGG TCT CAG GAC ACG CTG AAT AAT AAC 707 Asn Leu Phe Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn Asn Asn 165 170 175 TCT TTA GGC AAA AAG CAC AGT TGG CAG GAC CGT GTG TCT CGA TCA TCC 755 Ser Leu Gly Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg Ser Ser 180 185 190 TCC CCT CTG AAG ACA GGA GAA CAG ACG CCT CCA CAT GAA CAC ATC TGC 803 Ser Pro Leu Lys Thr Gly Glu Gln Thr Pro Pro His Glu His Ile Cys 195 200 205 CTG AGT GAT GAG CTG CCA CCC CAG GGC AGT CCT GTT CCC ACC CAG GAC 851 Leu Ser Asp Glu Leu Pro Pro Gln Gly Ser Pro Val Pro Thr Gln Asp 210 215 220 CGC GGC ACT TCC ACC GAC AGC CCT TGT CGC CGA AGT GCA GCC ACC CAG 899 Arg Gly Thr Ser Thr Asp Ser Pro Cys Arg Arg Ser Ala Ala Thr Gln 225 230 235 240 ATG GCA CCT CCA AGC GGT CCC CCT GCC ACT GCT CCT GGT GGC CGG GGC 947 Met Ala Pro Pro Ser Gly Pro Pro Ala Thr Ala Pro Gly Gly Arg Gly 245 250 255 CAC TCC CAT CGA GAC CGA ATC CAC TAC CAG GCA GAT GTG CGG CTC GAG 995 His Ser His Arg Asp Arg Ile His Tyr Gln Ala Asp Val Arg Leu Glu 260 265 270 GCG ACT GAG GAG ATC TAC CTG ACC CCA GTG CAG AGG CCC CCA GAC CCT 1043 Ala Thr Glu Glu Ile Tyr Leu Thr Pro Val Gln Arg Pro Pro Asp Pro 275 280 285 GCA GAA CCC ACC TCC ACC TTC ATG CCA CCC ACG GAG AGC CGG ATG TCA 1091 Ala Glu Pro Thr Ser Thr Phe Met Pro Pro Thr Glu Ser Arg Met Ser 290 295 300 GTT AGC TCG GAT CCA GAC CCT GCC GCT TAC TCT GTA ACT GCG GGG CGG 1139 Val Ser Ser Asp Pro Asp Pro Ala Ala Tyr Ser Val Thr Ala Gly Arg 305 310 315 320 CCA CAC CCC TCC ATC AGT GAA GAG GAT GAG GGC TTC GAC TGC CTG TCA 1187 Pro His Pro Ser Ile Ser Glu Glu Asp Glu Gly Phe Asp Cys Leu Ser 325 330 335 TCC CCA GAG CGA GCT GAG CCA CCA GGT GGA GGG TGG CGG GGA AGC CTC 1235 Ser Pro Glu Arg Ala Glu Pro Pro Gly Gly Gly Trp Arg Gly Ser Leu 340 345 350 GGG GAG CCA CCA CCG CCT CCA CGG GCC TCA CTG AGC TCG GAC ACC AGC 1283 Gly Glu Pro Pro Pro Pro Pro Arg Ala Ser Leu Ser Ser Asp Thr Ser 355 360 365 GCA CTG TCC TAC GAC TCG GTC AAG TAC ACA CTG GTG GTG GAT GAA CAT 1331 Ala Leu Ser Tyr Asp Ser Val Lys Tyr Thr Leu Val Val Asp Glu His 370 375 380 GCC CAG CTT GAG TTG GTG AGC CTG CGG CCG TGC TTT GGA GAT TAC AGT 1379 Ala Gln Leu Glu Leu Val Ser Leu Arg Pro Cys Phe Gly Asp Tyr Ser 385 390 395 400 GAC GAA AGC GAC TCT GCC ACT GTC TAT GAC AAC TGT GCC TCT GCC TCC 1427 Asp Glu Ser Asp Ser Ala Thr Val Tyr Asp Asn Cys Ala Ser Ala Ser 405 410 415 TCG CCC TAC GAG TCA GCC ATT GGT GAG GAG TAT GAG GAG GCC CCT CAG 1475 Ser Pro Tyr Glu Ser Ala Ile Gly Glu Glu Tyr Glu Glu Ala Pro Gln 420 425 430 CCC CGG CCT CCC ACC TGC CTC TCA GAG GAC TCC ACC CCG GAT GAG CCT 1523 Pro Arg Pro Pro Thr Cys Leu Ser Glu Asp Ser Thr Pro Asp Glu Pro 435 440 445 GAT GTC CAC TTC TCT AAG AAG TTT CTG AAT GTC TTC ATG AGT GGC CGC 1571 Asp Val His Phe Ser Lys Lys Phe Leu Asn Val Phe Met Ser Gly Arg 450 455 460 TCT CGT TCC TCC AGT GCT GAG TCC TTT GGG CTG TTC TCC TGC GTC ATC 1619 Ser Arg Ser Ser Ser Ala Glu Ser Phe Gly Leu Phe Ser Cys Val Ile 465 470 475 480 AAT GGG GAG GAG CAT GAG CAA ACC CAT CGG GCT ATA TTC AGG TTT GTG 1667 Asn Gly Glu Glu His Glu Gln Thr His Arg Ala Ile Phe Arg Phe Val 485 490 495 CCT CGG CAT GAA GAT GAA CTT GAG CTG GAA GTG GAT GAC CCC CTG CTG 1715 Pro Arg His Glu Asp Glu Leu Glu Leu Glu Val Asp Asp Pro Leu Leu 500 505 510 GTG GAG CTG CAG GCA GAA GAC TAT TGG TAT GAG GCC TAT AAC ATG CGC 1763 Val Glu Leu Gln Ala Glu Asp Tyr Trp Tyr Glu Ala Tyr Asn Met Arg 515 520 525 ACC GGA GCC CGC GGG GTC TTC CCT GCC TAC TAT GCC ATT GAG GTC ACC 1811 Thr Gly Ala Arg Gly Val Phe Pro Ala Tyr Tyr Ala Ile Glu Val Thr 530 535 540 AAG GAG CCT GAG CAC ATG GCA GCC CTT GCC AAA AAC AGC TGT GTC CTT 1859 Lys Glu Pro Glu His Met Ala Ala Leu Ala Lys Asn Ser Cys Val Leu 545 550 555 560 GAG ATC AGT GTC AGG GGT GTC AAG ATA GGC GTC AAA GCT GAT GAT GCT 1907 Glu Ile Ser Val Arg Gly Val Lys Ile Gly Val Lys Ala Asp Asp Ala 565 570 575 CTG GAG GCC AAG GGA AAT AAA TGT AGC CAC TTC TTC CAG CTA AAG AAC 1955 Leu Glu Ala Lys Gly Asn Lys Cys Ser His Phe Phe Gln Leu Lys Asn 580 585 590 ATC TCT TTC TGT GGA TAC CAT CCA AAG AAT AAC AAG TAC TTT GGG TTT 2003 Ile Ser Phe Cys Gly Tyr His Pro Lys Asn Asn Lys Tyr Phe Gly Phe 595 600 605 ATC ACT AAG CAC CCT GCT GAC CAC CGG TTT GCC TGC CAT GTC TTT GTG 2051 Ile Thr Lys His Pro Ala Asp His Arg Phe Ala Cys His Val Phe Val 610 615 620 TCT GAA GAT TCC ACC AAA GCC CTG GCG GAG TCT GTG GGG CGT GCA TTT 2099 Ser Glu Asp Ser Thr Lys Ala Leu Ala Glu Ser Val Gly Arg Ala Phe 625 630 635 640 CAG CAG TTC TAC AAG CAG TTT GTG GAG TAT ACC TGT CCT ACA GAA GAT 2147 Gln Gln Phe Tyr Lys Gln Phe Val Glu Tyr Thr Cys Pro Thr Glu Asp 645 650 655 ATC TAC TTG GAG TAGCAGCACC CCCACTCTCT GCAGCCCCTC AGCCCCAAGC 2199 Ile Tyr Leu Glu 660 CAGTGCAAGG ACAGCTGGCT GCTGACAGGA TGTGGTACTG CCACAAAAGA ATGGGGGAAT 2259 GAGGGCTGTT GGGTCGGGGG GGCCGGGGTT TGGGGAGAGG CAGATGCAGT TTATTGTAAT 2319 ATATGGGGTT AGATTAATCT ATGGAGGACA GTACAGGCTC TCTGGGGACT GGGGAAGGGT 2379 GGGGCTGGGG GGTGGGGGGT CAGGCCCCTG GCCACAGAGG GACTCCCTAG GAACAGAGGC 2439 ACTGTCCCAT CCTGGGCCTG TTTCATGCTA GGGGCCCTGG CTTTCTGGCT CTTGGCTCCT 2499 GCCTTGACAA AGCCCATGCC ACCTGGAAGT GTCCAGCTTC CCTTGTCCCC ACCTTGACCG 2559 GAGCCCTGAG CTCAGGCTGA GCCCACGCAC CTCCAAAGGA CTTTCCAGTA AGGAAATGGC 2619 AACGTGTGAC CGTGGAGACC CTGTTCTCAT CTGTGGGGCC TCTGGGCAGC TCCAACCTCC 2679 AGCCTGGCTA GCACACAGGT CCTCGCAAGG TTGTGTGTGC AAGGAGAGGG CCACAGTAAG 2739 CCCCATCTGC CAGGAAAAGG AGGCCTCTTA GCTGGCCCCA GCCAGCCGGT CACTGTCTTG 2799 TCACCTGGCT ACTATTAAAG TGCCATCTCG TGC 2832 27 amino acids amino acid linear peptide JIP-1 protein, amino acids 148-174 3 Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr Leu Asn Leu Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 155 amino acids amino acid linear protein JIP-1 protein, amino acids 127-281 4 Pro Lys Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln 1 5 10 15 Gly Pro Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr 20 25 30 Thr Leu Asn Leu Phe Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 35 40 45 Asn Asn Ser Leu Gly Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg 50 55 60 Ser Ser Ser Pro Leu Lys Thr Gly Glu Gln Thr Pro Pro His Glu His 65 70 75 80 Ile Cys Leu Ser Asp Glu Leu Pro Pro Gln Gly Ser Pro Val Pro Thr 85 90 95 Gln Asp Arg Gly Thr Ser Thr Asp Ser Pro Cys Arg Arg Ser Ala Ala 100 105 110 Thr Gln Met Ala Pro Pro Ser Gly Pro Pro Ala Thr Ala Pro Gly Gly 115 120 125 Arg Gly His Ser His Arg Asp Arg Ile His Tyr Gln Ala Asp Val Arg 130 135 140 Leu Glu Ala Thr Glu Glu Ile Tyr Leu Thr Pro 145 150 155 27 amino acids amino acid linear peptide c-Jun JNK-binding domain 5 Tyr Ser Asn Pro Lys Ile Leu Lys Gln Ser Met Thr Leu Asn Leu Ala 1 5 10 15 Asp Pro Val Gly Ser Leu Lys Pro His Leu Arg 20 25 27 amino acids amino acid linear peptide ATF-2 JNK-binding domain 6 His Leu Ala Val His Lys His Lys His Glu Met Thr Leu Lys Phe Gly 1 5 10 15 Pro Ala Arg Asn Asp Ser Val Ile Val Ala Asp 20 25 27 amino acids amino acid linear peptide JIP-1 mutant (159-162) G 7 Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Gly Gly Gly Gly Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 27 amino acids amino acid linear peptide JIP-1 mutant T159G 8 Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Gly Leu Asn Leu Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 27 amino acids amino acid linear peptide JIP-1 mutant L160G 9 Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr Gly Asn Leu Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 27 amino acids amino acid linear peptide JIP-1 mutant L162G 10 Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr Leu Asn Gly Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 27 amino acids amino acid linear peptide JIP-1 mutant K155G 11 Ser Gly Asp Thr Tyr Arg Pro Gly Arg Pro Thr Thr Leu Asn Leu Phe 1 5 10 15 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 20 25 27 amino acids amino acid linear peptide control peptide 12 Asn Ser Leu Gly Thr Asp Asp Thr Gln Tyr Ser Arg Arg Pro Pro Lys 1 5 10 15 Val Arg Gln Pro Asn Pro Thr Phe Thr Leu Leu 20 25 50 amino acids amino acid linear peptide JIP-1 amino acids 491-540 (SH3 domain) 13 Ala Ile Phe Arg Glu Val Pro Arg His Glu Asp Glu Leu Glu Leu Glu 1 5 10 15 Val Asp Asp Pro Leu Leu Val Glu Leu Gln Ala Glu Asp Tyr Trp Tyr 20 25 30 Glu Ala Tyr Asn Met Arg Thr Gly Ala Arg Gly Val Phe Pro Ala Tyr 35 40 45 Tyr Ala 50 8 amino acids amino acid linear peptide Flag eptiope 14 Asp Tyr Lys Asp Asp Asp Asp Lys 1 5 76 amino acids amino acid linear peptide JIP-1 amino acids 127-202 15 Pro Lys Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln 1 5 10 15 Gly Pro Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr 20 25 30 Thr Leu Asn Leu Phe Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn 35 40 45 Asn Asn Ser Leu Gly Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg 50 55 60 Ser Ser Ser Pro Leu Lys Thr Gly Glu Gln Thr Pro 65 70 75 79 amino acids amino acid linear peptide JIP-1 amino acids 203-281 16 Pro His Glu His Ile Cys Leu Ser Asp Glu Leu Pro Pro Gln Gly Ser 1 5 10 15 Pro Val Pro Thr Gln Asp Arg Gly Thr Ser Thr Asp Ser Pro Cys Arg 20 25 30 Arg Ser Ala Ala Thr Gln Met Ala Pro Pro Ser Gly Pro Pro Ala Thr 35 40 45 Ala Pro Gly Gly Arg Gly His Ser His Arg Asp Arg Ile His Tyr Gln 50 55 60 Ala Asp Val Arg Leu Glu Ala Thr Glu Glu Ile Tyr Leu Thr Pro 65 70 75 77 amino acids amino acid linear peptide JIP-1 amino acids 164-240 17 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn Asn Asn Ser Leu Gly 1 5 10 15 Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg Ser Ser Ser Pro Leu 20 25 30 Lys Thr Gly Glu Gln Thr Pro Pro His Glu His Ile Cys Leu Ser Asp 35 40 45 Glu Leu Pro Pro Gln Gly Ser Pro Val Pro Thr Gln Asp Arg Gly Thr 50 55 60 Ser Thr Asp Ser Pro Cys Arg Arg Ser Ala Ala Thr Gln 65 70 75 68 amino acids amino acid linear peptide JIP-1 amino acids 135-202 18 Pro Ala Pro Arg Ser Gln Gly Gln Gly Pro Gly Thr Gly Ser Gly Asp 1 5 10 15 Thr Tyr Arg Pro Lys Arg Pro Thr Thr Leu Asn Leu Phe Pro Gln Val 20 25 30 Pro Arg Ser Gln Asp Thr Leu Asn Asn Asn Ser Leu Gly Lys Lys His 35 40 45 Ser Trp Gln Asp Arg Val Ser Arg Ser Ser Ser Pro Leu Lys Thr Gly 50 55 60 Glu Gln Thr Pro 65 59 amino acids amino acid linear peptide JIP-1 amino acids 144-202 19 Pro Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr Thr 1 5 10 15 Leu Asn Leu Phe Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn Asn 20 25 30 Asn Ser Leu Gly Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg Ser 35 40 45 Ser Ser Pro Leu Lys Thr Gly Glu Gln Thr Pro 50 55 49 amino acids amino acid linear peptide JIP-1 amino acids 154-202 20 Pro Lys Arg Pro Thr Thr Leu Asn Leu Phe Pro Gln Val Pro Arg Ser 1 5 10 15 Gln Asp Thr Leu Asn Asn Asn Ser Leu Gly Lys Lys His Ser Trp Gln 20 25 30 Asp Arg Val Ser Arg Ser Ser Ser Pro Leu Lys Thr Gly Glu Gln Thr 35 40 45 Pro 39 amino acids amino acid linear peptide JIP-1 amino acids 164-202 21 Pro Gln Val Pro Arg Ser Gln Asp Thr Leu Asn Asn Asn Ser Leu Gly 1 5 10 15 Lys Lys His Ser Trp Gln Asp Arg Val Ser Arg Ser Ser Ser Pro Leu 20 25 30 Lys Thr Gly Glu Gln Thr Pro 35 17 amino acids amino acid linear peptide JIP-1 amino acids 127-143 22 Pro Lys Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln 1 5 10 15 Gly 27 amino acids amino acid linear peptide JIP-1 amino acids 127-153 23 Pro Lys Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln 1 5 10 15 Gly Pro Gly Thr Gly Ser Gly Asp Thr Tyr Arg 20 25 37 amino acids amino acid linear peptide JIP-1 amino acids 127-163 24 Pro Lys Ala Glu Ser Asn Gln Asp Pro Ala Pro Arg Ser Gln Gly Gln 1 5 10 15 Gly Pro Gly Thr Gly Ser Gly Asp Thr Tyr Arg Pro Lys Arg Pro Thr 20 25 30 Thr Leu Asn Leu Phe 35 20 base pairs nucleic acid single linear DNA 6...6 where R at position 6 is A or G 25 GARGARTTYG ARGAYGARGA 20 20 base pairs nucleic acid single linear DNA 6...6 where R at position 6 is A or G 26 GGNAARAARC AYAGNTGGCA 20 21 base pairs nucleic acid single linear DNA 7...7 where W at position 7 is A or T 27 CATRTTWTAN GCYTCWTACC A 21 23 base pairs nucleic acid single linear DNA 6...6 where Y at position 6 is C or T 28 AAYTGYTTKT ARAAYTGYTG RAA 23 33 base pairs nucleic acid single linear RNA 29 GGUAUCGAUA AGCUUGAUAU CGCUGUCCGG AGC 33 33 base pairs nucleic acid single linear RNA 30 AGAGGCACUG UCCCAUCCUG GGCCUGUUUC AUG 33

Claims (27)

What is claimed is:
1. A substantially pure JNK-interacting protein 1 (JIP-1) polypeptide or a biologically active fragment thereof.
2. The polypeptide of claim 1, wherein said polypeptide is modified by attachment of a hydrophobic moiety, to facilitate uptake of said polypeptide by cells.
3. The polypeptide of claim 2, wherein said polypeptide is modified by attachment of a peptide that facilitates uptake of said polypeptide by cells.
4. The polypeptide of claim 1, wherein said polypeptide comprises an amino acid sequence corresponding to the JNK-binding domain (JBD; SEQ ID NO:4) of wild type JIP-1.
5. The polypeptide of claim 1, said polypeptide comprising an amino acid sequence substantially identical to SEQ ID NO:1.
6. The polypeptide of claim 1, said polypeptide comprising an amino acid sequence substantially identical to a sequence selected from the group consisting SEQ ID NO:3; SEQ ID NO:4; SEQ ID NO:13; SEQ ID NO:15; SEQ ID NO:16; SEQ ID NO:17; SEQ ID NO:18; SEQ ID NO:19; SEQ ID NO:20; SEQ ID NO:21; SEQ ID NO:22; SEQ ID NO:22; SEQ ID NO:23; and SEQ ID NO:24.
7. The polypeptide of claim 1, wherein said polypeptide is of mammalian origin.
8. The polypeptide of claim 1, wherein said polypeptide is of human origin.
9. Isolated nucleic acid comprising a sequence encoding the polypeptide of claim 1, or its complement.
10. The nucleic acid of claim 9, wherein said nucleic acid comprises a sequence encoding a JNK-binding domain.
11. The nucleic acid of claim 9, said nucleic acid comprising a nucleotide sequence substantially identical to SEQ ID NO:12 or its complement.
12. The nucleic acid of claim 9, said nucleic acid encoding the amino acid sequence of SEQ ID NO:1.
13. A genetically engineered host cell comprising the nucleic acid of claim 9.
14. An expression vector comprising the nucleic acid of claim 9, operably linked to a nucleotide sequence regulatory element that controls expression of the nucleotide sequence in a host cell.
15. The nucleic acid of claim 9, wherein said nucleic acid is of mammalian origin.
16. The nucleic acid of claim 9, wherein said nucleic acid is of human origin.
17. A method of treating a patient having a pathological condition associated with abnormal expression or activity of JNK, or at risk of developing a pathological condition associated with abnormal activity or expression of JNK, said method comprising administering to the patient a therapeutically effective amount of a JIP-1 nucleic acid.
18. The method of claim 17, wherein said pathological condition is a neurodegenerative disease.
19. The method of claim 17, wherein said neurodegenerative disease is selected from the group consisting of Parkinson's disease and Alzheimer's disease.
20. The method of claim 17, wherein said pathological condition is a blood clot.
21. The method of claim 17, wherein said condition is stroke.
22. The method of claim 17, wherein said pathological condition is malignancy.
23. The method of claim 17, wherein said pathological condition is leukemia.
24. The method of claim 23, wherein said leukemia is chronic myelogenous leukemia.
25. The method of claim 17, wherein said pathological condition is an autoimmune disease.
26. The method of claim 17, wherein said pathological condition is inflammation.
27. A method of treating a patient having a pathological condition associated with abnormal expression or activity of JNK, or at risk of developing a pathological condition associated with abnormal activity or expression of JNK, said method comprising administering to the patient a therapeutically effective amount of a JIP-1 polypeptide.
US10/115,178 1997-04-28 2002-04-02 Inhibitors of the JNK signal transduction pathway and methods of use Abandoned US20020119135A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US10/115,178 US20020119135A1 (en) 1997-04-28 2002-04-02 Inhibitors of the JNK signal transduction pathway and methods of use
US11/474,513 US7838294B2 (en) 1997-04-28 2006-06-23 Inhibitors of the JNK signal transduction pathway and methods of use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/819,177 US6043083A (en) 1997-04-28 1997-04-28 Inhibitors of the JNK signal transduction pathway and methods of use
US09/419,587 US6410693B1 (en) 1997-04-28 1999-10-18 Inhibitors of the JNK signal transduction pathway and methods of use
US10/115,178 US20020119135A1 (en) 1997-04-28 2002-04-02 Inhibitors of the JNK signal transduction pathway and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/419,587 Continuation US6410693B1 (en) 1997-04-28 1999-10-18 Inhibitors of the JNK signal transduction pathway and methods of use

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/474,513 Continuation US7838294B2 (en) 1997-04-28 2006-06-23 Inhibitors of the JNK signal transduction pathway and methods of use

Publications (1)

Publication Number Publication Date
US20020119135A1 true US20020119135A1 (en) 2002-08-29

Family

ID=25227408

Family Applications (4)

Application Number Title Priority Date Filing Date
US08/819,177 Expired - Lifetime US6043083A (en) 1997-04-28 1997-04-28 Inhibitors of the JNK signal transduction pathway and methods of use
US09/419,587 Expired - Lifetime US6410693B1 (en) 1997-04-28 1999-10-18 Inhibitors of the JNK signal transduction pathway and methods of use
US10/115,178 Abandoned US20020119135A1 (en) 1997-04-28 2002-04-02 Inhibitors of the JNK signal transduction pathway and methods of use
US11/474,513 Expired - Fee Related US7838294B2 (en) 1997-04-28 2006-06-23 Inhibitors of the JNK signal transduction pathway and methods of use

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US08/819,177 Expired - Lifetime US6043083A (en) 1997-04-28 1997-04-28 Inhibitors of the JNK signal transduction pathway and methods of use
US09/419,587 Expired - Lifetime US6410693B1 (en) 1997-04-28 1999-10-18 Inhibitors of the JNK signal transduction pathway and methods of use

Family Applications After (1)

Application Number Title Priority Date Filing Date
US11/474,513 Expired - Fee Related US7838294B2 (en) 1997-04-28 2006-06-23 Inhibitors of the JNK signal transduction pathway and methods of use

Country Status (4)

Country Link
US (4) US6043083A (en)
EP (1) EP1017710A4 (en)
AU (1) AU7167298A (en)
WO (1) WO1998049188A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008118626A2 (en) * 2007-03-08 2008-10-02 Burnham Institute For Medical Research Inhibitors of jnk and methods for identifying inhibitors of jnk
WO2010151638A1 (en) 2009-06-25 2010-12-29 Medical College Of Georgia Research Institute, Inc. Jnk inhibitors for use in treating spinal muscular atrophy
US8501812B2 (en) 2005-03-29 2013-08-06 University Of Massachusetts Therapeutic methods for type I diabetes
WO2016100909A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College METHODS FOR GENERATING STEM CELL-DERIVED β CELLS AND USES THEREOF

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6294350B1 (en) * 1997-06-05 2001-09-25 Dalhousie University Methods for treating fibroproliferative diseases
US6943000B2 (en) 1997-10-03 2005-09-13 University Of Massachusetts JNK3 modulators and methods of use
EP1087230A1 (en) * 1999-09-24 2001-03-28 Boehringer Ingelheim International GmbH Method for identifying compounds useful in the therapy of bone disorders
EP1088822A1 (en) 1999-09-28 2001-04-04 Applied Research Systems ARS Holding N.V. Pharmaceutically active sulfonyl hydrazide derivatives
EP1088815A1 (en) 1999-09-28 2001-04-04 Applied Research Systems ARS Holding N.V. Pharmaceutically active sulfonyl amino acid derivatives
EP1088821A1 (en) 1999-09-28 2001-04-04 Applied Research Systems ARS Holding N.V. Pharmaceutically active sulfonamide derivatives
US8183339B1 (en) 1999-10-12 2012-05-22 Xigen S.A. Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US20040082509A1 (en) * 1999-10-12 2004-04-29 Christophe Bonny Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US6610820B1 (en) * 1999-10-12 2003-08-26 University Of Lausanne Cell-permeable peptide inhibitors of the JNK signal transduction pathway
US20030108539A1 (en) * 2000-02-14 2003-06-12 Christophe Bonny Cell-permeable peptide inhibitors of the JNK signal transduction pathway
EP1110957A1 (en) 1999-12-24 2001-06-27 Applied Research Systems ARS Holding N.V. Benzazole derivatives and their use as JNK modulators
US6428967B1 (en) * 2000-05-01 2002-08-06 Board Of Regents, The University Of Texas System LDL receptor signaling pathways
US6346416B1 (en) * 2000-08-29 2002-02-12 Isis Pharmaceuticals, Inc. Antisense inhibition of HPK/GCK-like kinase expression
EP1193268A1 (en) 2000-09-27 2002-04-03 Applied Research Systems ARS Holding N.V. Pharmaceutically active sulfonamide derivatives bearing both lipophilic and ionisable moieties as inhibitors of protein Junkinases
EP1193256A1 (en) * 2000-09-27 2002-04-03 Applied Research Systems ARS Holding N.V. Pharmaceutically active benzsulfonamide derivatives as inhibitors of JNK proteins
US7122658B1 (en) * 2000-11-22 2006-10-17 Pioneer Hi-Bred International, Inc. Seed-preferred regulatory elements and uses thereof
AUPR279101A0 (en) * 2001-01-30 2001-02-22 Cytopia Pty Ltd Protein kinase signalling
US7232897B2 (en) * 2001-04-24 2007-06-19 Harvard University, President And Fellows Of Harvard College Compositions and methods for modulating NH2-terminal Jun Kinase activity
US6696453B2 (en) 2001-05-21 2004-02-24 Alcon, Inc. Use of NF-κB inhibitors to treat dry eye disorders
US7803749B2 (en) 2002-01-09 2010-09-28 Xigen Sa Peptide inhibitors of MKK7 kinase binding to insulin binding proteins
US7003536B2 (en) * 2002-08-15 2006-02-21 Comsys Communications & Signal Processing Ltd. Reduced complexity fast hadamard transform
AU2003278727A1 (en) * 2002-09-20 2004-04-08 Alcon, Inc. Use of cytokine synthesis inhibitors for the treatment of dry eye disorders
WO2004094591A2 (en) * 2003-04-16 2004-11-04 The Regents Of The University Of Colorado Three-dimensional structures of hdac9 and cabin1 and compound structures and methods related thereto
WO2007031098A1 (en) * 2005-09-12 2007-03-22 Xigen S.A. Cell-permeable peptide inhibitors of the jnk signal transduction pathway
US8080517B2 (en) * 2005-09-12 2011-12-20 Xigen Sa Cell-permeable peptide inhibitors of the JNK signal transduction pathway
WO2008025564A2 (en) * 2006-08-31 2008-03-06 Nexigen Gmbh Methods for detecting protein-peptide interactions
US9061009B2 (en) * 2007-06-08 2015-06-23 University Of Massachusetts Mixed lineage kinases and metabolic disorders
WO2009143865A1 (en) * 2008-05-30 2009-12-03 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
WO2009143864A1 (en) * 2008-05-30 2009-12-03 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of chronic or non-chronic inflammatory digestive diseases
US20100183633A1 (en) * 2008-12-04 2010-07-22 University Of Massachusetts Interleukin 6 and tumor necrosis factor alpha as biomarkers of jnk inhibition
WO2010072228A1 (en) 2008-12-22 2010-07-01 Xigen S.A. Novel transporter constructs and transporter cargo conjugate molecules
JP5457813B2 (en) * 2009-12-16 2014-04-02 ルネサスエレクトロニクス株式会社 ADPLL circuit, semiconductor device and portable information device
WO2011160653A1 (en) 2010-06-21 2011-12-29 Xigen S.A. Novel jnk inhibitor molecules
CA2807036C (en) 2010-10-14 2018-01-16 Xigen S.A. Use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of chronic or non-chronic inflammatory eye diseases
WO2013091670A1 (en) 2011-12-21 2013-06-27 Xigen S.A. Novel jnk inhibitor molecules for treatment of various diseases
WO2014206426A1 (en) 2013-06-26 2014-12-31 Xigen Inflammation Ltd. New use for jnk inhibitor molecules for treatment of various diseases
WO2014206427A1 (en) 2013-06-26 2014-12-31 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
CA2903275A1 (en) * 2013-06-26 2014-12-31 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
WO2016207413A1 (en) * 2015-06-26 2016-12-29 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of mild cognitive impairment
WO2015197097A1 (en) 2014-06-26 2015-12-30 Xigen Inflammation Ltd. New use for jnk inhibitor molecules for treatment of various diseases
WO2016055160A2 (en) * 2014-10-08 2016-04-14 Xigen Inflammation Ltd. New use of cell-permeable peptide inhibitors of the jnk signal transduction pathway for the treatment of various diseases
ES2949982T3 (en) * 2014-06-26 2023-10-04 Xigen Inflammation Ltd Cell-Permeable Peptide Inhibitors of the JNK Signal Transduction Pathway for the Treatment of Cystitis

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5534426A (en) * 1993-07-19 1996-07-09 The Regents Of The University Of California Oncoprotein protein kinase
US5880261A (en) * 1997-04-03 1999-03-09 Waeber; Gerard Transcription factor Islet-Brain 1 (IB1)
US6610820B1 (en) 1999-10-12 2003-08-26 University Of Lausanne Cell-permeable peptide inhibitors of the JNK signal transduction pathway

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8501812B2 (en) 2005-03-29 2013-08-06 University Of Massachusetts Therapeutic methods for type I diabetes
WO2008118626A2 (en) * 2007-03-08 2008-10-02 Burnham Institute For Medical Research Inhibitors of jnk and methods for identifying inhibitors of jnk
WO2008118626A3 (en) * 2007-03-08 2009-05-07 Burnham Inst Medical Research Inhibitors of jnk and methods for identifying inhibitors of jnk
WO2010151638A1 (en) 2009-06-25 2010-12-29 Medical College Of Georgia Research Institute, Inc. Jnk inhibitors for use in treating spinal muscular atrophy
WO2016100909A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College METHODS FOR GENERATING STEM CELL-DERIVED β CELLS AND USES THEREOF

Also Published As

Publication number Publication date
EP1017710A4 (en) 2003-01-02
WO1998049188A1 (en) 1998-11-05
US7838294B2 (en) 2010-11-23
AU7167298A (en) 1998-11-24
EP1017710A1 (en) 2000-07-12
US20070003517A1 (en) 2007-01-04
US6410693B1 (en) 2002-06-25
US6043083A (en) 2000-03-28

Similar Documents

Publication Publication Date Title
US7838294B2 (en) Inhibitors of the JNK signal transduction pathway and methods of use
US6627405B1 (en) 53BP2 complexes
WO1999015556A1 (en) Huchordin and uses thereof
US6395548B1 (en) Methods of modulating of angiogenesis
US7585497B2 (en) Induction of apoptosis and cell growth inhibition by protein 4.33
US6607880B1 (en) Methods and reagents for modulating apoptosis
US6670450B1 (en) Protein and gene involved in myocyte differentiation
US20030022837A1 (en) Modulation of cell division by an early mitotic inhibitor protein
US6558918B1 (en) Nucleic acid that encode a cell growth regulatory protein
AU735512B2 (en) Modulators of BRAC1 activity
EP1450852B1 (en) Ptp10d nucleic acids and peptides involved in the regulation of energy homeostasis
EP1226279B1 (en) Modulation of neuroendocrine differentiation by protein 25.1
AU784443B2 (en) Novel cell cycle checkpoint genes and proteins encoded thereby
EP1373500B1 (en) Gene involved in v(d)j recombination and/or dna repair
WO1998046641A9 (en) Novel polypeptides within the growth factor superfamily
US6372467B1 (en) P54s6k and p85s6k genes, proteins, primers, probes, and detection methods
US20020127594A1 (en) Don-1 gene and polypeptides and uses therefor
US6680167B2 (en) Molecules of the card-related protein family and uses thereof
KR20020069140A (en) A Novel TRAF6 Inhibiting Protein
KR100721484B1 (en) Nucleic acid, polypeptides and antibodies relating to human suppressor of fused
US20050119206A1 (en) Cg8327, cg10823, cg18418, cg15862, cg3768, cg11447 and cg16750 homologous proteins involved in the regulation of energy homeostasis
CA2331152A1 (en) Cell cycle regulating factor
WO1998008861A1 (en) Rp compositions and therapeutic and diagnostic uses therefor
AU1355501A (en) Modulation of neuroendocrine differentiation by protein 25.1
JP2000083678A (en) Human proteinkinase hoacf72

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION