WO2000017236A2 - Apoptosis related protein and uses therefor - Google Patents

Apoptosis related protein and uses therefor Download PDF

Info

Publication number
WO2000017236A2
WO2000017236A2 PCT/US1999/022270 US9922270W WO0017236A2 WO 2000017236 A2 WO2000017236 A2 WO 2000017236A2 US 9922270 W US9922270 W US 9922270W WO 0017236 A2 WO0017236 A2 WO 0017236A2
Authority
WO
WIPO (PCT)
Prior art keywords
arp
nucleic acid
polypeptide
protein
seq
Prior art date
Application number
PCT/US1999/022270
Other languages
French (fr)
Other versions
WO2000017236A8 (en
WO2000017236A3 (en
Inventor
Mehran M. Khodadoust
Original Assignee
Millennium Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Millennium Pharmaceuticals, Inc. filed Critical Millennium Pharmaceuticals, Inc.
Priority to AU65005/99A priority Critical patent/AU6500599A/en
Publication of WO2000017236A2 publication Critical patent/WO2000017236A2/en
Publication of WO2000017236A3 publication Critical patent/WO2000017236A3/en
Publication of WO2000017236A8 publication Critical patent/WO2000017236A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides 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
    • C07K14/4747Apoptosis related proteins

Definitions

  • programmed cell death occurs in both vertebrate and invertebrate species and is characterized by unique morphological alterations, such as cytoplasmic contraction and chromatin condensation, as well as by specific DNA cleavage into oligonucleosomal fragments. Unlike necrosis, programmed cell death or apoptosis is an irreversible process which in most systems appears to depend on the expression of a specific set of novel "death genes". Deregulation of this process contributes to the pathogenesis of several diseases including cancer, immunodeficiency,autoimmune diseases, and neurodegenerative disorders (Thompson C.B. et al. (1995) Science 267: 1456).
  • SARP Secreted Apoptosis-Related Proteins
  • the present invention is based, at least in part, on the discovery of novel apoptosis related protein (ARP) family members, referred to herein as "ARP" nucleic acid and protein molecules.
  • ARP apoptosis related protein
  • the ARP molecules of the present invention are useful as modulating agents for regulating a variety of cellular processes. Accordingly, in one aspect, this invention provides isolated nucleic acid molecules encoding ARP proteins or biologically active portions thereof, as well as nucleic acid fragments suitable as primers or hybridization probes for the detection of ARP-encoding nucleic acids.
  • an ARP nucleic acid molecule of the invention is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or more homologous to the nucleotide sequence (e.g., to the entire length of the nucleotide sequence) shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a complement thereof.
  • the isolated nucleic acid molecule includes the nucleotide sequence shown SEQ ID NO: 1 or 3, or a complement thereof.
  • the nucleic acid molecule includes SEQ ID NO: 3 and nucleotides 1-73 of SEQ ID NO: 1.
  • the nucleic acid molecule includes SEQ ID NO:3 and nucleotides 1133-1380 of SEQ ID NO:l.
  • the nucleic acid molecule consists of the nucleotide sequence shown in SEQ ID NO:l or 3.
  • the nucleic acid molecule includes a fragment of at least 308 nucleotides of the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or a complement thereof.
  • an ARP nucleic acid molecule includes a nucleotide sequence encoding a protein having an amino acid sequence sufficiently homologous to the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the
  • an ARP nucleic acid molecule includes a nucleotide sequence encoding a protein having an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%>, 80%), 85%», 90%), 95%>, 98% or more homologous to the amino acid sequence of SEQ ID NO:2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • an isolated nucleic acid molecule encodes the amino acid sequence of human ARP.
  • the nucleic acid molecule includes a nucleotide sequence encoding a protein having the amino acid sequence of SEQ ID NO: 2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • the nucleic acid molecule is at least 308 nucleotides in length.
  • the nucleic acid molecule is at least 308 nucleotides in length and encodes a protein having an ARP activity (as described herein).
  • nucleic acid molecules preferably ARP nucleic acid molecules, which specifically detect ARP nucleic acid molecules relative to nucleic acid molecules encoding non- ARP proteins.
  • a nucleic acid molecule is at least 308, 308-350, 350-400, 400-450, 450-500 or more nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence shown in SEQ ID NO:l, the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • the nucleic acid molecules are at least 15 (e.g., contiguous) nucleotides in length and hybridize under stringent conditions to nucleotides 117-139 of SEQ ID NO : 1. In other preferred embodiments, the nucleic acid molecules comprise nucleotides 117-139 of SEQ ID NO: 1.
  • the nucleic acid molecule encodes a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number , wherein the nucleic acid molecule hybridizes to a nucleic acid molecule comprising SEQ ID NO:l or SEQ ID NO: 3 under stringent conditions.
  • Another embodiment of the invention provides an isolated nucleic acid molecule which is antisense to an ARP nucleic acid molecule, e.g., the coding strand of an ARP nucleic acid molecule.
  • Another aspect of the invention provides a vector comprising an ARP nucleic acid molecule.
  • the vector is a recombinant expression vector.
  • the invention provides a host cell containing a vector of the invention.
  • the invention also provides a method for producing a protein, preferably an ARP protein, by culturing in a suitable medium, a host cell, e.g., a mammalian host cell such as a non-human mammalian cell, of the invention containing a recombinant expression vector, such that the protein is produced.
  • the isolated protein preferably an ARP protein
  • the isolated protein includes at least one cysteine rich domain.
  • the isolated protein, preferably an ARP protein includes at least one cysteine rich domain and at least one EGF-like domain.
  • the isolated protein, preferably an ARP protein includes at least one cysteine rich domain, at least one EGF- like domain, and at least one Laminin EGF-like domain.
  • the protein preferably an ARP protein, includes at least one cysteine rich domain and has an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%), 98%) or more homologous to the amino acid sequence of SEQ ID NO:2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • the protein preferably an ARP protein, includes at least one cysteine rich domain and plays a role in apoptosis or programmed cell death.
  • the protein preferably an ARP protein, includes at least one cysteine rich domain and is encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l or SEQ ID NO:3.
  • the invention features fragments of the protein having the amino acid sequence of SEQ ID NO:2, wherein the fragment comprises at least 15 amino acids (e.g., contiguous amino acids) of the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with the ATCC as Accession Number .
  • the protein preferably an amino acid sequence having the amino acid sequence having the amino acid sequence of SEQ ID NO:2, wherein the fragment comprises at least 15 amino acids (e.g., contiguous amino acids) of the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with the ATCC as Accession Number .
  • the protein preferably an amino acids (e.g., contiguous amino acids) of the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with the ATCC as Accession Number .
  • the protein preferably an amino acid sequence having the amino acid sequence having the amino acid
  • ARP protein has the amino acid sequence of SEQ ID NO:2.
  • the invention features an isolated protein, preferably an ARP protein, which is encoded by a nucleic acid molecule consisting of a nucleotide sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to a nucleotide sequence of SEQ ID NO: 1 , SEQ ID NO:3, or a complement thereof.
  • This invention further features an isolated protein, preferably an ARP protein, which is encoded by a nucleic acid molecule consisting of a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or a complement thereof.
  • the proteins of the present invention or portions thereof e.g., biologically active portions thereof, can be operatively linked to a non- ARP polypeptide (e.g., heterologous amino acid sequences) to form fusion proteins.
  • the invention further features antibodies, such as monoclonal or polyclonal antibodies, that specifically bind proteins of the invention, preferably ARP proteins.
  • the ARP proteins or biologically active portions thereof can be incorporated into pharmaceutical compositions, which optionally include pharmaceutically acceptable carriers.
  • the present invention provides a method for detecting the presence of an ARP nucleic acid molecule, protein or polypeptide in a biological sample by contacting the biological sample with an agent capable of detecting an ARP nucleic acid molecule, protein or polypeptide such that the presence of an ARP nucleic acid molecule, protein or polypeptide is detected in the biological sample.
  • the present invention provides a method for detecting the presence of ARP activity in a biological sample by contacting the biological sample with an agent capable of detecting an indicator of ARP activity such that the presence of ARP activity is detected in the biological sample.
  • the invention provides a method for modulating ARP activity comprising contacting a cell capable of expressing ARP with an agent that modulates ARP activity such that ARP activity in the cell is modulated.
  • the agent inhibits ARP activity.
  • the agent stimulates ARP activity.
  • the agent is an antibody that specifically binds to an ARP protein.
  • the agent modulates expression of ARP by modulating transcription of an ARP gene or translation of an ARP mRNA.
  • the agent is a nucleic acid molecule having a nucleotide sequence that is antisense to the coding strand of an ARP mRNA or an ARP gene.
  • the methods of the present invention are used to treat a subject having a disorder characterized by aberrant ARP protein or nucleic acid expression or activity by administering an agent which is an ARP modulator to the subject.
  • the ARP modulator is an ARP protein.
  • the ARP modulator is an ARP nucleic acid molecule.
  • the ARP modulator is a peptide, peptidomimetic, or other small molecule.
  • the disorder characterized by aberrant ARP protein or nucleic acid expression is a disorder characterized by deregulated programmed cell death.
  • the present invention also provides a diagnostic assay for identifying the presence or absence of a genetic alteration characterized by at least one of (i) aberrant modification or mutation of a gene encoding an ARP protein; (ii) mis-regulation of the gene; and (iii) aberrant post-translational modification of an ARP protein, wherein a wild-type form of the gene encodes a protein with an ARP activity.
  • the invention provides a method for identifying a compound that binds to or modulates the activity of an ARP protein, by providing an indicator composition comprising an ARP protein having ARP activity, contacting the indicator composition with a test compound, and determining the effect of the test compound on ARP activity in the indicator composition to identify a compound that modulates the activity of an ARP protein.
  • Figure 1 depicts the cDNA sequence and predicted amino acid sequence of human ARP.
  • the nucleotide sequence corresponds to nucleic acids 1 to 1380 of SEQ ID NO:l .
  • the amino acid sequence corresponds to amino acids 1 to 353 of SEQ ID NO:2.
  • the coding region without the 5' and 3' untranslated regions of the human ARP gene is shown in SEQ ID NO:3.
  • Figure 2 depicts a structural, hydrophobicity, and antigenicity analysis of the human ARP protein.
  • Figure 3 depicts the results of a search which was performed against the HMM database and which resulted in the identification of an EGF-like domain in the human ARP protein.
  • Figure 4 depicts the results of a search which was performed against the HMM database and which resulted in the identification of a Laminin EGF-like domain in the human ARP protein.
  • Figure 5 depicts the results from a northern blot analysis.
  • ARP protein and nucleic acid molecules comprise a family of molecules having certain conserved structural and functional features.
  • family when referring to the protein and nucleic acid molecules of the invention is intended to mean two or more proteins or nucleic acid molecules having a common structural domain or motif and having sufficient amino acid or nucleotide sequence homology as defined herein.
  • family members can be naturally or non-naturally occurring and can be from either the same or different species.
  • a family can contain a first protein of human origin, as well as other, distinct proteins of human origin or alternatively, can contain homologues of non-human origin.
  • Members of a family may also have common functional characteristics.
  • cyste rich domain includes a protein domain having an amino acid sequence of at least about 20 amino acids of which at least about 2 amino acids are cysteine residues.
  • a cysteine rich domain includes at least about 30, more preferably at least about 35-40 amino acid residues, of which at least about 2, preferably at least about 3, more preferably at least about 4, 5 or 6 amino acids are cysteine residues.
  • Cysteine-rich domains having lengths of 45-50 or 60 amino acid residues and having up to 7, 8, 9 or 10 cysteine residues are also within the scope of this invention. Cysteine rich domains are described in, for example, Lodish H. et al. Molecular Cell Biology, (Scientific American Books Inc., New York, N.Y., 1995), the contents of which are incorporated herein by reference.
  • an ARP of the present invention is identified based on the presence of an "EGF-like domain" in the protein or corresponding nucleic acid molecule.
  • the term "EGF-like domain” includes a protein domain having an amino acid sequence of about 55-90 amino acid residues and having a bit score for the alignment of the sequence to the EGF-like domain (HMM) of at least 6.
  • an EGF-like domain includes at least about 60-85, more preferably about 65- 80 amino acid residues, or about 70-79 amino acids and has a bit score for the alignment of the sequence to the EGF-like domain (HMM) of at least 7-10, more preferably 10-30, more preferably 30-50, even more preferably 50-75, 75-100, 100-200 or greater.
  • the EGF-like domain HMM has been assigned the PFAM Accession PF00008 (http://genome.wustl.edu/Pfam/WWWdata/EGF.html).
  • the amino acid sequence of the protein is searched against a database of HMMs (e.g., the Pfam database, release 2.1) using the default parameters
  • the hmmsf program which is available as part of the HMMER package of search programs, is a family specific default program for PF00435 and a score of 15 is the default threshold score for determining a hit.
  • the threshold score for determining a hit can be lowered (e.g., to 8 bits).
  • a description of the Pfam database can be found in Sonhammer et al. (1997) Proteins 28(3)405-420 and a detailed description of HMMs can be found, for example, in Gribskov et ⁇ /.(1990) Meth.
  • an ARP of the present invention is identified based on the presence of a "Laminin EGF-like domain" in the protein or corresponding nucleic acid molecule.
  • the term "Laminin EGF-like domain” includes a protein domain having an amino acid sequence of about 30-90 amino acid residues and having a bit score for the alignment of the sequence to the Laminin EGF-like domain (HMM) of at least 6.
  • the Laminin EGF-like domain includes about 40-80, more preferably about 40-60 amino acid residues, or about 40-50 amino acid residues and has a bit score for the alignment of the sequence to the Laminin EGF-like domain (HMM) of at least 7-10, more preferably 10-30, more preferably 30-50, even more preferably 50- 75, 75-100, 100-200 or greater.
  • the Laminin EGF-like domain HMM has been assigned the PFAM Accession PF00053 (http://genome.wustl.edu/Pfam/WWWdata/EGF.html).
  • the results of the search are set forth in Figure 4. Accordingly, ARP proteins having at least 50-60% homology, preferably about 60-70%), more preferably about 70-80%, or about 80-90%) homology with a Laminin EGF-like domain of human ARP are within the scope of the invention.
  • the ARP protein of the present invention are believed to play a role in apoptosis or programmed cell death.
  • programmed cell death includes a genetically regulated process involved in the normal development of multicellular organisms. This process occurs in cells destined for removal in a variety of normal situations, including larval development of the nematode c. elegans, insect metamorphosis, development in mammalian embryos including the nephrogenic zone in the developing kidney, and regression or atrophy (e.g., in the prostrate after castration).
  • Programmed cell death can occur in many cells following the withdrawal of growth and trophic factors, or as a result of nutritional deprivation, hormone treatment, ultraviolet irradiation, and exposure to toxic and infectious agents including reactive oxygen species and phosphatase inhibitors, e.g., okadaic acid, calcium ionphones, and a number of cancer chemotherapeutic agents.
  • toxic and infectious agents including reactive oxygen species and phosphatase inhibitors, e.g., okadaic acid, calcium ionphones, and a number of cancer chemotherapeutic agents.
  • the ARP proteins by participating in a programmed cell death pathway, can modulate a programmed cell death pathway activity and provide novel diagnostic targets and therapeutic agents for disorders characterized by deregulated programmed cell death, particularly in cells that express ARP, e.g., the heart.
  • a "disorder characterized by deregulated programmed cell death” includes a disorder, disease or condition which is characterized by a deregulation, e.g., an upregulation or a downregulation, of programmed cell death.
  • a deregulation e.g., an upregulation or a downregulation
  • disorders characterized by deregulated programmed cell death include profilerative disorders, e.g., cancer such as chronic lymphocytic leukemia or colorectal cancer; and neurodegenerative disorders, e.g., Alzheimer's disease, dementias related to Alzheimer's disease (such as Pick's disease), Parkinson's and other Lewy diffuse body diseases, multiple sclerosis, amyotrophic lateral sclerosis, progressive supranuclear palsy, epilepsy, Jakob-Creutzfieldt disease, or AIDS related dementias.
  • profilerative disorders e.g., cancer such as chronic lymphocytic leukemia or colorectal cancer
  • neurodegenerative disorders e.g., Alzheimer's disease, dementias related to Alzheimer's disease (such as Pick's disease), Parkinson's and other Lewy diffuse body diseases, multiple sclerosis, amyotrophic lateral sclerosis, progressive supranuclear palsy, epilepsy, Jakob-Creutzfieldt disease, or
  • Isolated proteins of the present invention preferably ARP proteins, have an amino acid sequence sufficiently homologous to the amino acid sequence of SEQ ID NO: 2 or are encoded by a nucleotide sequence sufficiently homologous to SEQ ID NO:l or SEQ ID NO:3.
  • the term "sufficiently homologous” refers to a first amino acid or nucleotide sequence which contains a sufficient or minimum number of identical or equivalent (e.g., an amino acid residue which has a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences share common structural domains or motifs and/or a common functional activity.
  • amino acid or nucleotide sequences which share common structural domains have at least 30%, 40%, or 50% homology, preferably 60%) homology, more preferably 70%-80%>, and even more preferably 90-95% homology across the amino acid sequences of the domains and contain at least one and preferably two structural domains or motifs, are defined herein as sufficiently homologous.
  • amino acid or nucleotide sequences which share at least 30%, 40%, or 50%, preferably 60%, more preferably 70-80%, or 90-95% homology and share a common functional activity are defined herein as sufficiently homologous.
  • an "ARP activity”, “biological activity of ARP” or “functional activity of ARP”, refers to an activity exerted by an ARP protein, polypeptide or nucleic acid molecule on an ARP responsive cell or on an ARP protein substrate, as determined in vivo, or in vitro, according to standard techniques.
  • an ARP activity is a direct activity, such as an association with an ARP-out molecule.
  • a "target molecule” or “binding partner” is a molecule with which an ARP protein binds or interacts in nature, such that ARP -mediated function is achieved.
  • An ARP target molecule can be a non- ARP molecule or an ARP protein or polypeptide of the present invention.
  • an ARP target molecule is an ARP ligand.
  • an ARP activity is an indirect activity, such as a cellular signaling activity mediated by interaction of the ARP protein with an ARP ligand.
  • ARP proteins and polypeptides having an ARP activity are isolated ARP proteins and polypeptides having an ARP activity.
  • Preferred proteins are ARP proteins having at least one cysteine rich domain and, preferably, an ARP activity.
  • Other preferred proteins are ARP proteins having at least one cysteine rich domain, an EGF- like domain and, preferably, an ARP activity.
  • Other preferred proteins are ARP proteins having at least one cysteine rich domain, an EGF-like domain, a Laminin EGF-like domain and, preferably, an ARP activity.
  • Additional preferred proteins have at least one cysteine rich domain, an EGF-like domain, a Laminin EGF-like domain and are, preferably, encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l or SEQ ID NO:3.
  • the nucleotide sequence of the isolated human ARP cDNA and the predicted amino acid sequence of the human ARP polypeptide are shown in Figure 1 and in SEQ ID NOs:l and 2, respectively.
  • a plasmid containing the nucleotide sequence encoding human ARP was deposited with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, VA 20110-2209, on and assigned Accession Number . This deposit will be maintained under the terms of the Budapest
  • the human ARP gene which is approximately 1380 nucleotides in length, encodes a protein having a molecular weight of approximately 40.5 kD and which is approximately 353 amino acid residues in length.
  • the large isoform of the human ARP gene is expressed in all tisues tested.
  • the small isoform of the human ARP gene is expressed mainly in the heart, the skeletal muscle, the placenta, and the pancreas.
  • nucleic acid molecules that encode ARP proteins or biologically active portions thereof, as well as nucleic acid fragments sufficient for use as hybridization probes to identify ARP-encoding nucleic acid molecules (e.g., ARP mRNA) and fragments for use as PCR primers for the amplification or mutation of ARP nucleic acid molecules.
  • ARP-encoding nucleic acid molecules e.g., ARP mRNA
  • fragments for use as PCR primers for the amplification or mutation of ARP nucleic acid molecules.
  • nucleic acid molecule is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs.
  • the nucleic acid molecule can be single-stranded or double- stranded, but preferably is double-stranded DNA.
  • isolated nucleic acid molecule includes nucleic acid molecules which are separated from other nucleic acid molecules which are present in the natural source of the nucleic acid.
  • isolated includes nucleic acid molecules which are separated from the chromosome with which the genomic DNA is naturally associated.
  • an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived.
  • the isolated nucleic acid molecule can contain less than about 5 kb, 4kb, 3kb, 2kb, 1 kb, 0.5 kb or 0.1 kb of nucleotides which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived.
  • an "isolated" nucleic acid molecule such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized.
  • a nucleic acid molecule of the present invention e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein.
  • ARP nucleic acid molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989).
  • nucleic acid molecule encompassing all or a portion of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number can be isolated by the polymerase chain reaction (PCR) using synthetic oligonucleotide primers designed based upon the sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • PCR polymerase chain reaction
  • a nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques.
  • the nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • oligonucleotides corresponding to ARP nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer.
  • an isolated nucleic acid molecule of the invention comprises the nucleotide sequence shown in SEQ ID NO: 1.
  • the sequence of SEQ ID NO:l corresponds to the human ARP cDNA.
  • This cDNA comprises sequences encoding the human ARP protein (i.e., "the coding region", from nucleotides 74-1132), as well as 5' untranslated sequences (nucleotides 1-73) and 3' untranslated sequences (nucleotides 1133-1380).
  • the nucleic acid molecule can comprise only the coding region of SEQ ID NO:l (e.g., nucleotides 74-1132, corresponding to SEQ ID NO:3).
  • an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the
  • DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion of any of these nucleotide sequences.
  • Accession Number is one which is sufficiently complementary to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , such that it can hybridize to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • an isolated nucleic acid molecule of the present invention comprises a nucleotide sequence which is at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to the entire length of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the entire length of the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion of any of these nucleotide sequences.
  • the nucleic acid molecule of the invention can comprise only a portion of the nucleic acid sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , for example, a fragment which can be used as a probe or primer or a fragment encoding a portion of an ARP protein, e.g., a biologically active portion of an ARP protein.
  • the nucleotide sequence determined from the cloning of the ARP gene allows for the generation of probes and primers designed for use in identifying and/or cloning other ARP family members, as well as ARP homologues from other species.
  • the probe/primer typically comprises substantially purified oligonucleotide.
  • the oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12 or 15, preferably about 20 or 25, more preferably about 30, 35, 40, 45, 50, 55, 60, 65, or 75 consecutive nucleotides of a sense sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , of an anti-sense sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or of a naturally occurring allelic variant or mutant of SEQ ID NO: 1 , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • a nucleic acid molecule of the present invention comprises a nucleotide sequence which is greater than 308, 308-350, 350-400, 400-450, 450-500, 500-550, 550-600, 600-650, 650-700, 700-750, 750-800, 800-850, 850-900, 900-950, 950-1000, or more nucleotides in length and hybridizes under stringent hybridization conditions to a nucleic acid molecule of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • Probes based on the ARP nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins.
  • the probe further comprises a label group attached thereto, e.g., the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co- factor.
  • Such probes can be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress an ARP protein, such as by measuring a level of an ARP- encoding nucleic acid in a sample of cells from a subject e.g., detecting ARP mRNA levels or determining whether a genomic ARP gene has been mutated or deleted.
  • a nucleic acid fragment encoding a "biologically active portion of an ARP protein” can be prepared by isolating a portion of the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , which encodes a polypeptide having an ARP biological activity (the biological activities of the ARP proteins are described herein), expressing the encoded portion of the ARP protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of the ARP protein.
  • the invention further encompasses nucleic acid molecules that differ from the nucleotide sequence shown in SEQ ID NO: 1 , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , due to degeneracy of the genetic code and thus encode the same ARP proteins as those encoded by the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NO:2.
  • DNA sequence polymo ⁇ hisms that lead to changes in the amino acid sequences of the ARP proteins may exist within a population (e.g., the human population).
  • Such genetic polymo ⁇ hism in the ARP genes may exist among individuals within a population due to natural allelic variation.
  • gene and “recombinant gene” refer to nucleic acid molecules which include an open reading frame encoding an ARP protein, preferably a mammalian ARP protein, and can further include non-coding regulatory sequences, and introns.
  • Allelic variants of human ARP include both functional and non-functional ARP proteins.
  • Functional allelic variants are naturally occurring amino acid sequence variants of the human ARP protein that maintain the ability to bind an ARP ligand and/or modulate programmed cell death.
  • Functional allelic variants will typically contain only conservative substitution of one or more amino acids of SEQ ID NO:2 or substitution, deletion or insertion of non-critical residues in non-critical regions of the protein.
  • Non-functional allelic variants are naturally occurring amino acid sequence variants of the human ARP protein that do not have the ability to either bind an ARP ligand and/or modulate programmed cell death. Non-functional allelic variants will typically contain a non-conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of SEQ ID NO:2 or a substitution, insertion or deletion in critical residues or critical regions.
  • the present invention further provides non-human orthologues of the human ARP protein. Orthologues of the human ARP protein are proteins that are isolated from non-human organisms and possess the same ARP ligand binding and/or modulation of programmed cell death capabilities of the human ARP protein. Orthologues of the human ARP protein can readily be identified as including an amino acid sequence that is substantially homologous to SEQ ID NO:2, as defined herein.
  • nucleic acid molecules encoding other ARP family members and, thus, which have a nucleotide sequence which differs from the ARP sequences of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number are intended to be within the scope of the invention.
  • another ARP cDNA can be identified based on the nucleotide sequence of human ARP.
  • nucleic acid molecules encoding ARP proteins from different species and which, thus, have a nucleotide sequence which differs from the ARP sequences of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number are intended to be within the scope of the invention.
  • a mouse ARP cDNA can be identified based on the nucleotide sequence of a human ARP.
  • Nucleic acid molecules corresponding to natural allelic variants and homologues of the ARP cDNAs of the invention can be isolated based on their homology to the ARP nucleic acids disclosed herein using the cDNAs disclosed herein, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. Nucleic acid molecules corresponding to natural allelic variants and homologues of the ARP cDNAs of the invention can further be isolated by mapping to the same chromosome or locus as the ARP gene.
  • an isolated nucleic acid molecule of the invention is at least 15, 20, 25, 30 or more nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number .
  • the nucleic acid is at least 30, 50, 100, 150, 200, 250, 300, 308, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, or 950 nucleotides in length.
  • hybridizes under stringent conditions is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other.
  • the conditions are such that sequences at least about 70%, more preferably at least about 80%), even more preferably at least about 85%) or 90%o homologous to each other typically remain hybridized to each other.
  • stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • a preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45C, followed by one or more washes in 0.2 X SSC, 0.1% SDS at 50°C, preferably at 55°C, more preferably at 60°C, and even more preferably at 65 °C.
  • SSC 6X sodium chloride/sodium citrate
  • 0.1% SDS 0.1% SDS at 50°C, preferably at 55°C, more preferably at 60°C, and even more preferably at 65 °C.
  • an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NOT corresponds to a naturally-occurring nucleic acid molecule.
  • a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
  • allelic variants of the ARP sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
  • non-essential amino acid residue is a residue that can be altered from the wild-type sequence of ARP (e.g., the sequence of SEQ ID NO:2) without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity.
  • amino acid residues that are conserved among the ARP proteins of the present invention are predicted to be particularly unamenable to alteration.
  • additional amino acid residues that are conserved between the ARP proteins of the present invention and other members of the GPCR families are not likely to be amenable to alteration.
  • nucleic acid molecules encoding ARP proteins that contain changes in amino acid residues that are not essential for activity. Such ARP proteins differ in amino acid sequence from SEQ ID NO:2, yet retain biological activity.
  • the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to SEQ ID NO:2.
  • An isolated nucleic acid molecule encoding an ARP protein homologous to the protein of SEQ ID NO:2 can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. Mutations can be introduced into SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis.
  • conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues.
  • a "conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine
  • nonpolar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • a predicted nonessential amino acid residue in an ARP protein is preferably replaced with another amino acid residue from the same side chain family.
  • mutations can be introduced randomly along all or part of an ARP coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for ARP biological activity to identify mutants that retain activity.
  • SEQ ID NO SEQ ID NO:3
  • the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
  • a mutant ARP protein can be assayed for the ability to (1) interact with a non- ARP protein molecule; (2) activate an ARP-dependent signal transduction pathway; or (3) modulate programmed cell death.
  • another aspect of the invention pertains to isolated nucleic acid molecules which are antisense thereto.
  • An "antisense" nucleic acid comprises a nucleotide sequence which is complementary to a "sense" nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence.
  • an antisense nucleic acid can hydrogen bond to a sense nucleic acid.
  • the antisense nucleic acid can be complementary to an entire ARP coding strand, or to only a portion thereof.
  • an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding ARP.
  • the term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues (e.g., the coding region of human ARP corresponds to SEQ ID NO:3).
  • the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding ARP.
  • noncoding region refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions).
  • antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing.
  • the antisense nucleic acid molecule can be complementary to the entire coding region of ARP mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of ARP mRNA.
  • the antisense oligonucleotide can be complementary to the region surrounding the translation start site of ARP mRNA.
  • An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length.
  • an antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid e.g., an antisense oligonucleotide
  • an antisense nucleic acid can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5- carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1- methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta-D- mannosylqueosine, 5'-methoxy
  • the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
  • the antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an ARP protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation.
  • the hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix.
  • An example of a route of administration of antisense nucleic acid molecules of the invention include direct injection at a tissue site.
  • antisense nucleic acid molecules can be modified to target selected cells and then administered systemically.
  • antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens.
  • the antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
  • the antisense nucleic acid molecule of the invention is an ⁇ -anomeric nucleic acid molecule.
  • An ⁇ -anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual ⁇ -units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids. Res. 15:6625-6641).
  • the antisense nucleic acid molecule can also comprise a 2'-o- methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett. 215:327-330).
  • an antisense nucleic acid of the invention is a ribozyme.
  • Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region.
  • ribozymes e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave ARP mRNA transcripts to thereby inhibit translation of ARP mRNA.
  • a ribozyme having specificity for an ARP-encoding nucleic acid can be designed based upon the nucleotide sequence of an ARP cDNA disclosed herein (i.e., SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number ).
  • a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an ARP- encoding mRNA. See, e.g., Cech et al. U.S. Patent No.
  • ARP mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel, D. and Szostak, J.W. (1993) Science 261:1411-1418.
  • ARP gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the ARP (e.g., the ARP promoter and/or enhancers) to form triple helical structures that prevent transcription of the ARP gene in target cells.
  • nucleotide sequences complementary to the regulatory region of the ARP e.g., the ARP promoter and/or enhancers
  • ARP promoter and/or enhancers e.g., the ARP promoter and/or enhancers
  • the ARP nucleic acid molecules of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule.
  • the deoxyribose phosphate backbone of the nucleic acid molecules can be modified to generate peptide nucleic acids (see Hyrup B. et al. (1996) Bioorganic & Medicinal Chemistry 4 (1): 5-23).
  • peptide nucleic acids refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained.
  • the neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength.
  • the synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup B. et al. (1996) supra; Perry-O'Keefe et al. Proc. Natl. Acad. Sci. 93: 14670-675.
  • PNAs of ARP nucleic acid molecules can be used in therapeutic and diagnostic applications.
  • PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, for example, inducing transcription or translation arrest or inhibiting replication.
  • PNAs of ARP nucleic acid molecules can also be used in the analysis of single base pair mutations in a gene, (e.g., by PNA- directed PCR clamping); as 'artificial restriction enzymes' when used in combination with other enzymes, (e.g., SI nucleases (Hyrup B. (1996) supra)); or as probes or primers for DNA sequencing or hybridization (Hyrup B. et al. (1996) supra; Perry- O'Keefe supra).
  • PNAs of ARP can be modified, (e.g., to enhance their stability or cellular uptake), by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art.
  • PNA-DNA chimeras of ARP nucleic acid molecules can be generated which may combine the advantageous properties of PNA and DNA.
  • Such chimeras allow DNA recognition enzymes, (e.g., RNAse H and DNA polymerases), to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity.
  • PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup B. (1996) supra).
  • the synthesis of PNA-DNA chimeras can be performed as described in Hyrup B. (1996) supra and Finn P.J. et al. (1996) Nucleic Acids Res. 24 (17): 3357-63.
  • a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used as a between the PNA and the 5' end of DNA (Mag, M. et al. (1989) Nucleic Acid Res. 17: 5973-88). PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment (Finn P.J. et al. (1996) supra).
  • chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment (Peterser, K.H. et al. (1975) Bioorganic Med. Chem. Lett. 5: 1119-11124).
  • 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 (see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Acad. Sci. USA 84:648-652; PCT Publication No. W088/09810) or the blood-brain barrier (see, e.g., PCT Publication No. W089/10134).
  • peptides e.g., for targeting host cell receptors in vivo
  • agents facilitating transport across the cell membrane see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Aca
  • oligonucleotides can be modified with hybridization- triggered cleavage agents (See, e.g., Krol et al. (1988) Bio-Techniques 6:958-976) or intercalating agents. (See, e.g., Zon (1988) Pharm. Res. 5:539-549).
  • the oligonucleotide may be conjugated to another molecule, (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent).
  • ARP proteins and Anti-ARP Antibodies
  • ARP proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques.
  • ARP proteins are produced by recombinant DNA techniques.
  • Alternative to recombinant expression an ARP protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
  • an “isolated” or “purified” protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the ARP protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized.
  • the language “substantially free of cellular material” includes preparations of ARP protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • the language "substantially free of cellular material” includes preparations of ARP protein having less than about 30% (by dry weight) of non- ARP protein (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non- ARP protein, still more preferably less than about 10% of non- ARP protein, and most preferably less than about 5% non- ARP protein.
  • non- ARP protein also referred to herein as a "contaminating protein”
  • the ARP protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of ARP protein in which the protein is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein.
  • the language “substantially free of chemical precursors or other chemicals” includes preparations of ARP protein having less than about 30% (by dry weight) of chemical precursors or non- ARP chemicals, more preferably less than about 20% chemical precursors or non- ARP chemicals, still more preferably less than about 10% chemical precursors or non- ARP chemicals, and most preferably less than about 5% chemical precursors or non- ARP chemicals.
  • a "biologically active portion" of an ARP protein includes a fragment of an ARP protein which participates in an interaction between an ARP molecule and a non- ARP molecule.
  • Biologically active portions of an ARP protein include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequence of the ARP protein, e.g., the amino acid sequence shown in SEQ ID NO:2, which include less amino acids than the full length ARP proteins, and exhibit at least one activity of an ARP protein.
  • biologically active portions comprise a domain or motif with at least one activity of the ARP protein, e.g., modulating cellular programmed cell death.
  • a biologically active portion of an ARP protein can be a polypeptide which is, for example, 10, 25, 50, 100, 200 or more amino acids in length.
  • Biologically active portions of an ARP protein can be used as targets for developing agents which modulate an ARP mediated activity, e.g., programmed cell death.
  • a biologically active portion of an ARP protein comprises at least one cysteine rich domain, at least one EGF-like domain, or at least one Laminin EGF-like domain. It is to be understood that a preferred biologically active portion of an ARP protein of the present invention may contain at least one of the above-identified structural domains. A more preferred biologically active portion of an ARP protein may contain at least two of the above-identified structural domains. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native ARP protein.
  • the ARP protein has an amino acid sequence shown in SEQ ID NO:2.
  • the ARP protein is substantially homologous to SEQ ID NO:2, and retains the functional activity of the protein of SEQ ID NO:2, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail in subsection I above.
  • the ARP protein is a protein which comprises an amino acid sequence at least about 50%, 55%>, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to SEQ ID NO:2.
  • the sequences are aligned for optimal comparison pu ⁇ oses (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence and non-homologous sequences can be disregarded for comparison pu ⁇ oses).
  • the length of a reference sequence aligned for comparison pu ⁇ oses is at least 30%, preferably at least 40%, more preferably at least 50%>, even more preferably at least 60%, and even more preferably at least 70%, 80%, or 90% of the length of the reference sequence (e.g., when aligning a second sequence having 177 amino acid residues, to the ARP amino acid sequence of SEQ ID NO:2, at least 80, preferably at least 100, more preferably at least 120, even more preferably at least 140, and even more preferably at least 150, 160 or 170 amino acid residues are aligned).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid "homology” is equivalent to amino acid or nucleic acid "identity”).
  • the comparison of sequences and determination of percent homology between two sequences can be accomplished using a mathematical algorithim.
  • a preferred, non- limiting example of a mathematical algorithim utilized for the comparison of sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-68, modified as in Karlin and Altschul (1993) Proc. Natl Acad. Sci. USA 90:5873-77. Such an algorithm is inco ⁇ orated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J Mol. Biol 215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res.
  • the percent homology between two amino acid sequences can be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4.
  • the percent homology between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using a gap weight of 50 and a length weight of 3.
  • an ARP "chimeric protein” or “fusion protein” comprises an ARP polypeptide operatively linked to a non-ARP polypeptide.
  • An “ARP polypeptide” refers to a polypeptide having an amino acid sequence corresponding to ARP
  • a non-ARP polypeptide refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to the ARP protein, e.g., a protein which is different from the ARP protein and which is derived from the same or a different organism.
  • the ARP polypeptide can correspond to all or a portion of an ARP protein.
  • an ARP fusion protein comprises at least one biologically active portion of an ARP protein. In another preferred embodiment, an ARP fusion protein comprises at least two biologically active portions of an ARP protein.
  • the term "operatively linked" is intended to indicate that the ARP polypeptide and the non-ARP polypeptide are fused in-frame to each other. The non-ARP polypeptide can be fused to the N-terminus or C-terminus of the ARP polypeptide.
  • the fusion protein is a GST-ARP fusion protein in which the ARP sequences are fused to the C-terminus of the GST sequences.
  • Such fusion proteins can facilitate the purification of recombinant ARP.
  • the fusion protein is an ARP protein containing a heterologous signal sequence at its N-terminus.
  • ARP fusion proteins of the invention can be inco ⁇ orated into pharmaceutical compositions and administered to a subject in vivo.
  • the ARP fusion proteins can be used to affect the bioavailability of an ARP substrate.
  • ARP fusion proteins may be useful therapeutically for the treatment of disorders caused by, for example, (i) aberrant modification or mutation of a gene encoding an ARP protein; (ii) mis-regulation of the ARP gene; and (iii) aberrant post-translational modification of an ARP protein.
  • the ARP-fusion proteins of the invention can be used as immunogens to produce anti-ARP antibodies in a subject, to purify ARP ligands and in screening assays to identify molecules which inhibit the interaction of ARP with an ARP substrate.
  • an ARP chimeric or fusion protein of the invention is produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, for example by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992).
  • many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide).
  • An ARP- encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the ARP protein.
  • the present invention also pertains to variants of the ARP proteins which function as either ARP agonists (mimetics) or as ARP antagonists. Variants of the ARP proteins can be generated by mutagenesis, e.g., discrete point mutation or truncation of an ARP protein.
  • An agonist of the ARP proteins can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of an ARP protein.
  • An antagonist of an ARP protein can inhibit one or more of the activities of the naturally occurring form of the ARP protein by, for example, competitively modulating an ARP- mediated activity of an ARP protein.
  • specific biological effects can be elicited by treatment with a variant of limited function.
  • treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the ARP protein.
  • variants of an ARP protein which function as either ARP agonists (mimetics) or as ARP antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of an ARP protein for ARP protein agonist or antagonist activity.
  • a variegated library of ARP variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library.
  • a variegated library of ARP variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential ARP sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of ARP sequences therein.
  • a degenerate set of potential ARP sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of ARP sequences therein.
  • methods which can be used to produce libraries of potential ARP variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector.
  • degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential ARP sequences.
  • Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang, S.A. (1983) Tetrahedron 39:3; Itakura et al. ( ⁇ 9%4) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477.
  • libraries of fragments of an ARP protein coding sequence can be used to generate a variegated population of ARP fragments for screening and subsequent selection of variants of an ARP protein.
  • a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an ARP coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with SI nuclease, and ligating the resulting fragment library into an expression vector.
  • an expression library can be derived which encodes N-terminal, C-terminal and internal fragments of various sizes of the ARP protein.
  • Recrusive ensemble mutagenesis (REM), a new technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify ARP variants (Arkin and Yourvan (1992) Proc. Natl. Acad. Sci. USA 59:7811-7815; Delgrave et al. (1993) Protein Engineering 6(3):327-331).
  • cell based assays can be exploited to analyze a variegated ARP library.
  • a library of expression vectors can be transfected into a cell line which ordinarily responds to a particular ligand in an ARP-dependent manner.
  • the transfected cells are then contacted with the ligand and the effect of expression of the mutant on signaling by the ligand can be detected, e.g., by measuring cell survival or the activity of an ARP -regulated transcription factor.
  • Plasmid DNA can then be recovered from the cells which score for inhibition, or alternatively, potentiation of signaling by the ligand, and the individual clones further characterized.
  • An isolated ARP protein, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind ARP using standard techniques for polyclonal and monoclonal antibody preparation.
  • a full-length ARP protein can be used or, alternatively, the invention provides antigenic peptide fragments of ARP for use as immunogens.
  • the antigenic peptide of ARP comprises at least 8 amino acid residues of the amino acid sequence shown in SEQ ID NO:2 and encompasses an epitope of ARP such that an antibody raised against the peptide forms a specific immune complex with ARP.
  • the antigenic peptide comprises at least 10 amino acid residues, more preferably at least 15 amino acid residues, even more preferably at least 20 amino acid residues, and most preferably at least 30 amino acid residues.
  • Preferred epitopes encompassed by the antigenic peptide are regions of ARP that are located on the surface of the protein, e.g., hydrophilic regions, as well as regions with high antigenicity (see, for example, Figure 2).
  • An ARP immunogen typically is used to prepare antibodies by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal) with the immunogen.
  • An appropriate immunogenic preparation can contain, for example, recombinantly expressed ARP protein or a chemically synthesized ARP polypeptide.
  • the preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent. Immunization of a suitable subject with an immunogenic ARP preparation induces a polyclonal anti-ARP antibody response. Accordingly, another aspect of the invention pertains to anti-ARP antibodies.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site which specifically binds (immunoreacts with) an antigen, such as ARP.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin.
  • the invention provides polyclonal and monoclonal antibodies that bind ARP.
  • monoclonal antibody or “monoclonal antibody composition”, as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of ARP.
  • a monoclonal antibody composition thus typically displays a single binding affinity for a particular ARP protein with which it immunoreacts.
  • Polyclonal anti-ARP antibodies can be prepared as described above by immunizing a suitable subject with an ARP immunogen.
  • the anti-ARP antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized ARP.
  • ELISA enzyme linked immunosorbent assay
  • the antibody molecules directed against ARP can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) N ⁇ twre 256:495-497) (see also, Brown et al. (1981) J Immunol. 127:539-46; Brown et al. (1980) J Biol. Chem .255:4980-83; Yeh et al. (1976) Proc. Natl. Acad. Sci. USA 76:2927-31 ; and Yeh et al. (1982) Int. J.
  • an immortal cell line typically a myeloma
  • lymphocytes typically splenocytes
  • ARP immunogen as described above
  • the immortal cell line e.g., a myeloma cell line
  • the immortal cell line is derived from the same mammalian species as the lymphocytes.
  • murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line.
  • Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium").
  • HAT medium culture medium containing hypoxanthine, aminopterin and thymidine
  • Any of a number of myeloma cell lines can be used as a fusion partner according to standard techniques, e.g., the P3-NSl/l-Ag4-l, P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines. These myeloma lines are available from ATCC.
  • HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG").
  • PEG polyethylene glycol
  • Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed).
  • Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind ARP, e.g., using a standard ELISA assay.
  • a monoclonal anti-ARP antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with ARP to thereby isolate immunoglobulin library members that bind ARP.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01 ; and the Stratagene SurfZAPTM Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, Ladner et al. U.S.
  • recombinant anti-ARP antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant D ⁇ A techniques, are within the scope of the invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant D ⁇ A techniques known in the art, for example using methods described in Robinson et al. International Application No. PCT/US86/02269; Akira, et al. European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al. European Patent Application 173,494; Neuberger et al. PCT International Publication No.
  • An anti-ARP antibody (e.g., monoclonal antibody) can be used to isolate ARP by standard techniques, such as affinity chromatography or immunoprecipitation.
  • An anti- ARP antibody can facilitate the purification of natural ARP from cells and of recombinantly produced ARP expressed in host cells.
  • an anti-ARP antibody can be used to detect ARP protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the ARP protein.
  • Anti-ARP antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen.
  • Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance.
  • detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125 I,
  • vectors preferably expression vectors, containing a nucleic acid encoding an ARP protein (or a portion thereof).
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • vectors e.g., non-episomal mammalian vectors
  • Other vectors are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • certain vectors are capable of directing the expression of genes to which they are operatively linked.
  • Such vectors are referred to herein as "expression vectors".
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector can be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno- associated viruses), which serve equivalent functions.
  • the recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid sequence to be expressed.
  • "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cells and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like.
  • the expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., ARP proteins, mutant forms of ARP proteins, fusion proteins, and the like).
  • the recombinant expression vectors of the invention can be designed for expression of ARP proteins in prokaryotic or eukaryotic cells.
  • ARP proteins can be expressed in bacterial cells such as E. coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein.
  • Such fusion vectors typically serve three pu ⁇ oses: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • enzymes, and their cognate recognition sequences include Factor Xa, thrombin and enterokinase.
  • Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D.B. and Johnson, K.S.
  • GST glutathione S-transferase
  • Purified fusion proteins can be utilized in ARP activity assays, (e.g., direct assays or competitive assays described in detail below), or to generate antibodies specific for ARP proteins, for example.
  • an ARP fusion protein expressed in a retroviral expression vector of the present invention can be utilized to infect bone marrow cells which are subsequently transplanted into irradiated recipients. The pathology of the subject recipient is then examined after sufficient time has passed (e.g., six (6) weeks).
  • Suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) and pET l id (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 60-89).
  • Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid t ⁇ -lac fusion promoter.
  • Target gene expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
  • One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 119-128).
  • Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al., (1992) Nucleic Acids Res. 20:2111-2118).
  • Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
  • the ARP expression vector is a yeast expression vector.
  • yeast expression vectors for expression in yeast S. cerivisae include pYepSecl (Baldari, et al., (1987) Embo J. 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-
  • ARP proteins can be expressed in insect cells using baculovirus expression vectors.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39).
  • a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, B. (1987) Nature 329:840) and pMT2PC (Kaufman et al.
  • the expression vector's control functions are often provided by viral regulatory elements.
  • promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40.
  • suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed, Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.
  • the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid).
  • tissue-specific regulatory elements are known in the art.
  • suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1:268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBOJ.
  • promoters are also encompassed, for example the murine hox promoters (Kessel and Grass (1990) Science 249:374-379) and the ⁇ -fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
  • the invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to ARP mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA.
  • the antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced.
  • a high efficiency regulatory region the activity of which can be determined by the cell type into which the vector is introduced.
  • Another aspect of the invention pertains to host cells into which an ARP nucleic acid molecule of the invention is introduced, e.g., an ARP nucleic acid molecule within a recombinant expression vector or an ARP nucleic acid molecule in a form suitable for homologous recombination in the genome of a host cell (e.g., an ARP nucleic acid molecule which includes ARP nucleotide sequences and additional 5' and 3' flanking sequences necessary for homologous recombination).
  • host cell and "recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • an ARP protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • mammalian cells such as Chinese hamster ovary cells (CHO) or COS cells.
  • Other suitable host cells are known to those skilled in the art.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and “transfection” are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those which confer resistance to drags, such as G418, hygromycin and methotrexate.
  • Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding an ARP protein or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drag selection (e.g., cells that have inco ⁇ orated the selectable marker gene will survive, while the other cells die).
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) an ARP protein.
  • the invention further provides methods for producing an ARP protein using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding an ARP protein has been introduced) in a suitable medium such that an ARP protein is produced.
  • the method further comprises isolating an ARP protein from the medium or the host cell.
  • the host cells of the invention can also be used to produce non-human transgenic animals.
  • a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which ARP-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous ARP sequences have been introduced into their genome or homologous recombinant animals in which endogenous ARP sequences have been altered. Such animals are useful for studying the function and/or activity of an ARP and for identifying and/or evaluating modulators of ARP activity.
  • a "transgenic animal” is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene.
  • transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, and the like.
  • a transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal.
  • a "homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous ARP gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
  • a transgenic animal of the invention can be created by introducing an ARP- encoding nucleic acid into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retro viral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal.
  • the ARP cDNA sequence of SEQ ID NOT can be introduced as a transgene into the genome of a non-human animal.
  • a nonhuman homologue of a human ARP gene such as a mouse or rat ARP gene, can be used as a transgene.
  • an ARP gene homologue such as another GPCR family member, can be isolated based on hybridization to the ARP cDNA sequences of SEQ ID NOT, SEQ ID NO:3, or the DNA insert of the plasmid deposited with ATCC as
  • transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009, both by Leder et al, U.S. Patent No. 4,873,191 by Wagner et al.
  • transgenic founder animal can be identified based upon the presence of an ARP transgene in its genome and/or expression of ARP mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene encoding an ARP protein can further be bred to other transgenic animals carrying other transgenes.
  • a vector which contains at least a portion of an ARP gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the ARP gene.
  • the ARP gene can be a human gene (e.g., the cDNA of SEQ ID NO:3), but more preferably, is a non- human homologue of a human ARP gene (e.g., a cDNA isolated by stringent hybridization with the nucleotide sequence of SEQ ID NOT).
  • a mouse ARP gene can be used to construct a homologous recombination nucleic acid molecule, e.g., a vector, suitable for altering an endogenous ARP gene in the mouse genome.
  • the homologous recombination nucleic acid molecule is designed such that, upon homologous recombination, the endogenous ARP gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector).
  • the homologous recombination nucleic acid molecule can be designed such that, upon homologous recombination, the endogenous ARP gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous ARP protein).
  • the altered portion of the ARP gene is flanked at its 5' and 3' ends by additional nucleotide sequence of the ARP gene to allow for homologous recombination to occur between the exogenous ARP gene carried by the homologous recombination nucleic acid molecule and an endogenous ARP gene in a cell, e.g., an embryonic stem cell.
  • the additional flanking ARP nucleic acid sequence is of sufficient length for successful homologous recombination with the endogenous gene.
  • homologous recombination nucleic acid molecule typically, several kilobases of flanking DNA (both at the 5' and 3' ends) are included in the homologous recombination nucleic acid molecule (see, e.g., Thomas, K.R. and Capecchi, M. R. (1987) Cell 51:503 for a description of homologous recombination vectors).
  • the homologous recombination nucleic acid molecule is introduced into a cell, e.g., an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced ARP gene has homologously recombined with the endogenous ARP gene are selected (see e.g., Li, E. et al. (1992) Cell 69:915).
  • the selected cells can then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see e.g., Bradley, A. in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E.J. Robertson, ed. (IRL, Oxford, 1987) pp. 113-152).
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term.
  • Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene.
  • homologous recombination nucleic acid molecules e.g., vectors, or homologous recombinant animals are described further in Bradley, A. (1991) Current Opinion in Biotechnology 2:823-829 and in PCT International Publication Nos.: WO 90/11354 by Le Mouellec et al.; WO 91/01140 by Smithies et al.; WO 92/0968 by Zijlstra et al; and WO 93/04169 by Bems et al.
  • transgenic non-humans animals can be produced which contain selected systems which allow for regulated expression of the transgene.
  • a system is the cre/loxP recombinase system of bacteriophage PI .
  • cre/loxP recombinase system for a description of the cre/loxP recombinase system, see, e.g., Lakso et al. (1992) Proc. Natl. Acad. Sci. USA 89:6232-6236.
  • Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355.
  • mice containing transgenes encoding both the Cre recombinase and a selected protein are required.
  • Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
  • Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, I. et al. (1997) Nature 385:810- 813 and PCT International Publication Nos. WO 97/07668 and WO 97/07669.
  • a cell e.g., a somatic cell
  • the quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated.
  • the recontructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal.
  • the offspring bome of this female foster animal will be a clone of the animal from which the cell, e.g., the somatic cell, is isolated.
  • compositions suitable for administration can be inco ⁇ orated into pharmaceutical compositions suitable for administration.
  • Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and abso ⁇ tion delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be inco ⁇ orated into the compositions.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents
  • antibacterial agents such as benzyl alcohol or methyl parabens
  • antioxidants
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged abso ⁇ tion of the injectable compositions can be brought about by including in the composition an agent which delays abso ⁇ tion, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by inco ⁇ orating the active compound (e.g., a fragment of an ARP protein or an anti-ARP antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • the active compound e.g., a fragment of an ARP protein or an anti-ARP antibody
  • dispersions are prepared by inco ⁇ orating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the pu ⁇ ose of oral therapeutic administration, the active compound can be inco ⁇ orated with excipients and used in the form of tablets, troches, or capsules, oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio
  • LD50/ED50 Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
  • the nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors.
  • Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054- 3057).
  • the pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • nucleic acid molecules, proteins, protein homologues, and antibodies described herein can be used in one or more of the following methods: a) screening assays; b) predictive medicine (e.g., diagnostic assays, prognostic assays, monitoring clinical trials, and pharmacogenetics); and c) methods of treatment (e.g., therapeutic and prophylactic).
  • an ARP protein of the invention has one or more of the following activities: (1) it interacts with a non-ARP protein molecule on the surface of the same cell which expresses it; (2) it interacts with a non-ARP protein molecule on the surface of a different cell; (3) it activates an ARP-dependent signal transduction pathway; and (4) it modulates programmed cell death, and, thus, can be used to, for example, (1) modulate the interaction with a non-ARP protein molecule on the surface of the same cell which expresses it; (2) modulate the interaction with a non-ARP protein molecule on the surface of a different cell; (3) to activate an ARP-dependent signal transduction pathway; and (4) to modulate programmed cell death.
  • the isolated nucleic acid molecules of the invention can be used, for example, to express ARP protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect ARP mRNA (e.g., in a biological sample) or a genetic alteration in an ARP gene, and to modulate ARP activity, as described further below.
  • ARP proteins can be used to treat disorders characterized by insufficient or excessive production of an ARP substrate or production of ARP inhibitors.
  • the ARP proteins can be used to screen for naturally occurring ARP substrates, to screen for drugs or compounds which modulate ARP activity, as well as to treat disorders characterized by insufficient or excessive production of ARP protein or production of ARP protein forms which have decreased or aberrant activity compared to ARP wild type protein (e.g., disorders associated with programmed cell death).
  • the anti-ARP antibodies of the invention can be used to detect and isolate ARP proteins, regulate the bioavailability of ARP proteins, and modulate ARP activity.
  • the invention provides a method (also referred to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) which bind to ARP proteins, have a stimulatory or inhibitory effect on, for example, ARP expression or ARP activity, or have a stimulatory or inhibitory effect on, for example, the expression or activity of ARP substrate.
  • the invention provides assays for screening candidate or test compounds which are substrates of an ARP protein or polypeptide or biologically active portion thereof.
  • the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of an ARP protein or polypeptide or biologically active portion thereof.
  • the test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection.
  • the biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, K.S. (1997) Anticancer Drug Des. 12: 145).
  • an assay is a cell-based assay in which a cell which expresses an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to modulate ARP activity is determined. Determining the ability of the test compound to modulate ARP activity can be accomplished by monitoring, for example, the survival of a cell which expresses ARP or the activity of an ARP-regulated transcription factor.
  • the cell for example, can be of mammalian origin.
  • the ability of the test compound to modulate ARP binding to a substrate or to bind to ARP can also be determined. Determining the ability of the test compound to modulate ARP binding to a substrate can be accomplished, for example, by coupling the ARP substrate with a radioisotope or enzymatic label such that binding of the ARP substrate to ARP can be determined by detecting the labeled ARP substrate in a complex. Determining the ability of the test compound to bind ARP can be accomplished, for example, by coupling the compound with a radioisotope or enzymatic label such that binding of the compound to ARP can be determined by detecting the labeled ARP compound in a complex.
  • compounds e.g., ARP substrates
  • compounds can be labeled with ⁇ l, ⁇ S, ⁇ C, or -1H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting.
  • compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
  • a microphysiometer can be used to detect the interaction of a compound with ARP without the labeling of either the compound or the ARP. McConnell, H. M. et al. (1992) Science 257:1906-1912.
  • a "microphysiometer” e.g., Cytosensor
  • LAPS light-addressable potentiometric sensor
  • an assay is a cell-based assay comprising contacting a cell expressing an ARP target molecule (e.g., an ARP substrate) with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the ARP target molecule. Determining the ability of the test compound to modulate the activity of an ARP target molecule can be accomplished, for example, by determining the ability of the ARP protein to bind to or interact with the ARP target molecule.
  • an ARP target molecule e.g., an ARP substrate
  • Determining the ability of the test compound to modulate the activity of an ARP target molecule can be accomplished, for example, by determining the ability of the ARP protein to bind to or interact with the ARP target molecule.
  • Determining the ability of the ARP protein or a biologically active fragment thereof, to bind to or interact with an ARP target molecule can be accomplished by one of the methods described above for determining direct binding. In a preferred embodiment, determining the ability of the ARP protein to bind to or interact with an ARP target molecule can be accomplished by determining the activity of the target molecule.
  • the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e., intracellular Ca2+, diacylglycerol, IP3, and the like), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising a target- responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a target-regulated cellular response.
  • a cellular second messenger of the target i.e., intracellular Ca2+, diacylglycerol, IP3, and the like
  • detecting catalytic/enzymatic activity of the target an appropriate substrate detecting the induction of a reporter gene (comprising a target- responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a target-regulated cellular
  • an assay of the present invention is a cell-free assay in which an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to bind to the ARP protein or biologically active portion thereof is determined.
  • Preferred biologically active portions of the ARP proteins to be used in assays of the present invention include fragments which participate in interactions with non-ARP molecules, e.g., fragments with high surface probability scores (see, for example, Figure 2). Binding of the test compound to the ARP protein can be determined either directly or indirectly as described above.
  • the assay includes contacting the ARP protein or biologically active portion thereof with a known compound which binds ARP to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an ARP protein, wherein determining the ability of the test compound to interact with an ARP protein comprises determining the ability of the test compound to preferentially bind to ARP or biologically active portion thereof as compared to the known compound.
  • the assay is a cell-free assay in which an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the ARP protein or biologically active portion thereof is determined.
  • Determining the ability of the test compound to modulate the activity of an ARP protein can be accomplished, for example, by determining the ability of the ARP protein to bind to an ARP target molecule by one of the methods described above for determining direct binding. Determining the ability of the ARP protein to bind to an ARP target molecule can also be accomplished using a technology such as real-time Biomolecular Interaction Analysis (BIA). Sjolander, S. and Urbaniczky, C.
  • BIOA is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore). Changes in the optical phenomenon of surface plasmon resonance
  • SPR SPR
  • determining the ability of the test compound to modulate the activity of an ARP protein can be accomplished by determining the ability of the ARP protein to further modulate the activity of a downstream effector of an ARP target molecule.
  • the activity of the effector molecule on an appropriate target can be determined or the binding of the effector to an appropriate target can be determined as previously described.
  • the cell-free assay involves contacting an ARP protein or biologically active portion thereof with a known compound which binds the ARP protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the ARP protein, wherein determining the ability of the test compound to interact with the ARP protein comprises determining the ability of the ARP protein to preferentially bind to or modulate the activity of an ARP target molecule.
  • the cell-free assays of the present invention are amenable to use of both soluble and/or membrane-bound forms of isolated proteins (e.g., ARP proteins or biologically active portions thereof).
  • a membrane-bound form of an isolated protein e.g., the large ARP isoform
  • a solubilizing agent such that the membrane-bound form of the isolated protein is maintained in solution.
  • non-ionic detergents such as n-oc
  • binding of a test compound to an ARP protein, or interaction of an ARP protein with a target molecule in the presence and absence of a candidate compound can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix.
  • glutathione-S-transferase/ ARP fusion proteins or glutathione-S- transferase/target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or ARP protein, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtitre plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of ARP binding or activity determined using standard techniques.
  • an ARP protein or an ARP target molecule can be immobilized utilizing conjugation of biotin and streptavidin.
  • Biotinylated ARP protein or target molecules can be prepared from biotin-NHS (N- hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with ARP protein or target molecules but which do not interfere with binding of the ARP protein to its target molecule can be derivatized to the wells of the plate, and unbound target or ARP protein trapped in the wells by antibody conjugation.
  • Methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the ARP protein or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the ARP protein or target molecule.
  • modulators of ARP expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of ARP mRNA or protein in the cell is determined. The level of expression of ARP mRNA or protein in the presence of the candidate compound is compared to the level of expression of ARP mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of ARP expression based on this comparison. For example, when expression of ARP mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of ARP mRNA or protein expression.
  • the candidate compound when expression of ARP mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of ARP mRNA or protein expression.
  • the level of ARP mRNA or protein expression in the cells can be determined by methods described herein for detecting ARP mRNA or protein.
  • the ARP proteins can be used as "bait proteins" in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol. Chem.
  • ARP-binding proteins proteins which bind to or interact with ARP
  • ARP-binding proteins proteins which bind to or interact with ARP
  • ARP-binding proteins are also likely to be involved in the propagation of signals by the ARP proteins or ARP targets as, for example, downstream elements of an ARP-mediated signaling pathway.
  • ARP-binding proteins are likely to be ARP inhibitors.
  • the two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains.
  • the assay utilizes two different DNA constracts.
  • the gene that codes for an ARP protein is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4).
  • a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein (“prey" or "sample” is fused to a gene that codes for the activation domain of the known transcription factor.
  • the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene which encodes the protein which interacts with the ARP protein.
  • a reporter gene e.g., LacZ
  • This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model.
  • an agent identified as described herein e.g., an ARP modulating agent, an antisense ARP nucleic acid molecule, an ARP-specific antibody, or an ARP-binding partner
  • an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent.
  • an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent.
  • this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.
  • cDNA sequences identified herein can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. These applications are described in the subsections below.
  • this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the ARP nucleotide sequences, described herein, can be used to map the location of the ARP genes on a chromosome. The mapping of the ARP sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease.
  • ARP genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the ARP nucleotide sequences. Computer analysis of the ARP sequences can be used to predict primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the ARP sequences will yield an amplified fragment. Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells).
  • Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the ARP nucleotide sequences to design oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes.
  • Other mapping strategies which can similarly be used to map an ARP sequence to its chromosome include in situ hybridization (described in Fan, Y. et al. (1990) Proc. Natl. Acad. Sci.
  • FISH Fluorescence in situ hybridization
  • Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical such as colcemid that disrupts the mitotic spindle.
  • the chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually.
  • the FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1 ,000 bases, and more preferably 2,000 bases will suffice to get good results at a reasonable amount of time. For a review of this technique, see Verma et al., Human Chromosomes: A Manual of Basic Techniques (Pergamon Press, New York 1988).
  • Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents corresponding to noncoding regions of the genes actually are preferred for mapping pu ⁇ oses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
  • a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymo ⁇ hisms.
  • the ARP sequences of the present invention can also be used to identify individuals from minute biological samples.
  • the United States military for example, is considering the use of restriction fragment length polymo ⁇ hism (RFLP) for identification of its personnel.
  • RFLP restriction fragment length polymo ⁇ hism
  • an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification.
  • This method does not suffer from the current limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult.
  • the sequences of the present invention are useful as additional DNA markers for RFLP (described in U.S. Patent 5,272,057).
  • sequences of the present invention can be used to provide an alternative technique which determines the actual base-by-base DNA sequence of selected portions of an individual's genome.
  • the ARP nucleotide sequences described herein can be used to prepare two PCR primers from the 5' and 3' ends of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it. Panels of corresponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences.
  • the sequences of the present invention can be used to obtain such identification sequences from individuals and from tissue.
  • the ARP nucleotide sequences of the invention uniquely represent portions of the human genome.
  • allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases.
  • Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification pu ⁇ oses. Because greater numbers of polymo ⁇ hisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals.
  • the noncoding sequences of SEQ ID NOT can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers which each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as that in SEQ ID NO:3 is used, a more appropriate number of primers for positive individual identification would be 500-2,000.
  • a panel of reagents from ARP nucleotide sequences described herein is used to generate a unique identification database for an individual, those same reagents can later be used to identify tissue from that individual.
  • Using the unique identification database positive identification of the individual, living or dead, can be made from extremely small tissue samples.
  • Forensic biology is a scientific field employing genetic typing of biological evidence found at a crime scene as a means for positively identifying, for example, a pe ⁇ etrator of a crime.
  • PCR technology can be used to amplify DNA sequences taken from very small biological samples such as tissues, e.g., hair or skin, or body fluids, e.g., blood, saliva, or semen found at a crime scene. The amplified sequence can then be compared to a standard, thereby allowing identification of the origin of the biological sample.
  • sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another "identification marker" (i.e. another DNA sequence that is unique to a particular individual).
  • an "identification marker” i.e. another DNA sequence that is unique to a particular individual.
  • actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments.
  • Sequences targeted to noncoding regions of SEQ ID NOT are particularly appropriate for this use as greater numbers of polymo ⁇ hisms occur in the noncoding regions, making it easier to differentiate individuals using this technique.
  • polynucleotide reagents include the ARP nucleotide sequences or portions thereof, e.g., fragments derived from the noncoding regions of SEQ ID NOT having a length of at least 20 bases, preferably at least 30 bases.
  • the ARP nucleotide sequences described herein can further be used to provide polynucleotide reagents, e.g., labeled or labelable probes which can be used in, for example, an in situ hybridization technique, to identify a specific tissue, e.g., brain tissue. This can be very useful in cases where a forensic pathologist is presented with a tissue of unknown origin. Panels of such ARP probes can be used to identify tissue by species and or by organ type.
  • these reagents e.g., ARP primers or probes can be used to screen tissue culture for contamination (i.e. screen for the presence of a mixture of different types of cells in a culture).
  • the present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, and monitoring clinical trials are used for prognostic (predictive) pu ⁇ oses to thereby treat an individual prophylactically.
  • one aspect of the present invention relates to diagnostic assays for determining ARP protein and/or nucleic acid expression as well as ARP activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant ARP expression or activity.
  • the invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with ARP protein, nucleic acid expression or activity. For example, mutations in an ARP gene can be assayed in a biological sample.
  • Such assays can be used for prognostic or predictive pu ⁇ ose to thereby phophylactically treat an individual prior to the onset of a disorder characterized by or associated with ARP protein, nucleic acid expression or activity.
  • Another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of ARP in clinical trials.
  • agents e.g., drugs, compounds
  • An exemplary method for detecting the presence or absence of ARP protein or nucleic acid in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting ARP protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes ARP protein such that the presence of ARP protein or nucleic acid is detected in the biological sample.
  • a preferred agent for detecting ARP mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to ARP mRNA or genomic DNA.
  • the nucleic acid probe can be, for example, a full-length ARP nucleic acid, such as the nucleic acid of SEQ ID NOT, SEQ ID NO:3, or the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion thereof, such as an oligonucleotide of at least
  • a preferred agent for detecting ARP protein is an antibody capable of binding to ARP protein, preferably an antibody with a detectable label.
  • Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used.
  • the term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • biological sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect ARP mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of ARP mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of ARP protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence.
  • In vitro techniques for detection of ARP genomic DNA include Southern hybridizations.
  • in vivo techniques for detection of ARP protein include introducing into a subject a labeled anti-ARP antibody.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • the biological sample contains protein molecules from the test subject.
  • the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject.
  • a preferred biological sample is a serum sample isolated by conventional means from a subject.
  • the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting ARP protein, mRNA, or genomic DNA, such that the presence of ARP protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of ARP protein, mRNA or genomic DNA in the control sample with the presence of ARP protein, mRNA or genomic DNA in the test sample.
  • kits for detecting the presence of ARP in a biological sample can comprise a labeled compound or agent capable of detecting ARP protein or mRNA in a biological sample; means for determining the amount of ARP in the sample; and means for comparing the amount of ARP in the sample with a standard.
  • the compound or agent can be packaged in a suitable container.
  • the kit can further comprise instructions for using the kit to detect ARP protein or nucleic acid.
  • the diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant ARP expression or activity.
  • aberrant includes an ARP expression or activity which deviates from the wild type ARP expression or activity.
  • Aberrant expression or activity includes increased or decreased expression or activity, as well as expression or activity which does not follow the wild type developmental pattern of expression or the subcellular pattern of expression.
  • aberrant ARP expression or activity is intended to include the cases in which a mutation in the ARP gene causes the ARP gene to be under-expressed or over-expressed and situations in which such mutations result in a non-functional ARP protein or a protein which does not function in a wild-type fashion, e.g., a protein which does not interact with an ARP ligand or one which interacts with a non-ARP ligand.
  • the assays described herein can be utilized to identify a subject having or at risk of developing a disorder associated with a misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder.
  • the prognostic assays can be utilized to identify a subject having or at risk for developing a disorder associated with a misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder.
  • the present invention provides a method for identifying a disease or disorder associated with aberrant ARP expression or activity in which a test sample is obtained from a subject and ARP protein or nucleic acid (e.g., mRNA or genomic DNA) is detected, wherein the presence of ARP protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with aberrant ARP expression or activity.
  • a test sample refers to a biological sample obtained from a subject of interest.
  • a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
  • the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate) to treat a disease or disorder associated with aberrant ARP expression or activity.
  • an agent e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate
  • such methods can be used to determine whether a subject can be effectively treated with an agent for a proliferative disorder, a differentiative disorder, or a gland associated disorder.
  • the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant ARP expression or activity in which a test sample is obtained and ARP protein or nucleic acid expression or activity is detected (e.g., wherein the abundance of ARP protein or nucleic acid expression or activity is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant ARP expression or activity).
  • the methods of the invention can also be used to detect genetic alterations in an ARP gene, thereby determining if a subject with the altered gene is at risk for a disorder characterized by misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder.
  • the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by at least one of an alteration affecting the integrity of a gene encoding an ARP -protein, or the mis- expression of the ARP gene.
  • such genetic alterations can be detected by ascertaining the existence of at least one of 1) a deletion of one or more nucleotides from an ARP gene; 2) an addition of one or more nucleotides to an ARP gene; 3) a substitution of one or more nucleotides of an ARP gene, 4) a chromosomal rearrangement of an ARP gene; 5) an alteration in the level of a messenger RNA transcript of an ARP gene, 6) aberrant modification of an ARP gene, such as of the methylation pattern of the genomic DNA, 7) the presence of a non- ild type splicing pattern of a messenger RNA transcript of an ARP gene, 8) a non-wild type level of an ARP -protein, 9) allelic loss of an ARP gene, and 10) inappropriate post-translational modification of an ARP -protein.
  • a preferred biological sample is a tissue or serum sample isolated by conventional means from a subject.
  • detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1994) Proc. Natl. Acad.
  • PCR polymerase chain reaction
  • LCR ligation chain reaction
  • This method can include the steps of collecting a sample of cells from a subject, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to an ARP gene under conditions such that hybridization and amplification of the ARP-gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein.
  • Alternative amplification methods include: self sustained sequence replication
  • mutations in an ARP gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns.
  • sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA.
  • sequence specific ribozymes see, for example, U.S. Patent No. 5,498,531 can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
  • genetic mutations in ARP can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotides probes (Cronin, M.T. et al. (1996) Human Mutation 7: 244-255; Kozal, M.J. et al. (1996) Nature Medicine 2: 753- 759).
  • oligonucleotides probes e.g., DNA or RNA
  • genetic mutations in ARP can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, M.T. et al. supra.
  • a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene.
  • any of a variety of sequencing reactions known in the art can be used to directly sequence the ARP gene and detect mutations by comparing the sequence of the sample ARP with the corresponding wild-type (control) sequence.
  • sequencing reactions include those based on techniques developed by Maxam and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995) Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36:127-162; and Griffin et al. (1993) Appl. Biochem. Biotechnol. 38:147-159).
  • RNA/RNA or RNA/DNA heteroduplexes Other methods for detecting mutations in the ARP gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242).
  • the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type ARP sequence with potentially mutant RNA or DNA obtained from a tissue sample.
  • the double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands.
  • RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with SI nuclease to enzymatically digesting the mismatched regions.
  • either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al. (1988) Proc. Natl Acad Sci USA 85 :4397; Saleeba et al. (1992) Methods Enzymol 217:286-295.
  • the control DNA or RNA can be labeled for detection.
  • the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in ARP cDNAs obtained from samples of cells.
  • DNA mismatch repair enzymes
  • the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662).
  • a probe based on an ARP sequence e.g., a wild-type ARP sequence
  • a cDNA or other DNA product from a test cell(s).
  • the duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S. Patent No. 5,459,039.
  • alterations in electrophoretic mobility will be used to identify mutations in ARP genes.
  • single strand conformation polymo ⁇ hism may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (orita et al. (1989) Proc Natl. Acad. Sci USA: 86:2766, see also Cotton (1993) Mutat. Res. 285:125-144; and Hayashi (1992) Genet. Anal. Tech. Appl. 9:73-79).
  • Single-stranded DNA fragments of sample and control ARP nucleic acids will be denatured and allowed to renature.
  • the secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change.
  • the DNA fragments may be labeled or detected with labeled probes.
  • the sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence.
  • the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet 7:5).
  • mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. ( 1985) Nature 313:495).
  • DGGE denaturing gradient gel electrophoresis
  • DGGE DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR.
  • a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:12753).
  • oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al. (1989) Proc. Natl Acad. Sci USA 86:6230).
  • Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA.
  • Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11 :238).
  • amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification.
  • the methods described herein may be performed, for example, by utilizing prepackaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving an ARP gene.
  • any cell type or tissue in which ARP is expressed may be utilized in the prognostic assays described herein.
  • Monitoring the influence of agents (e.g., drugs) on the expression or activity of an ARP protein can be applied not only in basic drag screening, but also in clinical trials.
  • agents e.g., drugs
  • the effectiveness of an agent determined by a screening assay as described herein to increase ARP gene expression, protein levels, or upregulate ARP activity can be monitored in clinical trials of subjects exhibiting decreased ARP gene expression, protein levels, or downregulated ARP activity.
  • the effectiveness of an agent determined by a screening assay to decrease ARP gene expression, protein levels, or downregulate ARP activity can be monitored in clinical trials of subjects exhibiting increased ARP gene expression, protein levels, or upregulated ARP activity.
  • the expression or activity of an ARP gene, and preferably, other genes that have been implicated in, for example, an ARP-associated disorder can be used as a "read out" or markers of the phenotype of a particular cell.
  • genes, including ARP that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) which modulates ARP activity (e.g., identified in a screening assay as described herein) can be identified.
  • an agent e.g., compound, drug or small molecule
  • ARP activity e.g., identified in a screening assay as described herein
  • agents on ARP-associated disorders e.g., proliferative disorders, differentiative disorders, or gland associated disorders
  • cells can be isolated and RNA prepared and analyzed for the levels of expression of ARP and other genes implicated in the ARP-associated disorder, respectively.
  • the levels of gene expression can be quantified by northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of ARP or other genes.
  • the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during treatment of the individual with the agent.
  • the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate identified by the screening assays described herein) including the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of expression of an ARP protein, mRNA, or genomic DNA in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the ARP protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the ARP protein, mRNA, or genomic DNA in the pre- administration sample with the ARP protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly.
  • an agent e.g., an agonist
  • increased administration of the agent may be desirable to increase the expression or activity of ARP to higher levels than detected, i.e., to increase the effectiveness of the agent.
  • decreased administration of the agent may be desirable to decrease expression or activity of ARP to lower levels than detected, i.e. to decrease the effectiveness of the agent.
  • ARP expression or activity may be used as an indicator of the effectiveness of an agent, even in the absence of an observable phenotypic response.
  • the present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant ARP expression or activity.
  • treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
  • “Pharmacogenomics”, as used herein, refers to the application of genomics technologies such as gene sequencing, statistical genetics, and gene expression analysis to drugs in clinical development and on the market.
  • the term refers the study of how a patient's genes determine his or her response to a drag (e.g., a patient's "drag response phenotype", or “drag response genotype”.)
  • a drag response genotype e.g., a patient's "drag response phenotype", or "drag response genotype”.
  • another aspect of the invention provides methods for tailoring an individual's prophylactic or therapeutic treatment with either the ARP molecules of the present invention or ARP modulators according to that individual's drag response genotype.
  • Pharmacogenomics allows a clinician or physician to target prophylactic or therapeutic treatments to patients who will most benefit from the treatment and to avoid treatment of patients who will experience toxic drag-related side effects.
  • the invention provides a method for preventing in a subject, a disease or condition associated with an aberrant ARP expression or activity, by administering to the subject an ARP or an agent which modulates ARP expression or at least one ARP activity.
  • Subjects at risk for a disease which is caused or contributed to by aberrant ARP expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein.
  • Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the ARP aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression.
  • an ARP, ARP agonist or ARP antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein.
  • the modulatory method of the invention involves contacting a cell with an ARP or agent that modulates one or more of the activities of ARP protein activity associated with the cell.
  • An agent that modulates ARP protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring target molecule of an ARP protein (e.g., an ARP substrate), an ARP antibody, an ARP agonist or antagonist, a peptidomimetic of an ARP agonist or antagonist, or other small molecule.
  • the agent stimulates one or more ARP activities.
  • stimulatory agents include active ARP protein and a nucleic acid molecule encoding ARP that has been introduced into the cell.
  • the agent inhibits one or more ARP activities.
  • inhibitory agents include antisense ARP nucleic acid molecules, anti-ARP antibodies, and ARP inhibitors.
  • the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., upregulates or downregulates) ARP expression or activity.
  • the method involves administering an ARP protein or nucleic acid molecule as therapy to compensate for reduced or aberrant ARP expression or activity. Stimulation of ARP activity is desirable in situations in which ARP is abnormally downregulated and/or in which increased ARP activity is likely to have a beneficial effect. For example, stimulation of ARP activity is desirable in situations in which an ARP is downregulated and/or in which increased ARP activity is likely to have a beneficial effect. Likewise, inhibition of ARP activity is desirable in situations in which ARP is abnormally upregulated and/or in which decreased ARP activity is likely to have a beneficial effect.
  • ARP molecules of the present invention as well as agents, or modulators which have a stimulatory or inhibitory effect on ARP activity (e.g., ARP gene expression) as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) ARP-associated disorders (e.g., programmed cell death associated disorders) associated with aberrant ARP activity.
  • ARP-associated disorders e.g., programmed cell death associated disorders
  • pharmacogenomics i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug
  • Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug.
  • a physician or clinician may consider applying knowledge obtained in relevant pharmacogenomics studies in determining whether to administer an ARP molecule or ARP modulator as well as tailoring the dosage and/or therapeutic regimen of treatment with an ARP molecule or ARP modulator.
  • Pharmacogenomics deals with clinically significant hereditary variations in the response to drags due to altered drug disposition and abnormal action in affected persons. See, for example, Eichelbaum, M. et al. (1996) Clin. Exp. Pharmacol. Physiol 23(10-11) :983-985 and Linder, M.W. et al. (1997) Clin. Chem. 43(2):254-266.
  • two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drags act on the body (altered drag action) or genetic conditions transmitted as single factors altering the way the body acts on drags (altered drug metabolism). These pharmacogenetic conditions can occur either as rare genetic defects or as naturally-occurring polymo ⁇ hisms.
  • G6PD glucose-6-phosphate dehydrogenase deficiency
  • oxidant drags anti-malarials, sulfonamides, analgesics, nitrofurans
  • a genome-wide association relies primarily on a high-resolution map of the human genome consisting of already known gene-related markers (e.g., a "bi- allelic” gene marker map which consists of 60,000-100,000 polymo ⁇ hic or variable sites on the human genome, each of which has two variants.)
  • gene-related markers e.g., a "bi- allelic” gene marker map which consists of 60,000-100,000 polymo ⁇ hic or variable sites on the human genome, each of which has two variants.
  • Such a high-resolution genetic map can be compared to a map of the genome of each of a statistically significant number of patients taking part in a Phase II/III drag trial to identify markers associated with a particular observed drug response or side effect.
  • such a high resolution map can be generated from a combination of some ten-million known single nucleotide polymo ⁇ hisms (SNPs) in the human genome.
  • SNP single nucleotide polymo ⁇ hisms
  • a "SNP" is a common alteration that occurs in a single nucleotide base in a stretch of DNA. For example, a SNP may occur once per every 1000 bases of DNA.
  • a SNP may be involved in a disease process, however, the vast majority may not be disease- associated.
  • individuals Given a genetic map based on the occurrence of such SNPs, individuals can be grouped into genetic categories depending on a particular pattern of SNPs in their individual genome. In such a manner, treatment regimens can be tailored to groups of genetically similar individuals, taking into account traits that may be common among such genetically similar individuals.
  • a method termed the "candidate gene approach” can be utilized to identify genes that predict drag response.
  • a gene that encodes a drags target e.g., an ARP protein of the present invention
  • all common variants of that gene can be fairly easily identified in the population and it can be determined if having one version of the gene versus another is associated with a particular drag response.
  • the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action.
  • the gene coding for CYP2D6 is highly polymo ⁇ hic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drag response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite mo ⁇ hine. The other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
  • a method termed the "gene expression profiling" can be utilized to identify genes that predict drag response.
  • a drag e.g., an ARP molecule or ARP modulator of the present invention
  • the gene expression of an animal dosed with a drag can give an indication whether gene pathways related to toxicity have been turned on.
  • Information generated from more than one of the above pharmacogenomics approaches can be used to determine appropriate dosage and treatment regimens for prophylactic or therapeutic treatment an individual.
  • This knowledge when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an ARP molecule or ARP modulator, such as a modulator identified by one of the exemplary screening assays described herein.
  • This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are inco ⁇ orated herein by reference.
  • the invention is based, at least in part, on the discovery of a human gene encoding a novel protein, referred to herein as ARP.
  • ARP a human gene encoding a novel protein
  • the human ARP was isolated from a human heart cDNA library purchased from Clonetch Marathon. Positive clones were isolated and subsequently 5' RACE-PCR was used to obtain the full length clone.
  • the sequence of the entire human clone was determined and found to contain an open reading frame of 353 amino acids termed human "ARP."
  • the nucleotide sequence encoding the human ARP protein is shown in Figure 1 and is set forth as SEQ ID NOT .
  • the full length protein encoded by this nucleic acid comprises about 353 amino acids and has the amino acid sequence shown in Figure 1 and set forth as SEQ ID NO:2.
  • the coding region (open reading frame) of SEQ ID NOT is set forth as SEQ ID NO:3.
  • Clone MHT comprising the entire coding region of human ARP was deposited with the American Type Culture Collection (ATCC®), 10801 University Boulevard, Manassas, VA 20110-2209, on , and assigned Accession No. .
  • the ARP protein is 80%> identical to the Cricetulus griseus HT protein (Accession Number U48852) over translated nucleotides 70 to 558.
  • a BLASTN 1.4.9 search using a score of 100 and a word length of 12 (Altschul et al. (1990) J. Mol. Biol. 215:403) of the nucleotide sequence of human ARP revealed that ARP is similar to the Homo sapiens cDNA clone 668306 (Accession Number AA243887).
  • the ARP nucleic acid molecule is 99%> identical to the Homo sapiens cDNA clone 668306 (Accession Number AA243887) over nucleotides 755 to 1062.
  • a search using the human ARP protein sequence was performed against the HMM database resulting in the identification of an EGF-like domain in the amino acid sequence of SEQ ID NO:2 (residuesl40 to 177). The results of the search are set forth in Figure 3. Using the same search, a Laminin EGF-like domain was also identified in the amino acid sequence of SEQ ID NO:2 (residuesl48 to 190). The results of this search are set forth in Figure 4.
  • This Example describes the tissue distribution of ARP mRNA, as determined by Northern blot hybridization.
  • Northern blot hybridizations with the various RNA samples were performed under standard conditions and washed under stringent conditions, i.e., 0.2xSSC at 65°C.
  • the DNA probe was radioactively labeled with 32p.dCTP using the Prime-It kit (Stratagene, La Jolla, CA) according to the instructions of the supplier.
  • Filters containing human mRNA MultiTissue Northern I and MultiTissue Northern II from Clontech, Palo Alto, CA
  • One isoform (larger message) could be a transmembrane protein (frizzled-like) and the other isoform could be the secreted form (smaller message).
  • the two isoforms show a clear pattern of tissue specificity. On the multiple tissue blot from Clonetech, the large transcript is found in almost all tissues whereas the smaller message is expressed mainly in the heart, skeletal muscle placenta and the pancreas (see Figure 5).
  • EXAMPLE 2 EXPRESSION OF RECOMBINANT ARP
  • ARP is expressed as a recombinant glutathione-S-transferase
  • GST fusion polypeptide in E. coli and the fusion polypeptide is isolated and characterized.
  • ARP is fused to GST and this fusion polypeptide is expressed in E. coli, e.g., strain PEB199.
  • Expression of the GST- ARP fusion protein in PEB199 is induced with IPTG.
  • the recombinant fusion polypeptide is purified from crade bacterial lysates of the induced PEB199 strain by affinity chromatography on glutathione beads. Using polyacrylamide gel electrophoretic analysis of the polypeptide purified from the bacterial lysates, the molecular weight of the resultant fusion polypeptide is determined.
  • the pcDNA/Amp vector by Invitrogen Co ⁇ oration (San Diego, CA) is used.
  • This vector contains an SV40 origin of replication, an ampicillin resistance gene, an E. coli replication origin, a CMV promoter followed by a polylinker region, and an SV40 intron and polyadenylation site.
  • a DNA fragment encoding the entire ARP protein and an HA tag (Wilson et al. (1984) Cell 37:767) or a FLAG tag fused in-frame to its 3' end of the fragment is cloned into the polylinker region of the vector, thereby placing the expression of the recombinant protein under the control of the CMV promoter.
  • the ARP DNA sequence is amplified by PCR using two primers.
  • the 5' primer contains the restriction site of interest followed by approximately twenty nucleotides of the ARP coding sequence starting from the initiation codon; the 3' end sequence contains complementary sequences to the other restriction site of interest, a translation stop codon, the HA tag or FLAG tag and the last 20 nucleotides of the ARP coding sequence.
  • the PCR amplified fragment and the pCDNA/Amp vector are digested with the appropriate restriction enzymes and the vector is dephosphorylated using the CIAP enzyme (New England Biolabs, Beverly, MA).
  • the two restriction sites chosen are different so that the ARP gene is inserted in the correct orientation.
  • the ligation mixture is transformed into E. coli cells (strains HB101, DH5a, SURE, available from Stratagene Cloning Systems, La Jolla, CA, can be used), the transformed culture is plated on ampicillin media plates, and resistant colonies are selected. Plasmid DNA is isolated from transformants and examined by restriction analysis for the presence of the correct fragment.
  • COS cells are subsequently transfected with the ARP-pcDNA/Amp plasmid DNA using the calcium phosphate or calcium chloride co-precipitation methods, DEAE- dextran-mediated transfection, lipofection, or electroporation.
  • Other suitable methods for transfecting host cells can be found in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.
  • the expression of the ARP polypeptide is detected by radiolabelling ( 35 S-methionine or 35 S-cysteine available from NEN, Boston, MA, can be used) and immunoprecipitation (Harlow, E. and Lane, D. Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1988) using an HA specific monoclonal antibody. Briefly, the cells are labelled for 8 hours with 35 S-methionine (or 35 S-cysteine). The culture media are then collected and the cells are lysed using detergents (RIPA buffer, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% DOC, 50 mM Tris, pH 7.5). Both the cell lysate and the culture media are precipitated with an HA specific monoclonal antibody. Precipitated polypeptides are then analyzed by SDS-PAGE.
  • DNA containing the ARP coding sequence is cloned directly into the polylinker of the pCDNA/Amp vector using the appropriate restriction sites.
  • the resulting plasmid is transfected into COS cells in the manner described above, and the expression of the ARP polypeptide is detected by radiolabelling and immunoprecipitation using an ARP specific monoclonal antibody.

Abstract

The invention provides isolated nucleic acids molecules, designated ARP nucleic acid molecules, which encode novel ARP family members. The invention also provides antisense nucleic acid molecules, recombinant expression vectors containing ARP nucleic acid molecules, host cells into which the expression vectors have been introduced, and nonhuman transgenic animals in which an ARP gene has been introduced or disrupted. The invention still further provides isolated ARP proteins, fusion proteins, antigenic peptides and anti-ARP antibodies. Diagnostic methods utilizing compositions of the invention are also provided.

Description

NOVEL APOPTOSIS RELATED PROTEINS AND USES THEREFOR
Background of the Invention
During normal embryonic and adult development of multicellular organisms, cells that are not necessary or deleterious are eliminated by a process referred to as programmed cell death or apoptosis (Ellis R.E. et al. (1991) Annual Rev. Cell Biol 7:663-698). Programmed cell death occurs in both vertebrate and invertebrate species and is characterized by unique morphological alterations, such as cytoplasmic contraction and chromatin condensation, as well as by specific DNA cleavage into oligonucleosomal fragments. Unlike necrosis, programmed cell death or apoptosis is an irreversible process which in most systems appears to depend on the expression of a specific set of novel "death genes". Deregulation of this process contributes to the pathogenesis of several diseases including cancer, immunodeficiency,autoimmune diseases, and neurodegenerative disorders (Thompson C.B. et al. (1995) Science 267: 1456).
Recently, a new family of molecules believed to play a role in apoptosis has been identified (Melkonyan H. S. et al. (1997) Proc. Natl. Acad. Sci. 94:13636-13641). These molecules are referred to as Secreted Apoptosis-Related Proteins (SARP) and have a cysteine rich domain homologous to the cysteine rich domain of the frizzled proteins, a class of D. melanogaster proteins which function as receptors for the Wingless protein (Zorn A. (1997) Current Biol. 7:R501-R504).
Summary of the Invention
The present invention is based, at least in part, on the discovery of novel apoptosis related protein (ARP) family members, referred to herein as "ARP" nucleic acid and protein molecules. The ARP molecules of the present invention are useful as modulating agents for regulating a variety of cellular processes. Accordingly, in one aspect, this invention provides isolated nucleic acid molecules encoding ARP proteins or biologically active portions thereof, as well as nucleic acid fragments suitable as primers or hybridization probes for the detection of ARP-encoding nucleic acids. In one embodiment, an ARP nucleic acid molecule of the invention is at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, or more homologous to the nucleotide sequence (e.g., to the entire length of the nucleotide sequence) shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a complement thereof.
In a preferred embodiment, the isolated nucleic acid molecule includes the nucleotide sequence shown SEQ ID NO: 1 or 3, or a complement thereof. In another embodiment, the nucleic acid molecule includes SEQ ID NO: 3 and nucleotides 1-73 of SEQ ID NO: 1. In another embodiment, the nucleic acid molecule includes SEQ ID NO:3 and nucleotides 1133-1380 of SEQ ID NO:l. In another preferred embodiment, the nucleic acid molecule consists of the nucleotide sequence shown in SEQ ID NO:l or 3. In another preferred embodiment, the nucleic acid molecule includes a fragment of at least 308 nucleotides of the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or a complement thereof. In another embodiment, an ARP nucleic acid molecule includes a nucleotide sequence encoding a protein having an amino acid sequence sufficiently homologous to the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the
DNA insert of the plasmid deposited with ATCC as Accession Number . In a preferred embodiment, an ARP nucleic acid molecule includes a nucleotide sequence encoding a protein having an amino acid sequence at least 50%, 55%, 60%, 65%, 70%, 75%>, 80%), 85%», 90%), 95%>, 98% or more homologous to the amino acid sequence of SEQ ID NO:2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number .
In another preferred embodiment, an isolated nucleic acid molecule encodes the amino acid sequence of human ARP. In yet another preferred embodiment, the nucleic acid molecule includes a nucleotide sequence encoding a protein having the amino acid sequence of SEQ ID NO: 2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number . In yet another preferred embodiment, the nucleic acid molecule is at least 308 nucleotides in length. In a further preferred embodiment, the nucleic acid molecule is at least 308 nucleotides in length and encodes a protein having an ARP activity (as described herein). Another embodiment of the invention features nucleic acid molecules, preferably ARP nucleic acid molecules, which specifically detect ARP nucleic acid molecules relative to nucleic acid molecules encoding non- ARP proteins. For example, in one embodiment, such a nucleic acid molecule is at least 308, 308-350, 350-400, 400-450, 450-500 or more nucleotides in length and hybridizes under stringent conditions to a nucleic acid molecule comprising the nucleotide sequence shown in SEQ ID NO:l, the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number , or a complement thereof. In preferred embodiments, the nucleic acid molecules are at least 15 (e.g., contiguous) nucleotides in length and hybridize under stringent conditions to nucleotides 117-139 of SEQ ID NO : 1. In other preferred embodiments, the nucleic acid molecules comprise nucleotides 117-139 of SEQ ID NO: 1.
In other preferred embodiments, the nucleic acid molecule encodes a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number , wherein the nucleic acid molecule hybridizes to a nucleic acid molecule comprising SEQ ID NO:l or SEQ ID NO: 3 under stringent conditions.
Another embodiment of the invention provides an isolated nucleic acid molecule which is antisense to an ARP nucleic acid molecule, e.g., the coding strand of an ARP nucleic acid molecule.
Another aspect of the invention provides a vector comprising an ARP nucleic acid molecule. In certain embodiments, the vector is a recombinant expression vector. In another embodiment, the invention provides a host cell containing a vector of the invention. The invention also provides a method for producing a protein, preferably an ARP protein, by culturing in a suitable medium, a host cell, e.g., a mammalian host cell such as a non-human mammalian cell, of the invention containing a recombinant expression vector, such that the protein is produced.
Another aspect of this invention features isolated or recombinant ARP proteins and polypeptides. In one embodiment, the isolated protein, preferably an ARP protein, includes at least one cysteine rich domain. In a preferred embodiment, the isolated protein, preferably an ARP protein, includes at least one cysteine rich domain and at least one EGF-like domain. In yet another preferred embodiment, the isolated protein, preferably an ARP protein, includes at least one cysteine rich domain, at least one EGF- like domain, and at least one Laminin EGF-like domain. In a preferred embodiment, the protein, preferably an ARP protein, includes at least one cysteine rich domain and has an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%), 98%) or more homologous to the amino acid sequence of SEQ ID NO:2 or the amino acid sequence encoded by the DNA insert of the plasmid deposited with ATCC as Accession Number . In another preferred embodiment, the protein, preferably an ARP protein, includes at least one cysteine rich domain and plays a role in apoptosis or programmed cell death. In yet another preferred embodiment, the protein, preferably an ARP protein, includes at least one cysteine rich domain and is encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l or SEQ ID NO:3.
In another embodiment, the invention features fragments of the protein having the amino acid sequence of SEQ ID NO:2, wherein the fragment comprises at least 15 amino acids (e.g., contiguous amino acids) of the amino acid sequence of SEQ ID NO:2 or an amino acid sequence encoded by the DNA insert of the plasmid deposited with the ATCC as Accession Number . In another embodiment, the protein, preferably an
ARP protein, has the amino acid sequence of SEQ ID NO:2.
In another embodiment, the invention features an isolated protein, preferably an ARP protein, which is encoded by a nucleic acid molecule consisting of a nucleotide sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to a nucleotide sequence of SEQ ID NO: 1 , SEQ ID NO:3, or a complement thereof. This invention further features an isolated protein, preferably an ARP protein, which is encoded by a nucleic acid molecule consisting of a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or a complement thereof. The proteins of the present invention or portions thereof, e.g., biologically active portions thereof, can be operatively linked to a non- ARP polypeptide (e.g., heterologous amino acid sequences) to form fusion proteins. The invention further features antibodies, such as monoclonal or polyclonal antibodies, that specifically bind proteins of the invention, preferably ARP proteins. In addition, the ARP proteins or biologically active portions thereof can be incorporated into pharmaceutical compositions, which optionally include pharmaceutically acceptable carriers.
In another aspect, the present invention provides a method for detecting the presence of an ARP nucleic acid molecule, protein or polypeptide in a biological sample by contacting the biological sample with an agent capable of detecting an ARP nucleic acid molecule, protein or polypeptide such that the presence of an ARP nucleic acid molecule, protein or polypeptide is detected in the biological sample.
In another aspect, the present invention provides a method for detecting the presence of ARP activity in a biological sample by contacting the biological sample with an agent capable of detecting an indicator of ARP activity such that the presence of ARP activity is detected in the biological sample.
In another aspect, the invention provides a method for modulating ARP activity comprising contacting a cell capable of expressing ARP with an agent that modulates ARP activity such that ARP activity in the cell is modulated. In one embodiment, the agent inhibits ARP activity. In another embodiment, the agent stimulates ARP activity. In one embodiment, the agent is an antibody that specifically binds to an ARP protein. In another embodiment, the agent modulates expression of ARP by modulating transcription of an ARP gene or translation of an ARP mRNA. In yet another embodiment, the agent is a nucleic acid molecule having a nucleotide sequence that is antisense to the coding strand of an ARP mRNA or an ARP gene.
In one embodiment, the methods of the present invention are used to treat a subject having a disorder characterized by aberrant ARP protein or nucleic acid expression or activity by administering an agent which is an ARP modulator to the subject. In one embodiment, the ARP modulator is an ARP protein. In another embodiment the ARP modulator is an ARP nucleic acid molecule. In yet another embodiment, the ARP modulator is a peptide, peptidomimetic, or other small molecule. In a preferred embodiment, the disorder characterized by aberrant ARP protein or nucleic acid expression is a disorder characterized by deregulated programmed cell death.
The present invention also provides a diagnostic assay for identifying the presence or absence of a genetic alteration characterized by at least one of (i) aberrant modification or mutation of a gene encoding an ARP protein; (ii) mis-regulation of the gene; and (iii) aberrant post-translational modification of an ARP protein, wherein a wild-type form of the gene encodes a protein with an ARP activity.
In another aspect the invention provides a method for identifying a compound that binds to or modulates the activity of an ARP protein, by providing an indicator composition comprising an ARP protein having ARP activity, contacting the indicator composition with a test compound, and determining the effect of the test compound on ARP activity in the indicator composition to identify a compound that modulates the activity of an ARP protein. Other features and advantages of the invention will be apparent from the following detailed description and claims.
Brief Description of the Drawings
Figure 1 depicts the cDNA sequence and predicted amino acid sequence of human ARP. The nucleotide sequence corresponds to nucleic acids 1 to 1380 of SEQ ID NO:l . The amino acid sequence corresponds to amino acids 1 to 353 of SEQ ID NO:2. The coding region without the 5' and 3' untranslated regions of the human ARP gene is shown in SEQ ID NO:3.
Figure 2 depicts a structural, hydrophobicity, and antigenicity analysis of the human ARP protein.
Figure 3 depicts the results of a search which was performed against the HMM database and which resulted in the identification of an EGF-like domain in the human ARP protein.
Figure 4 depicts the results of a search which was performed against the HMM database and which resulted in the identification of a Laminin EGF-like domain in the human ARP protein. Figure 5 depicts the results from a northern blot analysis.
Detailed Description of the Invention
The present invention is based, at least in part, on the discovery of novel Apoptosis Related Protein (ARP) family members, referred to herein as ARP protein and nucleic acid molecules. These molecules comprise a family of molecules having certain conserved structural and functional features. The term "family" when referring to the protein and nucleic acid molecules of the invention is intended to mean two or more proteins or nucleic acid molecules having a common structural domain or motif and having sufficient amino acid or nucleotide sequence homology as defined herein. Such family members can be naturally or non-naturally occurring and can be from either the same or different species. For example, a family can contain a first protein of human origin, as well as other, distinct proteins of human origin or alternatively, can contain homologues of non-human origin. Members of a family may also have common functional characteristics.
For example, the family of Apoptosis Related Proteins (ARPs), to which the ARP proteins of the present invention bear significant homology, comprise a "cysteine rich domain." As used herein, the term "cysteine rich domain" includes a protein domain having an amino acid sequence of at least about 20 amino acids of which at least about 2 amino acids are cysteine residues. Preferably, a cysteine rich domain includes at least about 30, more preferably at least about 35-40 amino acid residues, of which at least about 2, preferably at least about 3, more preferably at least about 4, 5 or 6 amino acids are cysteine residues. Cysteine-rich domains having lengths of 45-50 or 60 amino acid residues and having up to 7, 8, 9 or 10 cysteine residues are also within the scope of this invention. Cysteine rich domains are described in, for example, Lodish H. et al. Molecular Cell Biology, (Scientific American Books Inc., New York, N.Y., 1995), the contents of which are incorporated herein by reference.
In another embodiment, an ARP of the present invention is identified based on the presence of an "EGF-like domain" in the protein or corresponding nucleic acid molecule. As used herein, the term "EGF-like domain" includes a protein domain having an amino acid sequence of about 55-90 amino acid residues and having a bit score for the alignment of the sequence to the EGF-like domain (HMM) of at least 6. Preferably, an EGF-like domain includes at least about 60-85, more preferably about 65- 80 amino acid residues, or about 70-79 amino acids and has a bit score for the alignment of the sequence to the EGF-like domain (HMM) of at least 7-10, more preferably 10-30, more preferably 30-50, even more preferably 50-75, 75-100, 100-200 or greater. The EGF-like domain HMM has been assigned the PFAM Accession PF00008 (http://genome.wustl.edu/Pfam/WWWdata/EGF.html).
To identify the presence of an EGF-like domain in an ARP protein and make the determination that a protein of interest has a particular profile, the amino acid sequence of the protein is searched against a database of HMMs (e.g., the Pfam database, release 2.1) using the default parameters
(http://www.sanger.ac.uk/Software/Pfam/HMM_search). For example, the hmmsf program, which is available as part of the HMMER package of search programs, is a family specific default program for PF00435 and a score of 15 is the default threshold score for determining a hit. Alternatively, the threshold score for determining a hit can be lowered (e.g., to 8 bits). A description of the Pfam database can be found in Sonhammer et al. (1997) Proteins 28(3)405-420 and a detailed description of HMMs can be found, for example, in Gribskov et α/.(1990) Meth. Enzymol 183:146-159; Gribskov et α/.(1987) Proc. Natl Acad. Sci. USA 84:4355-4358; Krogh et α/.(1994) J Mol. Biol. 235:1501-1531; and Stultz et α/.(1993) Protein Sci. 2:305-314, the contents of which are incorporated herein by reference. A search was performed against the HMM database resulting in the identification of an EGF-like domain in the amino acid sequence of SEQ ID NO:2 (at about residues 140 to 177). The results of the search are set forth in Figure 3. Accordingly, ARP proteins having at least 50-60%> homology, preferably about
60-70%, more preferably about 70-80%, or about 80-90% homology with an EGF-like domain of human ARP are within the scope of the invention.
In another embodiment, an ARP of the present invention is identified based on the presence of a "Laminin EGF-like domain" in the protein or corresponding nucleic acid molecule. As used herein, the term "Laminin EGF-like domain" includes a protein domain having an amino acid sequence of about 30-90 amino acid residues and having a bit score for the alignment of the sequence to the Laminin EGF-like domain (HMM) of at least 6. Preferably, the Laminin EGF-like domain includes about 40-80, more preferably about 40-60 amino acid residues, or about 40-50 amino acid residues and has a bit score for the alignment of the sequence to the Laminin EGF-like domain (HMM) of at least 7-10, more preferably 10-30, more preferably 30-50, even more preferably 50- 75, 75-100, 100-200 or greater. The Laminin EGF-like domain HMM has been assigned the PFAM Accession PF00053 (http://genome.wustl.edu/Pfam/WWWdata/EGF.html). A search was performed against the HMM database, as described herein, resulting in the identification of a Laminin EGF-like domain in the amino acid sequence of SEQ ID NO:2 (at about residues 148 to 190). The results of the search are set forth in Figure 4. Accordingly, ARP proteins having at least 50-60% homology, preferably about 60-70%), more preferably about 70-80%, or about 80-90%) homology with a Laminin EGF-like domain of human ARP are within the scope of the invention.
The ARP protein of the present invention are believed to play a role in apoptosis or programmed cell death. As used herein, "programmed cell death" includes a genetically regulated process involved in the normal development of multicellular organisms. This process occurs in cells destined for removal in a variety of normal situations, including larval development of the nematode c. elegans, insect metamorphosis, development in mammalian embryos including the nephrogenic zone in the developing kidney, and regression or atrophy (e.g., in the prostrate after castration). Programmed cell death can occur in many cells following the withdrawal of growth and trophic factors, or as a result of nutritional deprivation, hormone treatment, ultraviolet irradiation, and exposure to toxic and infectious agents including reactive oxygen species and phosphatase inhibitors, e.g., okadaic acid, calcium ionphones, and a number of cancer chemotherapeutic agents. For a detailed description of programmed cell death see Trump B.F. et al. (1995) FASEB J. 9: 219-228 and Lee S. (1993) Curr. Opin. Cell Biol. 5: 286-291, the contents of which are incorporated herein by reference. Thus, the ARP proteins, by participating in a programmed cell death pathway, can modulate a programmed cell death pathway activity and provide novel diagnostic targets and therapeutic agents for disorders characterized by deregulated programmed cell death, particularly in cells that express ARP, e.g., the heart. As used herein, a "disorder characterized by deregulated programmed cell death" includes a disorder, disease or condition which is characterized by a deregulation, e.g., an upregulation or a downregulation, of programmed cell death. Programmed cell death deregulation can lead to deregulation of cellular proliferation and/or cell cycle progression. Examples of disorders characterized by deregulated programmed cell death include profilerative disorders, e.g., cancer such as chronic lymphocytic leukemia or colorectal cancer; and neurodegenerative disorders, e.g., Alzheimer's disease, dementias related to Alzheimer's disease (such as Pick's disease), Parkinson's and other Lewy diffuse body diseases, multiple sclerosis, amyotrophic lateral sclerosis, progressive supranuclear palsy, epilepsy, Jakob-Creutzfieldt disease, or AIDS related dementias. Isolated proteins of the present invention, preferably ARP proteins, have an amino acid sequence sufficiently homologous to the amino acid sequence of SEQ ID NO: 2 or are encoded by a nucleotide sequence sufficiently homologous to SEQ ID NO:l or SEQ ID NO:3. As used herein, the term "sufficiently homologous" refers to a first amino acid or nucleotide sequence which contains a sufficient or minimum number of identical or equivalent (e.g., an amino acid residue which has a similar side chain) amino acid residues or nucleotides to a second amino acid or nucleotide sequence such that the first and second amino acid or nucleotide sequences share common structural domains or motifs and/or a common functional activity. For example, amino acid or nucleotide sequences which share common structural domains have at least 30%, 40%, or 50% homology, preferably 60%) homology, more preferably 70%-80%>, and even more preferably 90-95% homology across the amino acid sequences of the domains and contain at least one and preferably two structural domains or motifs, are defined herein as sufficiently homologous. Furthermore, amino acid or nucleotide sequences which share at least 30%, 40%, or 50%, preferably 60%, more preferably 70-80%, or 90-95% homology and share a common functional activity are defined herein as sufficiently homologous.
As used interchangeably herein, an "ARP activity", "biological activity of ARP" or "functional activity of ARP", refers to an activity exerted by an ARP protein, polypeptide or nucleic acid molecule on an ARP responsive cell or on an ARP protein substrate, as determined in vivo, or in vitro, according to standard techniques. In one embodiment, an ARP activity is a direct activity, such as an association with an ARP- traget molecule. As used herein, a "target molecule" or "binding partner" is a molecule with which an ARP protein binds or interacts in nature, such that ARP -mediated function is achieved. An ARP target molecule can be a non- ARP molecule or an ARP protein or polypeptide of the present invention. In an exemplary embodiment, an ARP target molecule is an ARP ligand. Alternatively, an ARP activity is an indirect activity, such as a cellular signaling activity mediated by interaction of the ARP protein with an ARP ligand.
Accordingly, another embodiment of the invention features isolated ARP proteins and polypeptides having an ARP activity. Preferred proteins are ARP proteins having at least one cysteine rich domain and, preferably, an ARP activity. Other preferred proteins are ARP proteins having at least one cysteine rich domain, an EGF- like domain and, preferably, an ARP activity. Other preferred proteins are ARP proteins having at least one cysteine rich domain, an EGF-like domain, a Laminin EGF-like domain and, preferably, an ARP activity. Additional preferred proteins have at least one cysteine rich domain, an EGF-like domain, a Laminin EGF-like domain and are, preferably, encoded by a nucleic acid molecule having a nucleotide sequence which hybridizes under stringent hybridization conditions to a nucleic acid molecule comprising the nucleotide sequence of SEQ ID NO:l or SEQ ID NO:3. The nucleotide sequence of the isolated human ARP cDNA and the predicted amino acid sequence of the human ARP polypeptide are shown in Figure 1 and in SEQ ID NOs:l and 2, respectively. A plasmid containing the nucleotide sequence encoding human ARP was deposited with the American Type Culture Collection (ATCC), 10801 University Boulevard, Manassas, VA 20110-2209, on and assigned Accession Number . This deposit will be maintained under the terms of the Budapest
Treaty on the International Recognition of the Deposit of Microorganisms for the Purposes of Patent Procedure. This deposit was made merely as a convenience for those of skill in the art and is not an admission that a deposit is required under 35 U.S.C. §112. The human ARP gene, which is approximately 1380 nucleotides in length, encodes a protein having a molecular weight of approximately 40.5 kD and which is approximately 353 amino acid residues in length. The large isoform of the human ARP gene is expressed in all tisues tested. The small isoform of the human ARP gene is expressed mainly in the heart, the skeletal muscle, the placenta, and the pancreas.
Various aspects of the invention are described in further detail in the following subsections:
I. Isolated Nucleic Acid Molecules
One aspect of the invention pertains to isolated nucleic acid molecules that encode ARP proteins or biologically active portions thereof, as well as nucleic acid fragments sufficient for use as hybridization probes to identify ARP-encoding nucleic acid molecules (e.g., ARP mRNA) and fragments for use as PCR primers for the amplification or mutation of ARP nucleic acid molecules. As used herein, the term "nucleic acid molecule" is intended to include DNA molecules (e.g., cDNA or genomic DNA) and RNA molecules (e.g., mRNA) and analogs of the DNA or RNA generated using nucleotide analogs. The nucleic acid molecule can be single-stranded or double- stranded, but preferably is double-stranded DNA.
The term "isolated nucleic acid molecule" includes nucleic acid molecules which are separated from other nucleic acid molecules which are present in the natural source of the nucleic acid. For example, with regards to genomic DNA, the term "isolated" includes nucleic acid molecules which are separated from the chromosome with which the genomic DNA is naturally associated. Preferably, an "isolated" nucleic acid is free of sequences which naturally flank the nucleic acid (i.e., sequences located at the 5' and 3' ends of the nucleic acid) in the genomic DNA of the organism from which the nucleic acid is derived. For example, the isolated nucleic acid molecule can contain less than about 5 kb, 4kb, 3kb, 2kb, 1 kb, 0.5 kb or 0.1 kb of nucleotides which naturally flank the nucleic acid molecule in genomic DNA of the cell from which the nucleic acid is derived. Moreover, an "isolated" nucleic acid molecule, such as a cDNA molecule, can be substantially free of other cellular material, or culture medium when produced by recombinant techniques, or substantially free of chemical precursors or other chemicals when chemically synthesized. A nucleic acid molecule of the present invention, e.g., a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion thereof, can be isolated using standard molecular biology techniques and the sequence information provided herein. Using all or portion of the nucleic acid sequence of SEQ ID NO: 1 , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , as a hybridization probe, ARP nucleic acid molecules can be isolated using standard hybridization and cloning techniques (e.g., as described in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989).
Moreover, a nucleic acid molecule encompassing all or a portion of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number can be isolated by the polymerase chain reaction (PCR) using synthetic oligonucleotide primers designed based upon the sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number .
A nucleic acid of the invention can be amplified using cDNA, mRNA or alternatively, genomic DNA, as a template and appropriate oligonucleotide primers according to standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. Furthermore, oligonucleotides corresponding to ARP nucleotide sequences can be prepared by standard synthetic techniques, e.g., using an automated DNA synthesizer. In a preferred embodiment, an isolated nucleic acid molecule of the invention comprises the nucleotide sequence shown in SEQ ID NO: 1. The sequence of SEQ ID NO:l corresponds to the human ARP cDNA. This cDNA comprises sequences encoding the human ARP protein (i.e., "the coding region", from nucleotides 74-1132), as well as 5' untranslated sequences (nucleotides 1-73) and 3' untranslated sequences (nucleotides 1133-1380). Alternatively, the nucleic acid molecule can comprise only the coding region of SEQ ID NO:l (e.g., nucleotides 74-1132, corresponding to SEQ ID NO:3). In another preferred embodiment, an isolated nucleic acid molecule of the invention comprises a nucleic acid molecule which is a complement of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the
DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion of any of these nucleotide sequences. A nucleic acid molecule which is complementary to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number , is one which is sufficiently complementary to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , such that it can hybridize to the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number , thereby forming a stable duplex.
In still another preferred embodiment, an isolated nucleic acid molecule of the present invention comprises a nucleotide sequence which is at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to the entire length of the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the entire length of the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion of any of these nucleotide sequences. Moreover, the nucleic acid molecule of the invention can comprise only a portion of the nucleic acid sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , for example, a fragment which can be used as a probe or primer or a fragment encoding a portion of an ARP protein, e.g., a biologically active portion of an ARP protein. The nucleotide sequence determined from the cloning of the ARP gene allows for the generation of probes and primers designed for use in identifying and/or cloning other ARP family members, as well as ARP homologues from other species. The probe/primer typically comprises substantially purified oligonucleotide. The oligonucleotide typically comprises a region of nucleotide sequence that hybridizes under stringent conditions to at least about 12 or 15, preferably about 20 or 25, more preferably about 30, 35, 40, 45, 50, 55, 60, 65, or 75 consecutive nucleotides of a sense sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , of an anti-sense sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , or of a naturally occurring allelic variant or mutant of SEQ ID NO: 1 , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number . In an exemplary embodiment, a nucleic acid molecule of the present invention comprises a nucleotide sequence which is greater than 308, 308-350, 350-400, 400-450, 450-500, 500-550, 550-600, 600-650, 650-700, 700-750, 750-800, 800-850, 850-900, 900-950, 950-1000, or more nucleotides in length and hybridizes under stringent hybridization conditions to a nucleic acid molecule of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number .
Probes based on the ARP nucleotide sequences can be used to detect transcripts or genomic sequences encoding the same or homologous proteins. In preferred embodiments, the probe further comprises a label group attached thereto, e.g., the label group can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme co- factor. Such probes can be used as a part of a diagnostic test kit for identifying cells or tissue which misexpress an ARP protein, such as by measuring a level of an ARP- encoding nucleic acid in a sample of cells from a subject e.g., detecting ARP mRNA levels or determining whether a genomic ARP gene has been mutated or deleted.
A nucleic acid fragment encoding a "biologically active portion of an ARP protein" can be prepared by isolating a portion of the nucleotide sequence of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , which encodes a polypeptide having an ARP biological activity (the biological activities of the ARP proteins are described herein), expressing the encoded portion of the ARP protein (e.g., by recombinant expression in vitro) and assessing the activity of the encoded portion of the ARP protein. The invention further encompasses nucleic acid molecules that differ from the nucleotide sequence shown in SEQ ID NO: 1 , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , due to degeneracy of the genetic code and thus encode the same ARP proteins as those encoded by the nucleotide sequence shown in SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number . In another embodiment, an isolated nucleic acid molecule of the invention has a nucleotide sequence encoding a protein having an amino acid sequence shown in SEQ ID NO:2.
In addition to the ARP nucleotide sequences shown in SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , it will be appreciated by those skilled in the art that DNA sequence polymoφhisms that lead to changes in the amino acid sequences of the ARP proteins may exist within a population (e.g., the human population). Such genetic polymoφhism in the ARP genes may exist among individuals within a population due to natural allelic variation. As used herein, the terms "gene" and "recombinant gene" refer to nucleic acid molecules which include an open reading frame encoding an ARP protein, preferably a mammalian ARP protein, and can further include non-coding regulatory sequences, and introns.
Allelic variants of human ARP include both functional and non-functional ARP proteins. Functional allelic variants are naturally occurring amino acid sequence variants of the human ARP protein that maintain the ability to bind an ARP ligand and/or modulate programmed cell death. Functional allelic variants will typically contain only conservative substitution of one or more amino acids of SEQ ID NO:2 or substitution, deletion or insertion of non-critical residues in non-critical regions of the protein.
Non-functional allelic variants are naturally occurring amino acid sequence variants of the human ARP protein that do not have the ability to either bind an ARP ligand and/or modulate programmed cell death. Non-functional allelic variants will typically contain a non-conservative substitution, a deletion, or insertion or premature truncation of the amino acid sequence of SEQ ID NO:2 or a substitution, insertion or deletion in critical residues or critical regions. The present invention further provides non-human orthologues of the human ARP protein. Orthologues of the human ARP protein are proteins that are isolated from non-human organisms and possess the same ARP ligand binding and/or modulation of programmed cell death capabilities of the human ARP protein. Orthologues of the human ARP protein can readily be identified as including an amino acid sequence that is substantially homologous to SEQ ID NO:2, as defined herein.
Moreover, nucleic acid molecules encoding other ARP family members and, thus, which have a nucleotide sequence which differs from the ARP sequences of SEQ ID NO:l, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number are intended to be within the scope of the invention. For example, another ARP cDNA can be identified based on the nucleotide sequence of human ARP. Moreover, nucleic acid molecules encoding ARP proteins from different species, and which, thus, have a nucleotide sequence which differs from the ARP sequences of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number are intended to be within the scope of the invention. For example, a mouse ARP cDNA can be identified based on the nucleotide sequence of a human ARP.
Nucleic acid molecules corresponding to natural allelic variants and homologues of the ARP cDNAs of the invention can be isolated based on their homology to the ARP nucleic acids disclosed herein using the cDNAs disclosed herein, or a portion thereof, as a hybridization probe according to standard hybridization techniques under stringent hybridization conditions. Nucleic acid molecules corresponding to natural allelic variants and homologues of the ARP cDNAs of the invention can further be isolated by mapping to the same chromosome or locus as the ARP gene. Accordingly, in another embodiment, an isolated nucleic acid molecule of the invention is at least 15, 20, 25, 30 or more nucleotides in length and hybridizes under stringent conditions to the nucleic acid molecule comprising the nucleotide sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number . In other embodiment, the nucleic acid is at least 30, 50, 100, 150, 200, 250, 300, 308, 350, 400, 450, 500, 550, 600, 650, 700, 750, 800, 850, 900, or 950 nucleotides in length. As used herein, the term "hybridizes under stringent conditions" is intended to describe conditions for hybridization and washing under which nucleotide sequences at least 60% homologous to each other typically remain hybridized to each other. Preferably, the conditions are such that sequences at least about 70%, more preferably at least about 80%), even more preferably at least about 85%) or 90%o homologous to each other typically remain hybridized to each other. Such stringent conditions are known to those skilled in the art and can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6. A preferred, non-limiting example of stringent hybridization conditions are hybridization in 6X sodium chloride/sodium citrate (SSC) at about 45C, followed by one or more washes in 0.2 X SSC, 0.1% SDS at 50°C, preferably at 55°C, more preferably at 60°C, and even more preferably at 65 °C. Preferably, an isolated nucleic acid molecule of the invention that hybridizes under stringent conditions to the sequence of SEQ ID NOT corresponds to a naturally-occurring nucleic acid molecule. As used herein, a "naturally-occurring" nucleic acid molecule refers to an RNA or DNA molecule having a nucleotide sequence that occurs in nature (e.g., encodes a natural protein).
In addition to naturally-occurring allelic variants of the ARP sequences that may exist in the population, the skilled artisan will further appreciate that changes can be introduced by mutation into the nucleotide sequences of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number , thereby leading to changes in the amino acid sequence of the encoded ARP proteins, without altering the functional ability of the ARP proteins. For example, nucleotide substitutions leading to amino acid substitutions at "non-essential" amino acid residues can be made in the sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as
Accession Number . A "non-essential" amino acid residue is a residue that can be altered from the wild-type sequence of ARP (e.g., the sequence of SEQ ID NO:2) without altering the biological activity, whereas an "essential" amino acid residue is required for biological activity. For example, amino acid residues that are conserved among the ARP proteins of the present invention, are predicted to be particularly unamenable to alteration. Furthermore, additional amino acid residues that are conserved between the ARP proteins of the present invention and other members of the GPCR families are not likely to be amenable to alteration.
Accordingly, another aspect of the invention pertains to nucleic acid molecules encoding ARP proteins that contain changes in amino acid residues that are not essential for activity. Such ARP proteins differ in amino acid sequence from SEQ ID NO:2, yet retain biological activity. In one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence encoding a protein, wherein the protein comprises an amino acid sequence at least about 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to SEQ ID NO:2. An isolated nucleic acid molecule encoding an ARP protein homologous to the protein of SEQ ID NO:2 can be created by introducing one or more nucleotide substitutions, additions or deletions into the nucleotide sequence of SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , such that one or more amino acid substitutions, additions or deletions are introduced into the encoded protein. Mutations can be introduced into SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number by standard techniques, such as site-directed mutagenesis and PCR-mediated mutagenesis. Preferably, conservative amino acid substitutions are made at one or more predicted non-essential amino acid residues. A "conservative amino acid substitution" is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art. These families include amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, a predicted nonessential amino acid residue in an ARP protein is preferably replaced with another amino acid residue from the same side chain family. Alternatively, in another embodiment, mutations can be introduced randomly along all or part of an ARP coding sequence, such as by saturation mutagenesis, and the resultant mutants can be screened for ARP biological activity to identify mutants that retain activity. Following mutagenesis of SEQ ID NO , SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number , the encoded protein can be expressed recombinantly and the activity of the protein can be determined.
In a preferred embodiment, a mutant ARP protein can be assayed for the ability to (1) interact with a non- ARP protein molecule; (2) activate an ARP-dependent signal transduction pathway; or (3) modulate programmed cell death. In addition to the nucleic acid molecules encoding ARP proteins described above, another aspect of the invention pertains to isolated nucleic acid molecules which are antisense thereto. An "antisense" nucleic acid comprises a nucleotide sequence which is complementary to a "sense" nucleic acid encoding a protein, e.g., complementary to the coding strand of a double-stranded cDNA molecule or complementary to an mRNA sequence. Accordingly, an antisense nucleic acid can hydrogen bond to a sense nucleic acid. The antisense nucleic acid can be complementary to an entire ARP coding strand, or to only a portion thereof. In one embodiment, an antisense nucleic acid molecule is antisense to a "coding region" of the coding strand of a nucleotide sequence encoding ARP. The term "coding region" refers to the region of the nucleotide sequence comprising codons which are translated into amino acid residues (e.g., the coding region of human ARP corresponds to SEQ ID NO:3). In another embodiment, the antisense nucleic acid molecule is antisense to a "noncoding region" of the coding strand of a nucleotide sequence encoding ARP. The term "noncoding region" refers to 5' and 3' sequences which flank the coding region that are not translated into amino acids (i.e., also referred to as 5' and 3' untranslated regions).
Given the coding strand sequences encoding ARP disclosed herein (e.g., SEQ ID NO:3), antisense nucleic acids of the invention can be designed according to the rules of Watson and Crick base pairing. The antisense nucleic acid molecule can be complementary to the entire coding region of ARP mRNA, but more preferably is an oligonucleotide which is antisense to only a portion of the coding or noncoding region of ARP mRNA. For example, the antisense oligonucleotide can be complementary to the region surrounding the translation start site of ARP mRNA. An antisense oligonucleotide can be, for example, about 5, 10, 15, 20, 25, 30, 35, 40, 45 or 50 nucleotides in length. An antisense nucleic acid of the invention can be constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Examples of modified nucleotides which can be used to generate the antisense nucleic acid include 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5-iodouracil, hypoxanthine, xantine, 4-acetylcytosine, 5-(carboxyhydroxylmethyl) uracil, 5- carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1- methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2- methylguanine, 3-methylcytosine, 5-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-thiouracil, 2-thiouracil, 4-thiouracil, 5-methyluracil, uracil-5- oxyacetic acid methylester, uracil-5-oxyacetic acid (v), 5-methyl-2-thiouracil, 3-(3- amino-3-N-2-carboxypropyl) uracil, (acp3)w, and 2,6-diaminopurine. Alternatively, the antisense nucleic acid can be produced biologically using an expression vector into which a nucleic acid has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid will be of an antisense orientation to a target nucleic acid of interest, described further in the following subsection).
The antisense nucleic acid molecules of the invention are typically administered to a subject or generated in situ such that they hybridize with or bind to cellular mRNA and/or genomic DNA encoding an ARP protein to thereby inhibit expression of the protein, e.g., by inhibiting transcription and/or translation. The hybridization can be by conventional nucleotide complementarity to form a stable duplex, or, for example, in the case of an antisense nucleic acid molecule which binds to DNA duplexes, through specific interactions in the major groove of the double helix. An example of a route of administration of antisense nucleic acid molecules of the invention include direct injection at a tissue site. Alternatively, antisense nucleic acid molecules can be modified to target selected cells and then administered systemically. For example, for systemic administration, antisense molecules can be modified such that they specifically bind to receptors or antigens expressed on a selected cell surface, e.g., by linking the antisense nucleic acid molecules to peptides or antibodies which bind to cell surface receptors or antigens. The antisense nucleic acid molecules can also be delivered to cells using the vectors described herein. To achieve sufficient intracellular concentrations of the antisense molecules, vector constructs in which the antisense nucleic acid molecule is placed under the control of a strong pol II or pol III promoter are preferred.
In yet another embodiment, the antisense nucleic acid molecule of the invention is an α-anomeric nucleic acid molecule. An α-anomeric nucleic acid molecule forms specific double-stranded hybrids with complementary RNA in which, contrary to the usual β-units, the strands run parallel to each other (Gaultier et al. (1987) Nucleic Acids. Res. 15:6625-6641). The antisense nucleic acid molecule can also comprise a 2'-o- methylribonucleotide (Inoue et al. (1987) Nucleic Acids Res. 15:6131-6148) or a chimeric RNA-DNA analogue (Inoue et al. (1987) FEBS Lett. 215:327-330).
In still another embodiment, an antisense nucleic acid of the invention is a ribozyme. Ribozymes are catalytic RNA molecules with ribonuclease activity which are capable of cleaving a single-stranded nucleic acid, such as an mRNA, to which they have a complementary region. Thus, ribozymes (e.g., hammerhead ribozymes (described in Haselhoff and Gerlach (1988) Nature 334:585-591)) can be used to catalytically cleave ARP mRNA transcripts to thereby inhibit translation of ARP mRNA. A ribozyme having specificity for an ARP-encoding nucleic acid can be designed based upon the nucleotide sequence of an ARP cDNA disclosed herein (i.e., SEQ ID NOT, SEQ ID NO:3, or the nucleotide sequence of the DNA insert of the plasmid deposited with ATCC as Accession Number ). For example, a derivative of a Tetrahymena L-19 IVS RNA can be constructed in which the nucleotide sequence of the active site is complementary to the nucleotide sequence to be cleaved in an ARP- encoding mRNA. See, e.g., Cech et al. U.S. Patent No. 4,987,071 ; and Cech et al. U.S. Patent No. 5,116,742. Alternatively, ARP mRNA can be used to select a catalytic RNA having a specific ribonuclease activity from a pool of RNA molecules. See, e.g., Bartel, D. and Szostak, J.W. (1993) Science 261:1411-1418.
Alternatively, ARP gene expression can be inhibited by targeting nucleotide sequences complementary to the regulatory region of the ARP (e.g., the ARP promoter and/or enhancers) to form triple helical structures that prevent transcription of the ARP gene in target cells. See generally, Helene, C. (1991) Anticancer Drug Des. 6(6):569- 84; Helene, C. et al. (1992) Ann. N Y. Acad. Sci. 660:27-36; and Maher, L.J. (1992) Bioassays 14(12):807-15.
In yet another embodiment, the ARP nucleic acid molecules of the present invention can be modified at the base moiety, sugar moiety or phosphate backbone to improve, e.g., the stability, hybridization, or solubility of the molecule. For example, the deoxyribose phosphate backbone of the nucleic acid molecules can be modified to generate peptide nucleic acids (see Hyrup B. et al. (1996) Bioorganic & Medicinal Chemistry 4 (1): 5-23). As used herein, the terms "peptide nucleic acids" or "PNAs" refer to nucleic acid mimics, e.g., DNA mimics, in which the deoxyribose phosphate backbone is replaced by a pseudopeptide backbone and only the four natural nucleobases are retained. The neutral backbone of PNAs has been shown to allow for specific hybridization to DNA and RNA under conditions of low ionic strength. The synthesis of PNA oligomers can be performed using standard solid phase peptide synthesis protocols as described in Hyrup B. et al. (1996) supra; Perry-O'Keefe et al. Proc. Natl. Acad. Sci. 93: 14670-675. PNAs of ARP nucleic acid molecules can be used in therapeutic and diagnostic applications. For example, PNAs can be used as antisense or antigene agents for sequence-specific modulation of gene expression by, for example, inducing transcription or translation arrest or inhibiting replication. PNAs of ARP nucleic acid molecules can also be used in the analysis of single base pair mutations in a gene, (e.g., by PNA- directed PCR clamping); as 'artificial restriction enzymes' when used in combination with other enzymes, (e.g., SI nucleases (Hyrup B. (1996) supra)); or as probes or primers for DNA sequencing or hybridization (Hyrup B. et al. (1996) supra; Perry- O'Keefe supra).
In another embodiment, PNAs of ARP can be modified, (e.g., to enhance their stability or cellular uptake), by attaching lipophilic or other helper groups to PNA, by the formation of PNA-DNA chimeras, or by the use of liposomes or other techniques of drug delivery known in the art. For example, PNA-DNA chimeras of ARP nucleic acid molecules can be generated which may combine the advantageous properties of PNA and DNA. Such chimeras allow DNA recognition enzymes, (e.g., RNAse H and DNA polymerases), to interact with the DNA portion while the PNA portion would provide high binding affinity and specificity. PNA-DNA chimeras can be linked using linkers of appropriate lengths selected in terms of base stacking, number of bonds between the nucleobases, and orientation (Hyrup B. (1996) supra). The synthesis of PNA-DNA chimeras can be performed as described in Hyrup B. (1996) supra and Finn P.J. et al. (1996) Nucleic Acids Res. 24 (17): 3357-63. For example, a DNA chain can be synthesized on a solid support using standard phosphoramidite coupling chemistry and modified nucleoside analogs, e.g., 5'-(4-methoxytrityl)amino-5'-deoxy-thymidine phosphoramidite, can be used as a between the PNA and the 5' end of DNA (Mag, M. et al. (1989) Nucleic Acid Res. 17: 5973-88). PNA monomers are then coupled in a stepwise manner to produce a chimeric molecule with a 5' PNA segment and a 3' DNA segment (Finn P.J. et al. (1996) supra). Alternatively, chimeric molecules can be synthesized with a 5' DNA segment and a 3' PNA segment (Peterser, K.H. et al. (1975) Bioorganic Med. Chem. Lett. 5: 1119-11124).
In other embodiments, 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 (see, e.g., Letsinger et al. (1989) Proc. Natl. Acad. Sci. USA 86:6553-6556; Lemaitre et al. (1987) Proc. Natl. Acad. Sci. USA 84:648-652; PCT Publication No. W088/09810) or the blood-brain barrier (see, e.g., PCT Publication No. W089/10134). In addition, oligonucleotides can be modified with hybridization- triggered cleavage agents (See, e.g., Krol et al. (1988) Bio-Techniques 6:958-976) or intercalating agents. (See, e.g., Zon (1988) Pharm. Res. 5:539-549). To this end, the oligonucleotide may be conjugated to another molecule, (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, or hybridization-triggered cleavage agent).
II. Isolated ARP Proteins and Anti-ARP Antibodies One aspect of the invention pertains to isolated ARP proteins, and biologically active portions thereof, as well as polypeptide fragments suitable for use as immunogens to raise anti-ARP antibodies. In one embodiment, native ARP proteins can be isolated from cells or tissue sources by an appropriate purification scheme using standard protein purification techniques. In another embodiment, ARP proteins are produced by recombinant DNA techniques. Alternative to recombinant expression, an ARP protein or polypeptide can be synthesized chemically using standard peptide synthesis techniques.
An "isolated" or "purified" protein or biologically active portion thereof is substantially free of cellular material or other contaminating proteins from the cell or tissue source from which the ARP protein is derived, or substantially free from chemical precursors or other chemicals when chemically synthesized. The language "substantially free of cellular material" includes preparations of ARP protein in which the protein is separated from cellular components of the cells from which it is isolated or recombinantly produced. In one embodiment, the language "substantially free of cellular material" includes preparations of ARP protein having less than about 30% (by dry weight) of non- ARP protein (also referred to herein as a "contaminating protein"), more preferably less than about 20% of non- ARP protein, still more preferably less than about 10% of non- ARP protein, and most preferably less than about 5% non- ARP protein. When the ARP protein or biologically active portion thereof is recombinantly produced, it is also preferably substantially free of culture medium, i.e., culture medium represents less than about 20%, more preferably less than about 10%, and most preferably less than about 5% of the volume of the protein preparation.
The language "substantially free of chemical precursors or other chemicals" includes preparations of ARP protein in which the protein is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. In one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of ARP protein having less than about 30% (by dry weight) of chemical precursors or non- ARP chemicals, more preferably less than about 20% chemical precursors or non- ARP chemicals, still more preferably less than about 10% chemical precursors or non- ARP chemicals, and most preferably less than about 5% chemical precursors or non- ARP chemicals.
As used herein, a "biologically active portion" of an ARP protein includes a fragment of an ARP protein which participates in an interaction between an ARP molecule and a non- ARP molecule. Biologically active portions of an ARP protein include peptides comprising amino acid sequences sufficiently homologous to or derived from the amino acid sequence of the ARP protein, e.g., the amino acid sequence shown in SEQ ID NO:2, which include less amino acids than the full length ARP proteins, and exhibit at least one activity of an ARP protein. Typically, biologically active portions comprise a domain or motif with at least one activity of the ARP protein, e.g., modulating cellular programmed cell death. A biologically active portion of an ARP protein can be a polypeptide which is, for example, 10, 25, 50, 100, 200 or more amino acids in length. Biologically active portions of an ARP protein can be used as targets for developing agents which modulate an ARP mediated activity, e.g., programmed cell death.
In one embodiment, a biologically active portion of an ARP protein comprises at least one cysteine rich domain, at least one EGF-like domain, or at least one Laminin EGF-like domain. It is to be understood that a preferred biologically active portion of an ARP protein of the present invention may contain at least one of the above-identified structural domains. A more preferred biologically active portion of an ARP protein may contain at least two of the above-identified structural domains. Moreover, other biologically active portions, in which other regions of the protein are deleted, can be prepared by recombinant techniques and evaluated for one or more of the functional activities of a native ARP protein.
In a preferred embodiment, the ARP protein has an amino acid sequence shown in SEQ ID NO:2. In other embodiments, the ARP protein is substantially homologous to SEQ ID NO:2, and retains the functional activity of the protein of SEQ ID NO:2, yet differs in amino acid sequence due to natural allelic variation or mutagenesis, as described in detail in subsection I above. Accordingly, in another embodiment, the ARP protein is a protein which comprises an amino acid sequence at least about 50%, 55%>, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or more homologous to SEQ ID NO:2. To determine the percent homology of two amino acid sequences or of two nucleic acids, the sequences are aligned for optimal comparison puφoses (e.g., gaps can be introduced in the sequence of a first amino acid or nucleic acid sequence for optimal alignment with a second amino or nucleic acid sequence and non-homologous sequences can be disregarded for comparison puφoses). In a preferred embodiment, the length of a reference sequence aligned for comparison puφoses is at least 30%, preferably at least 40%, more preferably at least 50%>, even more preferably at least 60%, and even more preferably at least 70%, 80%, or 90% of the length of the reference sequence (e.g., when aligning a second sequence having 177 amino acid residues, to the ARP amino acid sequence of SEQ ID NO:2, at least 80, preferably at least 100, more preferably at least 120, even more preferably at least 140, and even more preferably at least 150, 160 or 170 amino acid residues are aligned). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are homologous at that position (i.e., as used herein amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity"). The percent homology between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % homology = # of identical positions/total # of positions x 100).
The comparison of sequences and determination of percent homology between two sequences can be accomplished using a mathematical algorithim. A preferred, non- limiting example of a mathematical algorithim utilized for the comparison of sequences is the algorithm of Karlin and Altschul (1990) Proc. Natl. Acad. Sci. USA 87:2264-68, modified as in Karlin and Altschul (1993) Proc. Natl Acad. Sci. USA 90:5873-77. Such an algorithm is incoφorated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J Mol. Biol 215:403-10. BLAST nucleotide searches can be performed with the NBLAST program, score = 100, wordlength = 12 to obtain nucleotide sequences homologous to ARP nucleic acid molecules of the invention. BLAST protein searches can be performed with the XBLAST program, score = 50, wordlength = 3 to obtain amino acid sequences homologous to ARP protein molecules of the invention. To obtain gapped alignments for comparison puφoses, Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res.
25(17):3389-3402. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. See http://www.ncbi.nlm.nih.gov. Another preferred, non-limiting example of a mathematical algorithim utilized for the comparison of sequences is the algorithm of Myers and Miller, CABIOS (1989). Such an algorithm is incoφorated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package. When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4 can be used. Additional algorithims for sequence analysis are known in the art, and include ADVANCE and ADAM, described in Torelli and Robotti (1994) Comput. Appl. Biosci. 10:3-5; and FASTA, described in Pearson and Lipman (1988) P.N.A.S. 85:2444-8.
In another preferred embodiment, the percent homology between two amino acid sequences can be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4. In yet another preferred embodiment, the percent homology between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package (available at http://www.gcg.com), using a gap weight of 50 and a length weight of 3.
The invention also provides ARP chimeric or fusion proteins. As used herein, an ARP "chimeric protein" or "fusion protein" comprises an ARP polypeptide operatively linked to a non-ARP polypeptide. An "ARP polypeptide" refers to a polypeptide having an amino acid sequence corresponding to ARP, whereas a "non-ARP polypeptide" refers to a polypeptide having an amino acid sequence corresponding to a protein which is not substantially homologous to the ARP protein, e.g., a protein which is different from the ARP protein and which is derived from the same or a different organism. Within an ARP fusion protein the ARP polypeptide can correspond to all or a portion of an ARP protein. In a preferred embodiment, an ARP fusion protein comprises at least one biologically active portion of an ARP protein. In another preferred embodiment, an ARP fusion protein comprises at least two biologically active portions of an ARP protein. Within the fusion protein, the term "operatively linked" is intended to indicate that the ARP polypeptide and the non-ARP polypeptide are fused in-frame to each other. The non-ARP polypeptide can be fused to the N-terminus or C-terminus of the ARP polypeptide.
For example, in one embodiment, the fusion protein is a GST-ARP fusion protein in which the ARP sequences are fused to the C-terminus of the GST sequences. Such fusion proteins can facilitate the purification of recombinant ARP.
In another embodiment, the fusion protein is an ARP protein containing a heterologous signal sequence at its N-terminus. In certain host cells (e.g., mammalian host cells), expression and/or secretion of ARP can be increased through use of a heterologous signal sequence. The ARP fusion proteins of the invention can be incoφorated into pharmaceutical compositions and administered to a subject in vivo. The ARP fusion proteins can be used to affect the bioavailability of an ARP substrate. Use of ARP fusion proteins may be useful therapeutically for the treatment of disorders caused by, for example, (i) aberrant modification or mutation of a gene encoding an ARP protein; (ii) mis-regulation of the ARP gene; and (iii) aberrant post-translational modification of an ARP protein.
Moreover, the ARP-fusion proteins of the invention can be used as immunogens to produce anti-ARP antibodies in a subject, to purify ARP ligands and in screening assays to identify molecules which inhibit the interaction of ARP with an ARP substrate. Preferably, an ARP chimeric or fusion protein of the invention is produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques, for example by employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992). Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST polypeptide). An ARP- encoding nucleic acid can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the ARP protein. The present invention also pertains to variants of the ARP proteins which function as either ARP agonists (mimetics) or as ARP antagonists. Variants of the ARP proteins can be generated by mutagenesis, e.g., discrete point mutation or truncation of an ARP protein. An agonist of the ARP proteins can retain substantially the same, or a subset, of the biological activities of the naturally occurring form of an ARP protein. An antagonist of an ARP protein can inhibit one or more of the activities of the naturally occurring form of the ARP protein by, for example, competitively modulating an ARP- mediated activity of an ARP protein. Thus, specific biological effects can be elicited by treatment with a variant of limited function. In one embodiment, treatment of a subject with a variant having a subset of the biological activities of the naturally occurring form of the protein has fewer side effects in a subject relative to treatment with the naturally occurring form of the ARP protein.
In one embodiment, variants of an ARP protein which function as either ARP agonists (mimetics) or as ARP antagonists can be identified by screening combinatorial libraries of mutants, e.g., truncation mutants, of an ARP protein for ARP protein agonist or antagonist activity. In one embodiment, a variegated library of ARP variants is generated by combinatorial mutagenesis at the nucleic acid level and is encoded by a variegated gene library. A variegated library of ARP variants can be produced by, for example, enzymatically ligating a mixture of synthetic oligonucleotides into gene sequences such that a degenerate set of potential ARP sequences is expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g., for phage display) containing the set of ARP sequences therein. There are a variety of methods which can be used to produce libraries of potential ARP variants from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be performed in an automatic DNA synthesizer, and the synthetic gene then ligated into an appropriate expression vector. Use of a degenerate set of genes allows for the provision, in one mixture, of all of the sequences encoding the desired set of potential ARP sequences. Methods for synthesizing degenerate oligonucleotides are known in the art (see, e.g., Narang, S.A. (1983) Tetrahedron 39:3; Itakura et al. (\9%4) Annu. Rev. Biochem. 53:323; Itakura et al. (1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477. In addition, libraries of fragments of an ARP protein coding sequence can be used to generate a variegated population of ARP fragments for screening and subsequent selection of variants of an ARP protein. In one embodiment, a library of coding sequence fragments can be generated by treating a double stranded PCR fragment of an ARP coding sequence with a nuclease under conditions wherein nicking occurs only about once per molecule, denaturing the double stranded DNA, renaturing the DNA to form double stranded DNA which can include sense/antisense pairs from different nicked products, removing single stranded portions from reformed duplexes by treatment with SI nuclease, and ligating the resulting fragment library into an expression vector. By this method, an expression library can be derived which encodes N-terminal, C-terminal and internal fragments of various sizes of the ARP protein.
Several techniques are known in the art for screening gene products of combinatorial libraries made by point mutations or truncation, and for screening cDNA libraries for gene products having a selected property. Such techniques are adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of ARP proteins. The most widely used techniques, which are amenable to high through-put analysis, for screening large gene libraries typically include cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates isolation of the vector encoding the gene whose product was detected. Recrusive ensemble mutagenesis (REM), a new technique which enhances the frequency of functional mutants in the libraries, can be used in combination with the screening assays to identify ARP variants (Arkin and Yourvan (1992) Proc. Natl. Acad. Sci. USA 59:7811-7815; Delgrave et al. (1993) Protein Engineering 6(3):327-331).
In one embodiment, cell based assays can be exploited to analyze a variegated ARP library. For example, a library of expression vectors can be transfected into a cell line which ordinarily responds to a particular ligand in an ARP-dependent manner. The transfected cells are then contacted with the ligand and the effect of expression of the mutant on signaling by the ligand can be detected, e.g., by measuring cell survival or the activity of an ARP -regulated transcription factor. Plasmid DNA can then be recovered from the cells which score for inhibition, or alternatively, potentiation of signaling by the ligand, and the individual clones further characterized.
An isolated ARP protein, or a portion or fragment thereof, can be used as an immunogen to generate antibodies that bind ARP using standard techniques for polyclonal and monoclonal antibody preparation. A full-length ARP protein can be used or, alternatively, the invention provides antigenic peptide fragments of ARP for use as immunogens. The antigenic peptide of ARP comprises at least 8 amino acid residues of the amino acid sequence shown in SEQ ID NO:2 and encompasses an epitope of ARP such that an antibody raised against the peptide forms a specific immune complex with ARP. Preferably, the antigenic peptide comprises at least 10 amino acid residues, more preferably at least 15 amino acid residues, even more preferably at least 20 amino acid residues, and most preferably at least 30 amino acid residues.
Preferred epitopes encompassed by the antigenic peptide are regions of ARP that are located on the surface of the protein, e.g., hydrophilic regions, as well as regions with high antigenicity (see, for example, Figure 2).
An ARP immunogen typically is used to prepare antibodies by immunizing a suitable subject, (e.g., rabbit, goat, mouse or other mammal) with the immunogen. An appropriate immunogenic preparation can contain, for example, recombinantly expressed ARP protein or a chemically synthesized ARP polypeptide. The preparation can further include an adjuvant, such as Freund's complete or incomplete adjuvant, or similar immunostimulatory agent. Immunization of a suitable subject with an immunogenic ARP preparation induces a polyclonal anti-ARP antibody response. Accordingly, another aspect of the invention pertains to anti-ARP antibodies. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site which specifically binds (immunoreacts with) an antigen, such as ARP. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin. The invention provides polyclonal and monoclonal antibodies that bind ARP. The term "monoclonal antibody" or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of ARP. A monoclonal antibody composition thus typically displays a single binding affinity for a particular ARP protein with which it immunoreacts.
Polyclonal anti-ARP antibodies can be prepared as described above by immunizing a suitable subject with an ARP immunogen. The anti-ARP antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized ARP. If desired, the antibody molecules directed against ARP can be isolated from the mammal (e.g., from the blood) and further purified by well known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, e.g., when the anti-ARP antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nαtwre 256:495-497) (see also, Brown et al. (1981) J Immunol. 127:539-46; Brown et al. (1980) J Biol. Chem .255:4980-83; Yeh et al. (1976) Proc. Natl. Acad. Sci. USA 76:2927-31 ; and Yeh et al. (1982) Int. J. Cancer 29:269-75), the more recent human B cell hybridoma technique (Kozbor et al. (1983) Immunol Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96) or trioma techniques. The technology for producing monoclonal antibody hybridomas is well known (see generally R. H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Coφ., New York, New York (1980); E. A. Lerner (1981) Yale J. Biol. Med, 54:387-402; M. L. Gefter et al. (1977) Somatic Cell Genet. 3:231-36). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an ARP immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds ARP.
Any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the puφose of generating an anti-ARP monoclonal antibody (see, e.g., G. Galfre et al. (1977) Nature 266:55052; Gefter et al. Somatic Cell Genet., cited supra; Lerner, Yale J. Biol. Med., cited supra; Kenneth, Monoclonal Antibodies, cited supra). Moreover, the ordinarily skilled worker will appreciate that there are many variations of such methods which also would be useful. Typically, the immortal cell line (e.g., a myeloma cell line) is derived from the same mammalian species as the lymphocytes. For example, murine hybridomas can be made by fusing lymphocytes from a mouse immunized with an immunogenic preparation of the present invention with an immortalized mouse cell line. Preferred immortal cell lines are mouse myeloma cell lines that are sensitive to culture medium containing hypoxanthine, aminopterin and thymidine ("HAT medium"). Any of a number of myeloma cell lines can be used as a fusion partner according to standard techniques, e.g., the P3-NSl/l-Ag4-l, P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines. These myeloma lines are available from ATCC. Typically, HAT-sensitive mouse myeloma cells are fused to mouse splenocytes using polyethylene glycol ("PEG"). Hybridoma cells resulting from the fusion are then selected using HAT medium, which kills unfused and unproductively fused myeloma cells (unfused splenocytes die after several days because they are not transformed). Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind ARP, e.g., using a standard ELISA assay.
Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal anti-ARP antibody can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with ARP to thereby isolate immunoglobulin library members that bind ARP. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01 ; and the Stratagene SurfZAP™ Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, Ladner et al. U.S. Patent No. 5,223,409; Kang et al. PCT International Publication No. WO 92/18619; Dower et al. PCT International Publication No. WO 91/17271; Winter et al. PCT International Publication WO 92/20791; Markland et al. PCT International Publication No. WO 92/15679; Breitling et al. PCT International Publication WO 93/01288; McCafferty et al. PCT International Publication No. WO 92/01047; Garrard et al. PCT International Publication No. WO 92/09690; Ladner et al. PCT International Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9:1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246:1275-1281; Griffiths et al. (1993) EMBO J 12:725-734; Hawkins et al. (1992) J. Mol. Biol. 226:889- 896; Clarkson et al. (1991) Nαtwre 352:624-628; Gram et al. (1992) Proc. Natl. Acad. Sci. USA 89:3576-3580; Garrad et al. (1991) Bio/Technology 9:1373-1377;
Hoogenboom et al. (1991) Nuc. Acid Res. 19:4133-4137; Barbas et al. (1991) Proc. Natl. Acad. Sci. USA 88:7978-7982; and McCafferty et al. Nαtwre (1990) 348:552-554.
Additionally, recombinant anti-ARP antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DΝA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DΝA techniques known in the art, for example using methods described in Robinson et al. International Application No. PCT/US86/02269; Akira, et al. European Patent Application 184,187; Taniguchi, M., European Patent Application 171,496; Morrison et al. European Patent Application 173,494; Neuberger et al. PCT International Publication No. WO 86/01533; Cabilly et al. U.S. Patent No. 4,816,567; Cabilly et al. European Patent Application 125,023; Better et al. (1988) Science 240:1041-1043; Liu et al. (1987) Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et al. (1987) J Immunol 139:3521-3526; Sun et al. (1987) Proc. Natl. Acad. Sci. USA 84:214-218; Nishimura et al. (1987) Cane. Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al. (1988) J Natl. Cancer Inst. 80:1553-1559); Morrison, S. L. (1985) Science 229:1202-1207; Oi et al. (1986) BioTechniques 4:214; Winter U.S. Patent 5,225,539; Jones et al. (1986) Nature 321 :552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol 141 :4053-4060.
An anti-ARP antibody (e.g., monoclonal antibody) can be used to isolate ARP by standard techniques, such as affinity chromatography or immunoprecipitation. An anti- ARP antibody can facilitate the purification of natural ARP from cells and of recombinantly produced ARP expressed in host cells. Moreover, an anti-ARP antibody can be used to detect ARP protein (e.g., in a cellular lysate or cell supernatant) in order to evaluate the abundance and pattern of expression of the ARP protein. Anti-ARP antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling (i.e., physically linking) the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, -galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include 125I, 131I, 35S or 3H.
III. Recombinant Expression Vectors and Host Cells
Another aspect of the invention pertains to vectors, preferably expression vectors, containing a nucleic acid encoding an ARP protein (or a portion thereof). As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as "expression vectors". In general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. In the present specification, "plasmid" and "vector" can be used interchangeably as the plasmid is the most commonly used form of vector. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adenoviruses and adeno- associated viruses), which serve equivalent functions.
The recombinant expression vectors of the invention comprise a nucleic acid of the invention in a form suitable for expression of the nucleic acid in a host cell, which means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cells and those which direct expression of the nucleotide sequence only in certain host cells (e.g., tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, and the like. The expression vectors of the invention can be introduced into host cells to thereby produce proteins or peptides, including fusion proteins or peptides, encoded by nucleic acids as described herein (e.g., ARP proteins, mutant forms of ARP proteins, fusion proteins, and the like). The recombinant expression vectors of the invention can be designed for expression of ARP proteins in prokaryotic or eukaryotic cells. For example, ARP proteins can be expressed in bacterial cells such as E. coli, insect cells (using baculovirus expression vectors) yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Expression of proteins in prokaryotes is most often carried out in E. coli with vectors containing constitutive or inducible promoters directing the expression of either fusion or non-fusion proteins. Fusion vectors add a number of amino acids to a protein encoded therein, usually to the amino terminus of the recombinant protein. Such fusion vectors typically serve three puφoses: 1) to increase expression of recombinant protein; 2) to increase the solubility of the recombinant protein; and 3) to aid in the purification of the recombinant protein by acting as a ligand in affinity purification. Often, in fusion expression vectors, a proteolytic cleavage site is introduced at the junction of the fusion moiety and the recombinant protein to enable separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Such enzymes, and their cognate recognition sequences, include Factor Xa, thrombin and enterokinase. Typical fusion expression vectors include pGEX (Pharmacia Biotech Inc; Smith, D.B. and Johnson, K.S. (1988) Gene 67:31-40), pMAL (New England Biolabs, Beverly, MA) and pRIT5 (Pharmacia, Piscataway, NJ) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the target recombinant protein.
Purified fusion proteins can be utilized in ARP activity assays, (e.g., direct assays or competitive assays described in detail below), or to generate antibodies specific for ARP proteins, for example. In a preferred embodiment, an ARP fusion protein expressed in a retroviral expression vector of the present invention can be utilized to infect bone marrow cells which are subsequently transplanted into irradiated recipients. The pathology of the subject recipient is then examined after sufficient time has passed (e.g., six (6) weeks).
Examples of suitable inducible non-fusion E. coli expression vectors include pTrc (Amann et al., (1988) Gene 69:301-315) and pET l id (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 60-89). Target gene expression from the pTrc vector relies on host RNA polymerase transcription from a hybrid tφ-lac fusion promoter. Target gene expression from the pET l id vector relies on transcription from a T7 gnlO-lac fusion promoter mediated by a coexpressed viral RNA polymerase (T7 gnl). This viral polymerase is supplied by host strains BL21(DE3) or HMS174(DE3) from a resident prophage harboring a T7 gnl gene under the transcriptional control of the lacUV 5 promoter.
One strategy to maximize recombinant protein expression in E. coli is to express the protein in a host bacteria with an impaired capacity to proteolytically cleave the recombinant protein (Gottesman, S., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, California (1990) 119-128). Another strategy is to alter the nucleic acid sequence of the nucleic acid to be inserted into an expression vector so that the individual codons for each amino acid are those preferentially utilized in E. coli (Wada et al., (1992) Nucleic Acids Res. 20:2111-2118). Such alteration of nucleic acid sequences of the invention can be carried out by standard DNA synthesis techniques.
In another embodiment, the ARP expression vector is a yeast expression vector. Examples of vectors for expression in yeast S. cerivisae include pYepSecl (Baldari, et al., (1987) Embo J. 6:229-234), pMFa (Kurjan and Herskowitz, (1982) Cell 30:933-
943), pJRY88 (Schultz et al, (1987) Gene 54:113-123), pYES2 (Invitrogen Coφoration, San Diego, CA), and picZ (InVitrogen Coφ, San Diego, CA).
Alternatively, ARP proteins can be expressed in insect cells using baculovirus expression vectors. Baculovirus vectors available for expression of proteins in cultured insect cells (e.g., Sf 9 cells) include the pAc series (Smith et al. (1983) Mol. Cell Biol. 3:2156-2165) and the pVL series (Lucklow and Summers (1989) Virology 170:31-39). In yet another embodiment, a nucleic acid of the invention is expressed in mammalian cells using a mammalian expression vector. Examples of mammalian expression vectors include pCDM8 (Seed, B. (1987) Nature 329:840) and pMT2PC (Kaufman et al. ( 1987) EMBO J 6: 187- 195). When used in mammalian cells, the expression vector's control functions are often provided by viral regulatory elements. For example, commonly used promoters are derived from polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40. For other suitable expression systems for both prokaryotic and eukaryotic cells see chapters 16 and 17 of Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed, Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989.
In another embodiment, the recombinant mammalian expression vector is capable of directing expression of the nucleic acid preferentially in a particular cell type (e.g., tissue-specific regulatory elements are used to express the nucleic acid). Tissue- specific regulatory elements are known in the art. Non-limiting examples of suitable tissue-specific promoters include the albumin promoter (liver-specific; Pinkert et al. (1987) Genes Dev. 1:268-277), lymphoid-specific promoters (Calame and Eaton (1988) Adv. Immunol. 43:235-275), in particular promoters of T cell receptors (Winoto and Baltimore (1989) EMBOJ. 8:729-733) and immunoglobulins (Banerji et al. (1983) Cell 33:729-740; Queen and Baltimore (1983) Cell 33:741-748), neuron-specific promoters (e.g., the neurofilament promoter; Byrne and Ruddle (1989) Proc. Natl. Acad. Sci. USA 86:5473-5477), pancreas-specific promoters (Edlund et al. (1985) Science 230:912-916), and mammary gland-specific promoters (e.g., milk whey promoter; U.S. Patent No. 4,873,316 and European Application Publication No. 264,166). Developmentally- regulated promoters are also encompassed, for example the murine hox promoters (Kessel and Grass (1990) Science 249:374-379) and the α-fetoprotein promoter (Campes and Tilghman (1989) Genes Dev. 3:537-546).
The invention further provides a recombinant expression vector comprising a DNA molecule of the invention cloned into the expression vector in an antisense orientation. That is, the DNA molecule is operatively linked to a regulatory sequence in a manner which allows for expression (by transcription of the DNA molecule) of an RNA molecule which is antisense to ARP mRNA. Regulatory sequences operatively linked to a nucleic acid cloned in the antisense orientation can be chosen which direct the continuous expression of the antisense RNA molecule in a variety of cell types, for instance viral promoters and/or enhancers, or regulatory sequences can be chosen which direct constitutive, tissue specific or cell type specific expression of antisense RNA. The antisense expression vector can be in the form of a recombinant plasmid, phagemid or attenuated virus in which antisense nucleic acids are produced under the control of a high efficiency regulatory region, the activity of which can be determined by the cell type into which the vector is introduced. For a discussion of the regulation of gene expression using antisense genes see Weintraub, H. et al., Antisense RNA as a molecular tool for genetic analysis, Reviews - Trends in Genetics, Vol. 1(1) 1986.
Another aspect of the invention pertains to host cells into which an ARP nucleic acid molecule of the invention is introduced, e.g., an ARP nucleic acid molecule within a recombinant expression vector or an ARP nucleic acid molecule in a form suitable for homologous recombination in the genome of a host cell (e.g., an ARP nucleic acid molecule which includes ARP nucleotide sequences and additional 5' and 3' flanking sequences necessary for homologous recombination). The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, an ARP protein can be expressed in bacterial cells such as E. coli, insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art.
Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing foreign nucleic acid (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, lipofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome. In order to identify and select these integrants, a gene that encodes a selectable marker (e.g., resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those which confer resistance to drags, such as G418, hygromycin and methotrexate. Nucleic acid encoding a selectable marker can be introduced into a host cell on the same vector as that encoding an ARP protein or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid can be identified by drag selection (e.g., cells that have incoφorated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) an ARP protein. Accordingly, the invention further provides methods for producing an ARP protein using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding an ARP protein has been introduced) in a suitable medium such that an ARP protein is produced. In another embodiment, the method further comprises isolating an ARP protein from the medium or the host cell. The host cells of the invention can also be used to produce non-human transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which ARP-coding sequences have been introduced. Such host cells can then be used to create non-human transgenic animals in which exogenous ARP sequences have been introduced into their genome or homologous recombinant animals in which endogenous ARP sequences have been altered. Such animals are useful for studying the function and/or activity of an ARP and for identifying and/or evaluating modulators of ARP activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens, amphibians, and the like. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, a "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous ARP gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
A transgenic animal of the invention can be created by introducing an ARP- encoding nucleic acid into the male pronuclei of a fertilized oocyte, e.g., by microinjection, retro viral infection, and allowing the oocyte to develop in a pseudopregnant female foster animal. The ARP cDNA sequence of SEQ ID NOT can be introduced as a transgene into the genome of a non-human animal. Alternatively, a nonhuman homologue of a human ARP gene, such as a mouse or rat ARP gene, can be used as a transgene. Alternatively, an ARP gene homologue, such as another GPCR family member, can be isolated based on hybridization to the ARP cDNA sequences of SEQ ID NOT, SEQ ID NO:3, or the DNA insert of the plasmid deposited with ATCC as
Accession Number (described further in subsection I above) and used as a transgene. Intronic sequences and polyadenylation signals can also be included in the transgene to increase the efficiency of expression of the transgene. A tissue-specific regulatory sequence(s) can be operably linked to an ARP transgene to direct expression of an ARP protein to particular cells. Methods for generating transgenic animals via embryo manipulation and microinjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent Nos. 4,736,866 and 4,870,009, both by Leder et al, U.S. Patent No. 4,873,191 by Wagner et al. and in Hogan, B., Manipulating the Mouse Embryo, (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Similar methods are used for production of other transgenic animals. A transgenic founder animal can be identified based upon the presence of an ARP transgene in its genome and/or expression of ARP mRNA in tissues or cells of the animals. A transgenic founder animal can then be used to breed additional animals carrying the transgene. Moreover, transgenic animals carrying a transgene encoding an ARP protein can further be bred to other transgenic animals carrying other transgenes.
To create a homologous recombinant animal, a vector is prepared which contains at least a portion of an ARP gene into which a deletion, addition or substitution has been introduced to thereby alter, e.g., functionally disrupt, the ARP gene. The ARP gene can be a human gene (e.g., the cDNA of SEQ ID NO:3), but more preferably, is a non- human homologue of a human ARP gene (e.g., a cDNA isolated by stringent hybridization with the nucleotide sequence of SEQ ID NOT). For example, a mouse ARP gene can be used to construct a homologous recombination nucleic acid molecule, e.g., a vector, suitable for altering an endogenous ARP gene in the mouse genome. In a preferred embodiment, the homologous recombination nucleic acid molecule is designed such that, upon homologous recombination, the endogenous ARP gene is functionally disrupted (i.e., no longer encodes a functional protein; also referred to as a "knock out" vector). Alternatively, the homologous recombination nucleic acid molecule can be designed such that, upon homologous recombination, the endogenous ARP gene is mutated or otherwise altered but still encodes functional protein (e.g., the upstream regulatory region can be altered to thereby alter the expression of the endogenous ARP protein). In the homologous recombination nucleic acid molecule, the altered portion of the ARP gene is flanked at its 5' and 3' ends by additional nucleotide sequence of the ARP gene to allow for homologous recombination to occur between the exogenous ARP gene carried by the homologous recombination nucleic acid molecule and an endogenous ARP gene in a cell, e.g., an embryonic stem cell. The additional flanking ARP nucleic acid sequence is of sufficient length for successful homologous recombination with the endogenous gene. Typically, several kilobases of flanking DNA (both at the 5' and 3' ends) are included in the homologous recombination nucleic acid molecule (see, e.g., Thomas, K.R. and Capecchi, M. R. (1987) Cell 51:503 for a description of homologous recombination vectors). The homologous recombination nucleic acid molecule is introduced into a cell, e.g., an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced ARP gene has homologously recombined with the endogenous ARP gene are selected (see e.g., Li, E. et al. (1992) Cell 69:915). The selected cells can then injected into a blastocyst of an animal (e.g., a mouse) to form aggregation chimeras (see e.g., Bradley, A. in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E.J. Robertson, ed. (IRL, Oxford, 1987) pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal and the embryo brought to term. Progeny harboring the homologously recombined DNA in their germ cells can be used to breed animals in which all cells of the animal contain the homologously recombined DNA by germline transmission of the transgene. Methods for constructing homologous recombination nucleic acid molecules, e.g., vectors, or homologous recombinant animals are described further in Bradley, A. (1991) Current Opinion in Biotechnology 2:823-829 and in PCT International Publication Nos.: WO 90/11354 by Le Mouellec et al.; WO 91/01140 by Smithies et al.; WO 92/0968 by Zijlstra et al; and WO 93/04169 by Bems et al.
In another embodiment, transgenic non-humans animals can be produced which contain selected systems which allow for regulated expression of the transgene. One example of such a system is the cre/loxP recombinase system of bacteriophage PI . For a description of the cre/loxP recombinase system, see, e.g., Lakso et al. (1992) Proc. Natl. Acad. Sci. USA 89:6232-6236. Another example of a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355. If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required. Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wilmut, I. et al. (1997) Nature 385:810- 813 and PCT International Publication Nos. WO 97/07668 and WO 97/07669. In brief, a cell, e.g., a somatic cell, from the transgenic animal can be isolated and induced to exit the growth cycle and enter G0 phase. The quiescent cell can then be fused, e.g., through the use of electrical pulses, to an enucleated oocyte from an animal of the same species from which the quiescent cell is isolated. The recontructed oocyte is then cultured such that it develops to morula or blastocyte and then transferred to pseudopregnant female foster animal. The offspring bome of this female foster animal will be a clone of the animal from which the cell, e.g., the somatic cell, is isolated.
IV. Pharmaceutical Compositions The ARP nucleic acid molecules, fragments of ARP proteins, and anti-ARP antibodies (also referred to herein as "active compounds") of the invention can be incoφorated into pharmaceutical compositions suitable for administration. Such compositions typically comprise the nucleic acid molecule, protein, or antibody and a pharmaceutically acceptable carrier. As used herein the language "pharmaceutically acceptable carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absoφtion delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incoφorated into the compositions.
A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic. Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF, Parsippany, NJ) or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absoφtion of the injectable compositions can be brought about by including in the composition an agent which delays absoφtion, for example, aluminum monostearate and gelatin. Sterile injectable solutions can be prepared by incoφorating the active compound (e.g., a fragment of an ARP protein or an anti-ARP antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incoφorating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the puφose of oral therapeutic administration, the active compound can be incoφorated with excipients and used in the form of tablets, troches, or capsules, oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or com starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
For administration by inhalation, the compounds are delivered in the form of an aerosol spray from pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
The compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery. In one embodiment, the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid.
Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Coφoration and Nova Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio
LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects. The data obtained from the cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.
The nucleic acid molecules of the invention can be inserted into vectors and used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Patent 5,328,470) or by stereotactic injection (see e.g., Chen et al. (1994) Proc. Natl. Acad. Sci. USA 91:3054- 3057). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.
The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.
V. Uses and Methods of the Invention
The nucleic acid molecules, proteins, protein homologues, and antibodies described herein can be used in one or more of the following methods: a) screening assays; b) predictive medicine (e.g., diagnostic assays, prognostic assays, monitoring clinical trials, and pharmacogenetics); and c) methods of treatment (e.g., therapeutic and prophylactic). As described herein, an ARP protein of the invention has one or more of the following activities: (1) it interacts with a non-ARP protein molecule on the surface of the same cell which expresses it; (2) it interacts with a non-ARP protein molecule on the surface of a different cell; (3) it activates an ARP-dependent signal transduction pathway; and (4) it modulates programmed cell death, and, thus, can be used to, for example, (1) modulate the interaction with a non-ARP protein molecule on the surface of the same cell which expresses it; (2) modulate the interaction with a non-ARP protein molecule on the surface of a different cell; (3) to activate an ARP-dependent signal transduction pathway; and (4) to modulate programmed cell death.
The isolated nucleic acid molecules of the invention can be used, for example, to express ARP protein (e.g., via a recombinant expression vector in a host cell in gene therapy applications), to detect ARP mRNA (e.g., in a biological sample) or a genetic alteration in an ARP gene, and to modulate ARP activity, as described further below. The ARP proteins can be used to treat disorders characterized by insufficient or excessive production of an ARP substrate or production of ARP inhibitors. In addition, the ARP proteins can be used to screen for naturally occurring ARP substrates, to screen for drugs or compounds which modulate ARP activity, as well as to treat disorders characterized by insufficient or excessive production of ARP protein or production of ARP protein forms which have decreased or aberrant activity compared to ARP wild type protein (e.g., disorders associated with programmed cell death). Moreover, the anti-ARP antibodies of the invention can be used to detect and isolate ARP proteins, regulate the bioavailability of ARP proteins, and modulate ARP activity.
A. Screening Assays:
The invention provides a method (also referred to herein as a "screening assay") for identifying modulators, i.e., candidate or test compounds or agents (e.g., peptides, peptidomimetics, small molecules or other drugs) which bind to ARP proteins, have a stimulatory or inhibitory effect on, for example, ARP expression or ARP activity, or have a stimulatory or inhibitory effect on, for example, the expression or activity of ARP substrate. In one embodiment, the invention provides assays for screening candidate or test compounds which are substrates of an ARP protein or polypeptide or biologically active portion thereof. In another embodiment, the invention provides assays for screening candidate or test compounds which bind to or modulate the activity of an ARP protein or polypeptide or biologically active portion thereof. The test compounds of the present invention can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to peptide libraries, while the other four approaches are applicable to peptide, non-peptide oligomer or small molecule libraries of compounds (Lam, K.S. (1997) Anticancer Drug Des. 12: 145).
Examples of methods for the synthesis of molecular libraries can be found in the art, for example in: DeWitt et al. (1993) Proc. Natl. Acad. Sci. U.S.A. 90:6909; Erb et al. (1994) Proc. Natl. Acad. Sci. USA 91:11422; Zuckermann et al. (1994). J. Med. Chem. 37:2678; Cho et al. (1993) Science 261 :1303; Carrell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2059; Carell et al. (1994) Angew. Chem. Int. Ed. Engl. 33:2061; and in Gallop et al. (1994) J Med. Chem. 37:1233. Libraries of compounds may be presented in solution (e.g., Houghten (1992)
Biotechniques 13:412-421), or on beads (Lam (1991) Nature 354:82-84), chips (Fodor (1993) Nature 364:555-556), bacteria (Ladner USP 5,223,409), spores (Ladner USP '409), plasmids (Cull et al. (1992) Proc Natl Acad Sci USA 89:1865-1869) or on phage (Scott and Smith (1990) Science 249:386-390); (Devlin (1990) Science 249:404-406); (Cwiria et al. (1990) Proc. Natl. Acad. Sci. 87:6378-6382); (Felici (1991) J Mol. Biol. 222:301-310); (Ladner supra.).
In one embodiment, an assay is a cell-based assay in which a cell which expresses an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to modulate ARP activity is determined. Determining the ability of the test compound to modulate ARP activity can be accomplished by monitoring, for example, the survival of a cell which expresses ARP or the activity of an ARP-regulated transcription factor. The cell, for example, can be of mammalian origin.
The ability of the test compound to modulate ARP binding to a substrate or to bind to ARP can also be determined. Determining the ability of the test compound to modulate ARP binding to a substrate can be accomplished, for example, by coupling the ARP substrate with a radioisotope or enzymatic label such that binding of the ARP substrate to ARP can be determined by detecting the labeled ARP substrate in a complex. Determining the ability of the test compound to bind ARP can be accomplished, for example, by coupling the compound with a radioisotope or enzymatic label such that binding of the compound to ARP can be determined by detecting the labeled ARP compound in a complex. For example, compounds (e.g., ARP substrates) can be labeled with ^^l, ^S, ^C, or -1H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting. Alternatively, compounds can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product.
It is also within the scope of this invention to determine the ability of a compound (e.g., an ARP substrate) to interact with ARP without the labeling of any of
< the interactants. For example, a microphysiometer can be used to detect the interaction of a compound with ARP without the labeling of either the compound or the ARP. McConnell, H. M. et al. (1992) Science 257:1906-1912. As used herein, a "microphysiometer" (e.g., Cytosensor) is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-addressable potentiometric sensor (LAPS). Changes in this acidification rate can be used as an indicator of the interaction between a compound and ARP.
In another embodiment, an assay is a cell-based assay comprising contacting a cell expressing an ARP target molecule (e.g., an ARP substrate) with a test compound and determining the ability of the test compound to modulate (e.g. stimulate or inhibit) the activity of the ARP target molecule. Determining the ability of the test compound to modulate the activity of an ARP target molecule can be accomplished, for example, by determining the ability of the ARP protein to bind to or interact with the ARP target molecule.
Determining the ability of the ARP protein or a biologically active fragment thereof, to bind to or interact with an ARP target molecule can be accomplished by one of the methods described above for determining direct binding. In a preferred embodiment, determining the ability of the ARP protein to bind to or interact with an ARP target molecule can be accomplished by determining the activity of the target molecule. For example, the activity of the target molecule can be determined by detecting induction of a cellular second messenger of the target (i.e., intracellular Ca2+, diacylglycerol, IP3, and the like), detecting catalytic/enzymatic activity of the target an appropriate substrate, detecting the induction of a reporter gene (comprising a target- responsive regulatory element operatively linked to a nucleic acid encoding a detectable marker, e.g., luciferase), or detecting a target-regulated cellular response.
In yet another embodiment, an assay of the present invention is a cell-free assay in which an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to bind to the ARP protein or biologically active portion thereof is determined. Preferred biologically active portions of the ARP proteins to be used in assays of the present invention include fragments which participate in interactions with non-ARP molecules, e.g., fragments with high surface probability scores (see, for example, Figure 2). Binding of the test compound to the ARP protein can be determined either directly or indirectly as described above. In a preferred embodiment, the assay includes contacting the ARP protein or biologically active portion thereof with a known compound which binds ARP to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with an ARP protein, wherein determining the ability of the test compound to interact with an ARP protein comprises determining the ability of the test compound to preferentially bind to ARP or biologically active portion thereof as compared to the known compound.
In another embodiment, the assay is a cell-free assay in which an ARP protein or biologically active portion thereof is contacted with a test compound and the ability of the test compound to modulate (e.g., stimulate or inhibit) the activity of the ARP protein or biologically active portion thereof is determined. Determining the ability of the test compound to modulate the activity of an ARP protein can be accomplished, for example, by determining the ability of the ARP protein to bind to an ARP target molecule by one of the methods described above for determining direct binding. Determining the ability of the ARP protein to bind to an ARP target molecule can also be accomplished using a technology such as real-time Biomolecular Interaction Analysis (BIA). Sjolander, S. and Urbaniczky, C. (1991) Anal. Chem. 63:2338-2345 and Szabo et al. (1995) Curr. Opin. Struct. Biol. 5:699-705. As used herein, "BIA" is a technology for studying biospecific interactions in real time, without labeling any of the interactants (e.g., BIAcore). Changes in the optical phenomenon of surface plasmon resonance
(SPR) can be used as an indication of real-time reactions between biological molecules.
In an alternative embodiment, determining the ability of the test compound to modulate the activity of an ARP protein can be accomplished by determining the ability of the ARP protein to further modulate the activity of a downstream effector of an ARP target molecule. For example, the activity of the effector molecule on an appropriate target can be determined or the binding of the effector to an appropriate target can be determined as previously described.
In yet another embodiment, the cell-free assay involves contacting an ARP protein or biologically active portion thereof with a known compound which binds the ARP protein to form an assay mixture, contacting the assay mixture with a test compound, and determining the ability of the test compound to interact with the ARP protein, wherein determining the ability of the test compound to interact with the ARP protein comprises determining the ability of the ARP protein to preferentially bind to or modulate the activity of an ARP target molecule. The cell-free assays of the present invention are amenable to use of both soluble and/or membrane-bound forms of isolated proteins (e.g., ARP proteins or biologically active portions thereof). In the case of cell-free assays in which a membrane-bound form of an isolated protein (e.g., the large ARP isoform) is used it may be desirable to utilize a solubilizing agent such that the membrane-bound form of the isolated protein is maintained in solution. Examples of such solubilizing agents include non-ionic detergents such as n-octylglucoside, n-dodecylglucoside, n-dodecylmaltoside, octanoyl- N-methylglucamide, decanoyl-N-methylglucamide, Triton® X-100, Triton® X-l 14, Thesit®, Isotridecypoly(ethylene glycol ether)n, 3-[(3- cholamidopropyl)dimethylamminio]-l -propane sulfonate (CHAPS), 3-[(3- cholamidopropyl)dimethylamminio]-2-hydroxy-l -propane sulfonate (CHAPSO), or N- dodecyl=N,N-dimethyl-3-ammonio-l -propane sulfonate.
In more than one embodiment of the above assay methods of the present invention, it may be desirable to immobilize either ARP or its target molecule to facilitate separation of complexed from uncomplexed forms of one or both of the proteins, as well as to accommodate automation of the assay. Binding of a test compound to an ARP protein, or interaction of an ARP protein with a target molecule in the presence and absence of a candidate compound, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein can be provided which adds a domain that allows one or both of the proteins to be bound to a matrix. For example, glutathione-S-transferase/ ARP fusion proteins or glutathione-S- transferase/target fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the test compound or the test compound and either the non-adsorbed target protein or ARP protein, and the mixture incubated under conditions conducive to complex formation (e.g., at physiological conditions for salt and pH). Following incubation, the beads or microtitre plate wells are washed to remove any unbound components, the matrix immobilized in the case of beads, complex determined either directly or indirectly, for example, as described above. Alternatively, the complexes can be dissociated from the matrix, and the level of ARP binding or activity determined using standard techniques.
Other techniques for immobilizing proteins on matrices can also be used in the screening assays of the invention. For example, either an ARP protein or an ARP target molecule can be immobilized utilizing conjugation of biotin and streptavidin. Biotinylated ARP protein or target molecules can be prepared from biotin-NHS (N- hydroxy-succinimide) using techniques known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical). Alternatively, antibodies reactive with ARP protein or target molecules but which do not interfere with binding of the ARP protein to its target molecule can be derivatized to the wells of the plate, and unbound target or ARP protein trapped in the wells by antibody conjugation. Methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the ARP protein or target molecule, as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the ARP protein or target molecule.
In another embodiment, modulators of ARP expression are identified in a method wherein a cell is contacted with a candidate compound and the expression of ARP mRNA or protein in the cell is determined. The level of expression of ARP mRNA or protein in the presence of the candidate compound is compared to the level of expression of ARP mRNA or protein in the absence of the candidate compound. The candidate compound can then be identified as a modulator of ARP expression based on this comparison. For example, when expression of ARP mRNA or protein is greater (statistically significantly greater) in the presence of the candidate compound than in its absence, the candidate compound is identified as a stimulator of ARP mRNA or protein expression. Alternatively, when expression of ARP mRNA or protein is less (statistically significantly less) in the presence of the candidate compound than in its absence, the candidate compound is identified as an inhibitor of ARP mRNA or protein expression. The level of ARP mRNA or protein expression in the cells can be determined by methods described herein for detecting ARP mRNA or protein.
In yet another aspect of the invention, the ARP proteins can be used as "bait proteins" in a two-hybrid assay or three-hybrid assay (see, e.g., U.S. Patent No. 5,283,317; Zervos et al. (1993) Cell 72:223-232; Madura et al. (1993) J Biol. Chem.
268:12046-12054; Bartel et al. (1993) Biotechniques 14:920-924; Iwabuchi et al. (1993) Oncogene 8:1693-1696; and Brent WO94/10300), to identify other proteins, which bind to or interact with ARP ("ARP-binding proteins" or "ARP-bp") and are involved in ARP activity. Such ARP-binding proteins are also likely to be involved in the propagation of signals by the ARP proteins or ARP targets as, for example, downstream elements of an ARP-mediated signaling pathway. Alternatively, such ARP-binding proteins are likely to be ARP inhibitors.
The two-hybrid system is based on the modular nature of most transcription factors, which consist of separable DNA-binding and activation domains. Briefly, the assay utilizes two different DNA constracts. In one construct, the gene that codes for an ARP protein is fused to a gene encoding the DNA binding domain of a known transcription factor (e.g., GAL-4). In the other constract, a DNA sequence, from a library of DNA sequences, that encodes an unidentified protein ("prey" or "sample") is fused to a gene that codes for the activation domain of the known transcription factor. If the "bait" and the "prey" proteins are able to interact, in vivo, forming an ARP- dependent complex, the DNA-binding and activation domains of the transcription factor are brought into close proximity. This proximity allows transcription of a reporter gene (e.g., LacZ) which is operably linked to a transcriptional regulatory site responsive to the transcription factor. Expression of the reporter gene can be detected and cell colonies containing the functional transcription factor can be isolated and used to obtain the cloned gene which encodes the protein which interacts with the ARP protein.
This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model. For example, an agent identified as described herein (e.g., an ARP modulating agent, an antisense ARP nucleic acid molecule, an ARP-specific antibody, or an ARP-binding partner) can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent. Furthermore, this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.
B. Detection Assays
Portions or fragments of the cDNA sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. These applications are described in the subsections below.
1. Chromosome Mapping
Once the sequence (or a portion of the sequence) of a gene has been isolated, this sequence can be used to map the location of the gene on a chromosome. This process is called chromosome mapping. Accordingly, portions or fragments of the ARP nucleotide sequences, described herein, can be used to map the location of the ARP genes on a chromosome. The mapping of the ARP sequences to chromosomes is an important first step in correlating these sequences with genes associated with disease.
Briefly, ARP genes can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp in length) from the ARP nucleotide sequences. Computer analysis of the ARP sequences can be used to predict primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers can then be used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the ARP sequences will yield an amplified fragment. Somatic cell hybrids are prepared by fusing somatic cells from different mammals (e.g., human and mouse cells). As hybrids of human and mouse cells grow and divide, they gradually lose human chromosomes in random order, but retain the mouse chromosomes. By using media in which mouse cells cannot grow, because they lack a particular enzyme, but human cells can, the one human chromosome that contains the gene encoding the needed enzyme, will be retained. By using various media, panels of hybrid cell lines can be established. Each cell line in a panel contains either a single human chromosome or a small number of human chromosomes, and a full set of mouse chromosomes, allowing easy mapping of individual genes to specific human chromosomes. (D'Eustachio P. et al. (1983) Science 220:919-924). Somatic cell hybrids containing only fragments of human chromosomes can also be produced by using human chromosomes with translocations and deletions. PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular sequence to a particular chromosome. Three or more sequences can be assigned per day using a single thermal cycler. Using the ARP nucleotide sequences to design oligonucleotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes. Other mapping strategies which can similarly be used to map an ARP sequence to its chromosome include in situ hybridization (described in Fan, Y. et al. (1990) Proc. Natl. Acad. Sci. USA, 87:6223-27), pre- screening with labeled flow-sorted chromosomes, and pre-selection by hybridization to chromosome specific cDNA libraries. Fluorescence in situ hybridization (FISH) of a DNA sequence to a metaphase chromosomal spread can further be used to provide a precise chromosomal location in one step. Chromosome spreads can be made using cells whose division has been blocked in metaphase by a chemical such as colcemid that disrupts the mitotic spindle. The chromosomes can be treated briefly with trypsin, and then stained with Giemsa. A pattern of light and dark bands develops on each chromosome, so that the chromosomes can be identified individually. The FISH technique can be used with a DNA sequence as short as 500 or 600 bases. However, clones larger than 1,000 bases have a higher likelihood of binding to a unique chromosomal location with sufficient signal intensity for simple detection. Preferably 1 ,000 bases, and more preferably 2,000 bases will suffice to get good results at a reasonable amount of time. For a review of this technique, see Verma et al., Human Chromosomes: A Manual of Basic Techniques (Pergamon Press, New York 1988).
Reagents for chromosome mapping can be used individually to mark a single chromosome or a single site on that chromosome, or panels of reagents can be used for marking multiple sites and/or multiple chromosomes. Reagents corresponding to noncoding regions of the genes actually are preferred for mapping puφoses. Coding sequences are more likely to be conserved within gene families, thus increasing the chance of cross hybridizations during chromosomal mapping.
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. (Such data are found, for example, in V. McKusick, Mendelian Inheritance in Man, available on-line through Johns Hopkins University Welch Medical Library). The relationship between a gene and a disease, mapped to the same chromosomal region, can then be identified through linkage analysis (co-inheritance of physically adjacent genes), described in, for example, Egeland, J. et al. (1987) Nature, 325:783-787. Moreover, differences in the DNA sequences between individuals affected and unaffected with a disease associated with the ARP gene, can be determined. If a mutation is observed in some or all of the affected individuals but not in any unaffected individuals, then the mutation is likely to be the causative agent of the particular disease. Comparison of affected and unaffected individuals generally involves first looking for structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR based on that DNA sequence. Ultimately, complete sequencing of genes from several individuals can be performed to confirm the presence of a mutation and to distinguish mutations from polymoφhisms.
2. Tissue Typing
The ARP sequences of the present invention can also be used to identify individuals from minute biological samples. The United States military, for example, is considering the use of restriction fragment length polymoφhism (RFLP) for identification of its personnel. In this technique, an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identification. This method does not suffer from the current limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult. The sequences of the present invention are useful as additional DNA markers for RFLP (described in U.S. Patent 5,272,057). Furthermore, the sequences of the present invention can be used to provide an alternative technique which determines the actual base-by-base DNA sequence of selected portions of an individual's genome. Thus, the ARP nucleotide sequences described herein can be used to prepare two PCR primers from the 5' and 3' ends of the sequences. These primers can then be used to amplify an individual's DNA and subsequently sequence it. Panels of corresponding DNA sequences from individuals, prepared in this manner, can provide unique individual identifications, as each individual will have a unique set of such DNA sequences due to allelic differences. The sequences of the present invention can be used to obtain such identification sequences from individuals and from tissue. The ARP nucleotide sequences of the invention uniquely represent portions of the human genome. Allelic variation occurs to some degree in the coding regions of these sequences, and to a greater degree in the noncoding regions. It is estimated that allelic variation between individual humans occurs with a frequency of about once per each 500 bases. Each of the sequences described herein can, to some degree, be used as a standard against which DNA from an individual can be compared for identification puφoses. Because greater numbers of polymoφhisms occur in the noncoding regions, fewer sequences are necessary to differentiate individuals. The noncoding sequences of SEQ ID NOT can comfortably provide positive individual identification with a panel of perhaps 10 to 1,000 primers which each yield a noncoding amplified sequence of 100 bases. If predicted coding sequences, such as that in SEQ ID NO:3 is used, a more appropriate number of primers for positive individual identification would be 500-2,000.
If a panel of reagents from ARP nucleotide sequences described herein is used to generate a unique identification database for an individual, those same reagents can later be used to identify tissue from that individual. Using the unique identification database, positive identification of the individual, living or dead, can be made from extremely small tissue samples.
3. Use of Partial ARP Sequences in Forensic Biology DNA-based identification techniques can also be used in forensic biology.
Forensic biology is a scientific field employing genetic typing of biological evidence found at a crime scene as a means for positively identifying, for example, a peφetrator of a crime. To make such an identification, PCR technology can be used to amplify DNA sequences taken from very small biological samples such as tissues, e.g., hair or skin, or body fluids, e.g., blood, saliva, or semen found at a crime scene. The amplified sequence can then be compared to a standard, thereby allowing identification of the origin of the biological sample.
The sequences of the present invention can be used to provide polynucleotide reagents, e.g., PCR primers, targeted to specific loci in the human genome, which can enhance the reliability of DNA-based forensic identifications by, for example, providing another "identification marker" (i.e. another DNA sequence that is unique to a particular individual). As mentioned above, actual base sequence information can be used for identification as an accurate alternative to patterns formed by restriction enzyme generated fragments. Sequences targeted to noncoding regions of SEQ ID NOT are particularly appropriate for this use as greater numbers of polymoφhisms occur in the noncoding regions, making it easier to differentiate individuals using this technique. Examples of polynucleotide reagents include the ARP nucleotide sequences or portions thereof, e.g., fragments derived from the noncoding regions of SEQ ID NOT having a length of at least 20 bases, preferably at least 30 bases. The ARP nucleotide sequences described herein can further be used to provide polynucleotide reagents, e.g., labeled or labelable probes which can be used in, for example, an in situ hybridization technique, to identify a specific tissue, e.g., brain tissue. This can be very useful in cases where a forensic pathologist is presented with a tissue of unknown origin. Panels of such ARP probes can be used to identify tissue by species and or by organ type.
In a similar fashion, these reagents, e.g., ARP primers or probes can be used to screen tissue culture for contamination (i.e. screen for the presence of a mixture of different types of cells in a culture).
C. Predictive Medicine:
The present invention also pertains to the field of predictive medicine in which diagnostic assays, prognostic assays, and monitoring clinical trials are used for prognostic (predictive) puφoses to thereby treat an individual prophylactically.
Accordingly, one aspect of the present invention relates to diagnostic assays for determining ARP protein and/or nucleic acid expression as well as ARP activity, in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or disorder, or is at risk of developing a disorder, associated with aberrant ARP expression or activity. The invention also provides for prognostic (or predictive) assays for determining whether an individual is at risk of developing a disorder associated with ARP protein, nucleic acid expression or activity. For example, mutations in an ARP gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive puφose to thereby phophylactically treat an individual prior to the onset of a disorder characterized by or associated with ARP protein, nucleic acid expression or activity.
Another aspect of the invention pertains to monitoring the influence of agents (e.g., drugs, compounds) on the expression or activity of ARP in clinical trials.
These and other agents are described in further detail in the following sections.
1. Diagnostic Assays
An exemplary method for detecting the presence or absence of ARP protein or nucleic acid in a biological sample involves obtaining a biological sample from a test subject and contacting the biological sample with a compound or an agent capable of detecting ARP protein or nucleic acid (e.g., mRNA, genomic DNA) that encodes ARP protein such that the presence of ARP protein or nucleic acid is detected in the biological sample. A preferred agent for detecting ARP mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to ARP mRNA or genomic DNA. The nucleic acid probe can be, for example, a full-length ARP nucleic acid, such as the nucleic acid of SEQ ID NOT, SEQ ID NO:3, or the DNA insert of the plasmid deposited with ATCC as Accession Number , or a portion thereof, such as an oligonucleotide of at least
15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to ARP mRNA or genomic DNA. Other suitable probes for use in the diagnostic assays of the invention are described herein.
A preferred agent for detecting ARP protein is an antibody capable of binding to ARP protein, preferably an antibody with a detectable label. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g., Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin. The term "biological sample" is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. That is, the detection method of the invention can be used to detect ARP mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo. For example, in vitro techniques for detection of ARP mRNA include Northern hybridizations and in situ hybridizations. In vitro techniques for detection of ARP protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofluorescence. In vitro techniques for detection of ARP genomic DNA include Southern hybridizations. Furthermore, in vivo techniques for detection of ARP protein include introducing into a subject a labeled anti-ARP antibody. For example, the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
In one embodiment, the biological sample contains protein molecules from the test subject. Alternatively, the biological sample can contain mRNA molecules from the test subject or genomic DNA molecules from the test subject. A preferred biological sample is a serum sample isolated by conventional means from a subject.
In another embodiment, the methods further involve obtaining a control biological sample from a control subject, contacting the control sample with a compound or agent capable of detecting ARP protein, mRNA, or genomic DNA, such that the presence of ARP protein, mRNA or genomic DNA is detected in the biological sample, and comparing the presence of ARP protein, mRNA or genomic DNA in the control sample with the presence of ARP protein, mRNA or genomic DNA in the test sample.
The invention also encompasses kits for detecting the presence of ARP in a biological sample. For example, the kit can comprise a labeled compound or agent capable of detecting ARP protein or mRNA in a biological sample; means for determining the amount of ARP in the sample; and means for comparing the amount of ARP in the sample with a standard. The compound or agent can be packaged in a suitable container. The kit can further comprise instructions for using the kit to detect ARP protein or nucleic acid.
2. Prognostic Assays
The diagnostic methods described herein can furthermore be utilized to identify subjects having or at risk of developing a disease or disorder associated with aberrant ARP expression or activity. As used herein, the term "aberrant" includes an ARP expression or activity which deviates from the wild type ARP expression or activity. Aberrant expression or activity includes increased or decreased expression or activity, as well as expression or activity which does not follow the wild type developmental pattern of expression or the subcellular pattern of expression. For example, aberrant ARP expression or activity is intended to include the cases in which a mutation in the ARP gene causes the ARP gene to be under-expressed or over-expressed and situations in which such mutations result in a non-functional ARP protein or a protein which does not function in a wild-type fashion, e.g., a protein which does not interact with an ARP ligand or one which interacts with a non-ARP ligand.
The assays described herein, such as the preceding diagnostic assays or the following assays, can be utilized to identify a subject having or at risk of developing a disorder associated with a misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder. Alternatively, the prognostic assays can be utilized to identify a subject having or at risk for developing a disorder associated with a misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder. Thus, the present invention provides a method for identifying a disease or disorder associated with aberrant ARP expression or activity in which a test sample is obtained from a subject and ARP protein or nucleic acid (e.g., mRNA or genomic DNA) is detected, wherein the presence of ARP protein or nucleic acid is diagnostic for a subject having or at risk of developing a disease or disorder associated with aberrant ARP expression or activity. As used herein, a "test sample" refers to a biological sample obtained from a subject of interest. For example, a test sample can be a biological fluid (e.g., serum), cell sample, or tissue.
Furthermore, the prognostic assays described herein can be used to determine whether a subject can be administered an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate) to treat a disease or disorder associated with aberrant ARP expression or activity. For example, such methods can be used to determine whether a subject can be effectively treated with an agent for a proliferative disorder, a differentiative disorder, or a gland associated disorder. Thus, the present invention provides methods for determining whether a subject can be effectively treated with an agent for a disorder associated with aberrant ARP expression or activity in which a test sample is obtained and ARP protein or nucleic acid expression or activity is detected (e.g., wherein the abundance of ARP protein or nucleic acid expression or activity is diagnostic for a subject that can be administered the agent to treat a disorder associated with aberrant ARP expression or activity).
The methods of the invention can also be used to detect genetic alterations in an ARP gene, thereby determining if a subject with the altered gene is at risk for a disorder characterized by misregulation in ARP protein activity or nucleic acid expression, such as a proliferative disorder, a differentiative disorder, or a gland associated disorder. In preferred embodiments, the methods include detecting, in a sample of cells from the subject, the presence or absence of a genetic alteration characterized by at least one of an alteration affecting the integrity of a gene encoding an ARP -protein, or the mis- expression of the ARP gene. For example, such genetic alterations can be detected by ascertaining the existence of at least one of 1) a deletion of one or more nucleotides from an ARP gene; 2) an addition of one or more nucleotides to an ARP gene; 3) a substitution of one or more nucleotides of an ARP gene, 4) a chromosomal rearrangement of an ARP gene; 5) an alteration in the level of a messenger RNA transcript of an ARP gene, 6) aberrant modification of an ARP gene, such as of the methylation pattern of the genomic DNA, 7) the presence of a non- ild type splicing pattern of a messenger RNA transcript of an ARP gene, 8) a non-wild type level of an ARP -protein, 9) allelic loss of an ARP gene, and 10) inappropriate post-translational modification of an ARP -protein. As described herein, there are a large number of assay known in the art which can be used for detecting alterations in an ARP gene. A preferred biological sample is a tissue or serum sample isolated by conventional means from a subject. In certain embodiments, detection of the alteration involves the use of a probe/primer in a polymerase chain reaction (PCR) (see, e.g., U.S. Patent Nos. 4,683,195 and 4,683,202), such as anchor PCR or RACE PCR, or, alternatively, in a ligation chain reaction (LCR) (see, e.g., Landegran et al. (1988) Science 241:1077-1080; and Nakazawa et al. (1994) Proc. Natl. Acad. Sci. USA 91 :360-364), the latter of which can be particularly useful for detecting point mutations in the ARP-gene (see Abravaya et al. (1995) Nucleic Acids Res .23:675-682). This method can include the steps of collecting a sample of cells from a subject, isolating nucleic acid (e.g., genomic, mRNA or both) from the cells of the sample, contacting the nucleic acid sample with one or more primers which specifically hybridize to an ARP gene under conditions such that hybridization and amplification of the ARP-gene (if present) occurs, and detecting the presence or absence of an amplification product, or detecting the size of the amplification product and comparing the length to a control sample. It is anticipated that PCR and/or LCR may be desirable to use as a preliminary amplification step in conjunction with any of the techniques used for detecting mutations described herein. Alternative amplification methods include: self sustained sequence replication
(Guatelli, J.C. et al., (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878). transcriptional amplification system (Kwoh, D.Y. et al., (1989) Proc. Natl Acad. Sci. USA 86:1173- 1177), Q-Beta Replicase (Lizardi, P.M. et al. (1988) Bio-Technology 6:1197), or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. These detection schemes are especially useful for the detection of nucleic acid molecules if such molecules are present in very low numbers.
In an alternative embodiment, mutations in an ARP gene from a sample cell can be identified by alterations in restriction enzyme cleavage patterns. For example, sample and control DNA is isolated, amplified (optionally), digested with one or more restriction endonucleases, and fragment length sizes are determined by gel electrophoresis and compared. Differences in fragment length sizes between sample and control DNA indicates mutations in the sample DNA. Moreover, the use of sequence specific ribozymes (see, for example, U.S. Patent No. 5,498,531) can be used to score for the presence of specific mutations by development or loss of a ribozyme cleavage site.
In other embodiments, genetic mutations in ARP can be identified by hybridizing a sample and control nucleic acids, e.g., DNA or RNA, to high density arrays containing hundreds or thousands of oligonucleotides probes (Cronin, M.T. et al. (1996) Human Mutation 7: 244-255; Kozal, M.J. et al. (1996) Nature Medicine 2: 753- 759). For example, genetic mutations in ARP can be identified in two dimensional arrays containing light-generated DNA probes as described in Cronin, M.T. et al. supra. Briefly, a first hybridization array of probes can be used to scan through long stretches of DNA in a sample and control to identify base changes between the sequences by making linear arrays of sequential overlapping probes. This step allows the identification of point mutations. This step is followed by a second hybridization array that allows the characterization of specific mutations by using smaller, specialized probe arrays complementary to all variants or mutations detected. Each mutation array is composed of parallel probe sets, one complementary to the wild-type gene and the other complementary to the mutant gene. In yet another embodiment, any of a variety of sequencing reactions known in the art can be used to directly sequence the ARP gene and detect mutations by comparing the sequence of the sample ARP with the corresponding wild-type (control) sequence. Examples of sequencing reactions include those based on techniques developed by Maxam and Gilbert ((1977) Proc. Natl. Acad. Sci. USA 74:560) or Sanger ((1977) Proc. Natl. Acad. Sci. USA 74:5463). It is also contemplated that any of a variety of automated sequencing procedures can be utilized when performing the diagnostic assays ((1995) Biotechniques 19:448), including sequencing by mass spectrometry (see, e.g., PCT International Publication No. WO 94/16101; Cohen et al. (1996) Adv. Chromatogr. 36:127-162; and Griffin et al. (1993) Appl. Biochem. Biotechnol. 38:147-159). Other methods for detecting mutations in the ARP gene include methods in which protection from cleavage agents is used to detect mismatched bases in RNA/RNA or RNA/DNA heteroduplexes (Myers et al. (1985) Science 230:1242). In general, the art technique of "mismatch cleavage" starts by providing heteroduplexes of formed by hybridizing (labeled) RNA or DNA containing the wild-type ARP sequence with potentially mutant RNA or DNA obtained from a tissue sample. The double-stranded duplexes are treated with an agent which cleaves single-stranded regions of the duplex such as which will exist due to basepair mismatches between the control and sample strands. For instance, RNA/DNA duplexes can be treated with RNase and DNA/DNA hybrids treated with SI nuclease to enzymatically digesting the mismatched regions. In other embodiments, either DNA/DNA or RNA/DNA duplexes can be treated with hydroxylamine or osmium tetroxide and with piperidine in order to digest mismatched regions. After digestion of the mismatched regions, the resulting material is then separated by size on denaturing polyacrylamide gels to determine the site of mutation. See, for example, Cotton et al. (1988) Proc. Natl Acad Sci USA 85 :4397; Saleeba et al. (1992) Methods Enzymol 217:286-295. In a preferred embodiment, the control DNA or RNA can be labeled for detection.
In still another embodiment, the mismatch cleavage reaction employs one or more proteins that recognize mismatched base pairs in double-stranded DNA (so called "DNA mismatch repair" enzymes) in defined systems for detecting and mapping point mutations in ARP cDNAs obtained from samples of cells. For example, the mutY enzyme of E. coli cleaves A at G/A mismatches and the thymidine DNA glycosylase from HeLa cells cleaves T at G/T mismatches (Hsu et al. (1994) Carcinogenesis 15:1657-1662). According to an exemplary embodiment, a probe based on an ARP sequence, e.g., a wild-type ARP sequence, is hybridized to a cDNA or other DNA product from a test cell(s). The duplex is treated with a DNA mismatch repair enzyme, and the cleavage products, if any, can be detected from electrophoresis protocols or the like. See, for example, U.S. Patent No. 5,459,039.
In other embodiments, alterations in electrophoretic mobility will be used to identify mutations in ARP genes. For example, single strand conformation polymoφhism (SSCP) may be used to detect differences in electrophoretic mobility between mutant and wild type nucleic acids (orita et al. (1989) Proc Natl. Acad. Sci USA: 86:2766, see also Cotton (1993) Mutat. Res. 285:125-144; and Hayashi (1992) Genet. Anal. Tech. Appl. 9:73-79). Single-stranded DNA fragments of sample and control ARP nucleic acids will be denatured and allowed to renature. The secondary structure of single-stranded nucleic acids varies according to sequence, the resulting alteration in electrophoretic mobility enables the detection of even a single base change. The DNA fragments may be labeled or detected with labeled probes. The sensitivity of the assay may be enhanced by using RNA (rather than DNA), in which the secondary structure is more sensitive to a change in sequence. In a preferred embodiment, the subject method utilizes heteroduplex analysis to separate double stranded heteroduplex molecules on the basis of changes in electrophoretic mobility (Keen et al. (1991) Trends Genet 7:5).
In yet another embodiment the movement of mutant or wild-type fragments in polyacrylamide gels containing a gradient of denaturant is assayed using denaturing gradient gel electrophoresis (DGGE) (Myers et al. ( 1985) Nature 313:495). When
DGGE is used as the method of analysis, DNA will be modified to insure that it does not completely denature, for example by adding a GC clamp of approximately 40 bp of high-melting GC-rich DNA by PCR. In a further embodiment, a temperature gradient is used in place of a denaturing gradient to identify differences in the mobility of control and sample DNA (Rosenbaum and Reissner (1987) Biophys Chem 265:12753).
Examples of other techniques for detecting point mutations include, but are not limited to, selective oligonucleotide hybridization, selective amplification, or selective primer extension. For example, oligonucleotide primers may be prepared in which the known mutation is placed centrally and then hybridized to target DNA under conditions which permit hybridization only if a perfect match is found (Saiki et al. (1986) Nature 324:163); Saiki et al. (1989) Proc. Natl Acad. Sci USA 86:6230). Such allele specific oligonucleotides are hybridized to PCR amplified target DNA or a number of different mutations when the oligonucleotides are attached to the hybridizing membrane and hybridized with labeled target DNA. Altematively, allele specific amplification technology which depends on selective PCR amplification may be used in conjunction with the instant invention. Oligonucleotides used as primers for specific amplification may carry the mutation of interest in the center of the molecule (so that amplification depends on differential hybridization) (Gibbs et al. (1989) Nucleic Acids Res. 17:2437-2448) or at the extreme 3' end of one primer where, under appropriate conditions, mismatch can prevent, or reduce polymerase extension (Prossner (1993) Tibtech 11 :238). In addition it may be desirable to introduce a novel restriction site in the region of the mutation to create cleavage-based detection (Gasparini et al. (1992) Mol. Cell Probes 6:1). It is anticipated that in certain embodiments amplification may also be performed using Taq ligase for amplification (Barany (1991) Proc. Natl. Acad. Sci USA 88:189). In such cases, ligation will occur only if there is a perfect match at the 3' end of the 5' sequence making it possible to detect the presence of a known mutation at a specific site by looking for the presence or absence of amplification. The methods described herein may be performed, for example, by utilizing prepackaged diagnostic kits comprising at least one probe nucleic acid or antibody reagent described herein, which may be conveniently used, e.g., in clinical settings to diagnose patients exhibiting symptoms or family history of a disease or illness involving an ARP gene. Furthermore, any cell type or tissue in which ARP is expressed may be utilized in the prognostic assays described herein.
3. Monitoring of Effects During Clinical Trials
Monitoring the influence of agents (e.g., drugs) on the expression or activity of an ARP protein (e.g., the modulation of cellular programmed cell death) can be applied not only in basic drag screening, but also in clinical trials. For example, the effectiveness of an agent determined by a screening assay as described herein to increase ARP gene expression, protein levels, or upregulate ARP activity, can be monitored in clinical trials of subjects exhibiting decreased ARP gene expression, protein levels, or downregulated ARP activity. Alternatively, the effectiveness of an agent determined by a screening assay to decrease ARP gene expression, protein levels, or downregulate ARP activity, can be monitored in clinical trials of subjects exhibiting increased ARP gene expression, protein levels, or upregulated ARP activity. In such clinical trials, the expression or activity of an ARP gene, and preferably, other genes that have been implicated in, for example, an ARP-associated disorder can be used as a "read out" or markers of the phenotype of a particular cell.
For example, and not by way of limitation, genes, including ARP, that are modulated in cells by treatment with an agent (e.g., compound, drug or small molecule) which modulates ARP activity (e.g., identified in a screening assay as described herein) can be identified. Thus, to study the effect of agents on ARP-associated disorders (e.g., proliferative disorders, differentiative disorders, or gland associated disorders), for example, in a clinical trial, cells can be isolated and RNA prepared and analyzed for the levels of expression of ARP and other genes implicated in the ARP-associated disorder, respectively. The levels of gene expression (e.g., a gene expression pattern) can be quantified by northern blot analysis or RT-PCR, as described herein, or alternatively by measuring the amount of protein produced, by one of the methods as described herein, or by measuring the levels of activity of ARP or other genes. In this way, the gene expression pattern can serve as a marker, indicative of the physiological response of the cells to the agent. Accordingly, this response state may be determined before, and at various points during treatment of the individual with the agent. In a preferred embodiment, the present invention provides a method for monitoring the effectiveness of treatment of a subject with an agent (e.g., an agonist, antagonist, peptidomimetic, protein, peptide, nucleic acid, small molecule, or other drag candidate identified by the screening assays described herein) including the steps of (i) obtaining a pre-administration sample from a subject prior to administration of the agent; (ii) detecting the level of expression of an ARP protein, mRNA, or genomic DNA in the preadministration sample; (iii) obtaining one or more post-administration samples from the subject; (iv) detecting the level of expression or activity of the ARP protein, mRNA, or genomic DNA in the post-administration samples; (v) comparing the level of expression or activity of the ARP protein, mRNA, or genomic DNA in the pre- administration sample with the ARP protein, mRNA, or genomic DNA in the post administration sample or samples; and (vi) altering the administration of the agent to the subject accordingly. For example, increased administration of the agent may be desirable to increase the expression or activity of ARP to higher levels than detected, i.e., to increase the effectiveness of the agent. Alternatively, decreased administration of the agent may be desirable to decrease expression or activity of ARP to lower levels than detected, i.e. to decrease the effectiveness of the agent. According to such an embodiment, ARP expression or activity may be used as an indicator of the effectiveness of an agent, even in the absence of an observable phenotypic response.
C. Methods of Treatment: The present invention provides for both prophylactic and therapeutic methods of treating a subject at risk of (or susceptible to) a disorder or having a disorder associated with aberrant ARP expression or activity. With regards to both prophylactic and therapeutic methods of treatment, such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics. "Pharmacogenomics", as used herein, refers to the application of genomics technologies such as gene sequencing, statistical genetics, and gene expression analysis to drugs in clinical development and on the market. More specifically, the term refers the study of how a patient's genes determine his or her response to a drag (e.g., a patient's "drag response phenotype", or "drag response genotype".) Thus, another aspect of the invention provides methods for tailoring an individual's prophylactic or therapeutic treatment with either the ARP molecules of the present invention or ARP modulators according to that individual's drag response genotype. Pharmacogenomics allows a clinician or physician to target prophylactic or therapeutic treatments to patients who will most benefit from the treatment and to avoid treatment of patients who will experience toxic drag-related side effects.
1. Prophylactic Methods
In one aspect, the invention provides a method for preventing in a subject, a disease or condition associated with an aberrant ARP expression or activity, by administering to the subject an ARP or an agent which modulates ARP expression or at least one ARP activity. Subjects at risk for a disease which is caused or contributed to by aberrant ARP expression or activity can be identified by, for example, any or a combination of diagnostic or prognostic assays as described herein. Administration of a prophylactic agent can occur prior to the manifestation of symptoms characteristic of the ARP aberrancy, such that a disease or disorder is prevented or, alternatively, delayed in its progression. Depending on the type of ARP aberrancy, for example, an ARP, ARP agonist or ARP antagonist agent can be used for treating the subject. The appropriate agent can be determined based on screening assays described herein.
2. Therapeutic Methods Another aspect of the invention pertains to methods of modulating ARP expression or activity for therapeutic puφoses. Accordingly, in an exemplary embodiment, the modulatory method of the invention involves contacting a cell with an ARP or agent that modulates one or more of the activities of ARP protein activity associated with the cell. An agent that modulates ARP protein activity can be an agent as described herein, such as a nucleic acid or a protein, a naturally-occurring target molecule of an ARP protein (e.g., an ARP substrate), an ARP antibody, an ARP agonist or antagonist, a peptidomimetic of an ARP agonist or antagonist, or other small molecule. In one embodiment, the agent stimulates one or more ARP activities. Examples of such stimulatory agents include active ARP protein and a nucleic acid molecule encoding ARP that has been introduced into the cell. In another embodiment, the agent inhibits one or more ARP activities. Examples of such inhibitory agents include antisense ARP nucleic acid molecules, anti-ARP antibodies, and ARP inhibitors. These modulatory methods can be performed in vitro (e.g., by culturing the cell with the agent) or, alternatively, in vivo (e.g., by administering the agent to a subject). As such, the present invention provides methods of treating an individual afflicted with a disease or disorder characterized by aberrant expression or activity of an ARP protein or nucleic acid molecule. In one embodiment, the method involves administering an agent (e.g., an agent identified by a screening assay described herein), or combination of agents that modulates (e.g., upregulates or downregulates) ARP expression or activity. In another embodiment, the method involves administering an ARP protein or nucleic acid molecule as therapy to compensate for reduced or aberrant ARP expression or activity. Stimulation of ARP activity is desirable in situations in which ARP is abnormally downregulated and/or in which increased ARP activity is likely to have a beneficial effect. For example, stimulation of ARP activity is desirable in situations in which an ARP is downregulated and/or in which increased ARP activity is likely to have a beneficial effect. Likewise, inhibition of ARP activity is desirable in situations in which ARP is abnormally upregulated and/or in which decreased ARP activity is likely to have a beneficial effect.
3. Pharmacogenomics The ARP molecules of the present invention, as well as agents, or modulators which have a stimulatory or inhibitory effect on ARP activity (e.g., ARP gene expression) as identified by a screening assay described herein can be administered to individuals to treat (prophylactically or therapeutically) ARP-associated disorders (e.g., programmed cell death associated disorders) associated with aberrant ARP activity. In conjunction with such treatment, pharmacogenomics (i.e., the study of the relationship between an individual's genotype and that individual's response to a foreign compound or drug) may be considered. Differences in metabolism of therapeutics can lead to severe toxicity or therapeutic failure by altering the relation between dose and blood concentration of the pharmacologically active drug. Thus, a physician or clinician may consider applying knowledge obtained in relevant pharmacogenomics studies in determining whether to administer an ARP molecule or ARP modulator as well as tailoring the dosage and/or therapeutic regimen of treatment with an ARP molecule or ARP modulator.
Pharmacogenomics deals with clinically significant hereditary variations in the response to drags due to altered drug disposition and abnormal action in affected persons. See, for example, Eichelbaum, M. et al. (1996) Clin. Exp. Pharmacol. Physiol 23(10-11) :983-985 and Linder, M.W. et al. (1997) Clin. Chem. 43(2):254-266. In general, two types of pharmacogenetic conditions can be differentiated. Genetic conditions transmitted as a single factor altering the way drags act on the body (altered drag action) or genetic conditions transmitted as single factors altering the way the body acts on drags (altered drug metabolism). These pharmacogenetic conditions can occur either as rare genetic defects or as naturally-occurring polymoφhisms. For example, glucose-6-phosphate dehydrogenase deficiency (G6PD) is a common inherited enzymopathy in which the main clinical complication is haemolysis after ingestion of oxidant drags (anti-malarials, sulfonamides, analgesics, nitrofurans) and consumption of fava beans.
One pharmacogenomics approach to identifying genes that predict drag response, known as "a genome-wide association", relies primarily on a high-resolution map of the human genome consisting of already known gene-related markers (e.g., a "bi- allelic" gene marker map which consists of 60,000-100,000 polymoφhic or variable sites on the human genome, each of which has two variants.) Such a high-resolution genetic map can be compared to a map of the genome of each of a statistically significant number of patients taking part in a Phase II/III drag trial to identify markers associated with a particular observed drug response or side effect. Alternatively, such a high resolution map can be generated from a combination of some ten-million known single nucleotide polymoφhisms (SNPs) in the human genome. As used herein, a "SNP" is a common alteration that occurs in a single nucleotide base in a stretch of DNA. For example, a SNP may occur once per every 1000 bases of DNA. A SNP may be involved in a disease process, however, the vast majority may not be disease- associated. Given a genetic map based on the occurrence of such SNPs, individuals can be grouped into genetic categories depending on a particular pattern of SNPs in their individual genome. In such a manner, treatment regimens can be tailored to groups of genetically similar individuals, taking into account traits that may be common among such genetically similar individuals.
Alternatively, a method termed the "candidate gene approach", can be utilized to identify genes that predict drag response. According to this method, if a gene that encodes a drags target is known (e.g., an ARP protein of the present invention), all common variants of that gene can be fairly easily identified in the population and it can be determined if having one version of the gene versus another is associated with a particular drag response. As an illustrative embodiment, the activity of drug metabolizing enzymes is a major determinant of both the intensity and duration of drug action. The discovery of genetic polymoφhisms of drug metabolizing enzymes (e.g., N-acetyltransferase 2 (NAT 2) and cytochrome P450 enzymes CYP2D6 and CYP2C19) has provided an explanation as to why some patients do not obtain the expected drug effects or show exaggerated drag response and serious toxicity after taking the standard and safe dose of a drag. These polymoφhisms are expressed in two phenotypes in the population, the extensive metabolizer (EM) and poor metabolizer (PM). The prevalence of PM is different among different populations. For example, the gene coding for CYP2D6 is highly polymoφhic and several mutations have been identified in PM, which all lead to the absence of functional CYP2D6. Poor metabolizers of CYP2D6 and CYP2C19 quite frequently experience exaggerated drag response and side effects when they receive standard doses. If a metabolite is the active therapeutic moiety, PM show no therapeutic response, as demonstrated for the analgesic effect of codeine mediated by its CYP2D6-formed metabolite moφhine. The other extreme are the so called ultra-rapid metabolizers who do not respond to standard doses. Recently, the molecular basis of ultra-rapid metabolism has been identified to be due to CYP2D6 gene amplification.
Alternatively, a method termed the "gene expression profiling", can be utilized to identify genes that predict drag response. For example, the gene expression of an animal dosed with a drag (e.g., an ARP molecule or ARP modulator of the present invention) can give an indication whether gene pathways related to toxicity have been turned on.
Information generated from more than one of the above pharmacogenomics approaches can be used to determine appropriate dosage and treatment regimens for prophylactic or therapeutic treatment an individual. This knowledge, when applied to dosing or drug selection, can avoid adverse reactions or therapeutic failure and thus enhance therapeutic or prophylactic efficiency when treating a subject with an ARP molecule or ARP modulator, such as a modulator identified by one of the exemplary screening assays described herein. This invention is further illustrated by the following examples which should not be construed as limiting. The contents of all references, patents and published patent applications cited throughout this application are incoφorated herein by reference.
EXAMPLES
EXAMPLE 1: IDENTIFICATION AND CHARACTERIZATION OF ARP cDNA
In this example, the identification and characterization of the gene encoding human ARP (also referred to as MHT) is described.
Isolation of the human ARP cDNA
The invention is based, at least in part, on the discovery of a human gene encoding a novel protein, referred to herein as ARP. The human ARP was isolated from a human heart cDNA library purchased from Clonetch Marathon. Positive clones were isolated and subsequently 5' RACE-PCR was used to obtain the full length clone.
The sequence of the entire human clone was determined and found to contain an open reading frame of 353 amino acids termed human "ARP." The nucleotide sequence encoding the human ARP protein is shown in Figure 1 and is set forth as SEQ ID NOT . The full length protein encoded by this nucleic acid comprises about 353 amino acids and has the amino acid sequence shown in Figure 1 and set forth as SEQ ID NO:2. The coding region (open reading frame) of SEQ ID NOT is set forth as SEQ ID NO:3. Clone MHT, comprising the entire coding region of human ARP was deposited with the American Type Culture Collection (ATCC®), 10801 University Boulevard, Manassas, VA 20110-2209, on , and assigned Accession No. .
Analysis of the Human ARP Molecule
A BLASTX 1.4 search, using a score of 100 and a word length of 3 (Altschul et al. (1990) J. Mol Biol. 215:403) of the translated nucleotide sequence of human ARP revealed that ARP is similar to the Cricetulus griseus HT protein (Accession Number U48852). The ARP protein is 80%> identical to the Cricetulus griseus HT protein (Accession Number U48852) over translated nucleotides 70 to 558.
A BLASTN 1.4.9 search, using a score of 100 and a word length of 12 (Altschul et al. (1990) J. Mol. Biol. 215:403) of the nucleotide sequence of human ARP revealed that ARP is similar to the Homo sapiens cDNA clone 668306 (Accession Number AA243887). The ARP nucleic acid molecule is 99%> identical to the Homo sapiens cDNA clone 668306 (Accession Number AA243887) over nucleotides 755 to 1062. A search using the human ARP protein sequence was performed against the HMM database resulting in the identification of an EGF-like domain in the amino acid sequence of SEQ ID NO:2 (residuesl40 to 177). The results of the search are set forth in Figure 3. Using the same search, a Laminin EGF-like domain was also identified in the amino acid sequence of SEQ ID NO:2 (residuesl48 to 190). The results of this search are set forth in Figure 4.
Tissue Distribution of ARP mRNA
This Example describes the tissue distribution of ARP mRNA, as determined by Northern blot hybridization.
Northern blot hybridizations with the various RNA samples were performed under standard conditions and washed under stringent conditions, i.e., 0.2xSSC at 65°C. The DNA probe was radioactively labeled with 32p.dCTP using the Prime-It kit (Stratagene, La Jolla, CA) according to the instructions of the supplier. Filters containing human mRNA (MultiTissue Northern I and MultiTissue Northern II from Clontech, Palo Alto, CA) were probed in ExpressHyb hybridization solution (Clontech) and washed at high stringency according to manufacturer's recommendations. Two isoforms were identified using this northern blot analysis, suggesting one form maybe a splice varient. One isoform (larger message) could be a transmembrane protein (frizzled-like) and the other isoform could be the secreted form (smaller message). The two isoforms show a clear pattern of tissue specificity. On the multiple tissue blot from Clonetech, the large transcript is found in almost all tissues whereas the smaller message is expressed mainly in the heart, skeletal muscle placenta and the pancreas (see Figure 5). EXAMPLE 2: EXPRESSION OF RECOMBINANT ARP
PROTEIN IN BACTERIAL CELLS
In this example, ARP is expressed as a recombinant glutathione-S-transferase
(GST) fusion polypeptide in E. coli and the fusion polypeptide is isolated and characterized. Specifically, ARP is fused to GST and this fusion polypeptide is expressed in E. coli, e.g., strain PEB199. Expression of the GST- ARP fusion protein in PEB199 is induced with IPTG. The recombinant fusion polypeptide is purified from crade bacterial lysates of the induced PEB199 strain by affinity chromatography on glutathione beads. Using polyacrylamide gel electrophoretic analysis of the polypeptide purified from the bacterial lysates, the molecular weight of the resultant fusion polypeptide is determined.
EXAMPLE 3: EXPRESSION OF RECOMBINANT ARP
PROTEIN IN COS CELLS
To express the ARP gene in COS cells, the pcDNA/Amp vector by Invitrogen Coφoration (San Diego, CA) is used. This vector contains an SV40 origin of replication, an ampicillin resistance gene, an E. coli replication origin, a CMV promoter followed by a polylinker region, and an SV40 intron and polyadenylation site. A DNA fragment encoding the entire ARP protein and an HA tag (Wilson et al. (1984) Cell 37:767) or a FLAG tag fused in-frame to its 3' end of the fragment is cloned into the polylinker region of the vector, thereby placing the expression of the recombinant protein under the control of the CMV promoter.
To constract the plasmid, the ARP DNA sequence is amplified by PCR using two primers. The 5' primer contains the restriction site of interest followed by approximately twenty nucleotides of the ARP coding sequence starting from the initiation codon; the 3' end sequence contains complementary sequences to the other restriction site of interest, a translation stop codon, the HA tag or FLAG tag and the last 20 nucleotides of the ARP coding sequence. The PCR amplified fragment and the pCDNA/Amp vector are digested with the appropriate restriction enzymes and the vector is dephosphorylated using the CIAP enzyme (New England Biolabs, Beverly, MA). Preferably the two restriction sites chosen are different so that the ARP gene is inserted in the correct orientation. The ligation mixture is transformed into E. coli cells (strains HB101, DH5a, SURE, available from Stratagene Cloning Systems, La Jolla, CA, can be used), the transformed culture is plated on ampicillin media plates, and resistant colonies are selected. Plasmid DNA is isolated from transformants and examined by restriction analysis for the presence of the correct fragment.
COS cells are subsequently transfected with the ARP-pcDNA/Amp plasmid DNA using the calcium phosphate or calcium chloride co-precipitation methods, DEAE- dextran-mediated transfection, lipofection, or electroporation. Other suitable methods for transfecting host cells can be found in Sambrook, J., Fritsh, E. F., and Maniatis, T. Molecular Cloning: A Laboratory Manual. 2nd, ed., Cold Spring Harbor Laboratory, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. The expression of the ARP polypeptide is detected by radiolabelling (35S-methionine or 35S-cysteine available from NEN, Boston, MA, can be used) and immunoprecipitation (Harlow, E. and Lane, D. Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1988) using an HA specific monoclonal antibody. Briefly, the cells are labelled for 8 hours with 35S-methionine (or 35S-cysteine). The culture media are then collected and the cells are lysed using detergents (RIPA buffer, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% DOC, 50 mM Tris, pH 7.5). Both the cell lysate and the culture media are precipitated with an HA specific monoclonal antibody. Precipitated polypeptides are then analyzed by SDS-PAGE.
Alternatively, DNA containing the ARP coding sequence is cloned directly into the polylinker of the pCDNA/Amp vector using the appropriate restriction sites. The resulting plasmid is transfected into COS cells in the manner described above, and the expression of the ARP polypeptide is detected by radiolabelling and immunoprecipitation using an ARP specific monoclonal antibody. Equivalents
Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

What is claimed:
1. An isolated nucleic acid molecule selected from the group consisting of:
(a) a nucleic acid molecule comprising the nucleotide sequence set forth in SEQ ID NOT; and
(b) a nucleic acid molecule comprising the nucleotide sequence set forth in SEQ ID NO:3.
2. An isolated nucleic acid molecule which encodes a polypeptide comprising the amino acid sequence set forth in SEQ ID NO:2.
3. An isolated nucleic acid molecule comprising the nucleotide sequence contained in the plasmid deposited with ATCC® as Accession Number .
4. An isolated nucleic acid molecule which encodes a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:2.
5. An isolated nucleic acid molecule selected from the group consisting of: a) a nucleic acid molecule comprising a nucleotide sequence which is at least 60%> homologous to the nucleotide sequence of SEQ ID NO: 1 , 3, or a complement thereof; b) a nucleic acid molecule comprising a fragment of at least 308 nucleotides of a nucleic acid comprising the nucleotide sequence of SEQ ID NOT, 3, or a complement thereof; c) a nucleic acid molecule which encodes a polypeptide comprising an amino acid sequence at least about 60% homologous to the amino acid sequence of
SEQ ID NO:2; and d) a nucleic acid molecule which encodes a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:2, wherein the fragment comprises at least 15 contiguous amino acid residues of the amino acid sequence of SEQ
ID NO:2.
6. An isolated nucleic acid molecule which hybridizes to the nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5 under stringent conditions.
7. An isolated nucleic acid molecule comprising a nucleotide sequence which is complementary to the nucleotide sequence of the nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5.
8. An isolated nucleic acid molecule comprising the nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5, and a nucleotide sequence encoding a heterologous polypeptide.
9. A vector comprising the nucleic acid molecule of any one of claims 1 , 2, 3, 4, or 5.
10. The vector of claim 9, which is an expression vector.
11. A host cell transfected with the expression vector of claim 9.
12. A method of producing a polypeptide comprising culturing a host cell transfected with the expression vector of claim 9 in an appropriate culture medium to, thereby, produce the polypeptide.
13. An isolated polypeptide selected from the group consisting of: a) a fragment of a polypeptide comprising the amino acid sequence of SEQ ID NO:2, wherein the fragment comprises at least 15 contiguous amino acids of SEQ ID NO:2; b) a naturally occurring allelic variant of a polypeptide comprising the amino acid sequence of SEQ ID NO:2, wherein the polypeptide is encoded by a nucleic acid molecule which hybridizes to a nucleic acid molecule comprising SEQ ID NOT or 3 under stringent conditions; c) a polypeptide which is encoded by a nucleic acid molecule comprising a nucleotide sequence which is at least 60 % homologous to a nucleic acid comprising the nucleotide sequence of SEQ ID NOT or 3; d) a polypeptide comprising an amino acid sequence which is at least 60% homologous to the amino acid sequence of SEQ ID NO:2.
14. The isolated polypeptide of claim 13 comprising the amino acid sequence of SEQ ID NO:2.
15. The polypeptide of claim 13, further comprising heterologous amino acid sequences.
16. An antibody which selectively binds to a polypeptide of claim 13.
17. A method for detecting the presence of a polypeptide of claim 13 in a sample comprising: a) contacting the sample with a compound which selectively binds to the polypeptide; and b) determining whether the compound binds to the polypeptide in the sample to thereby detect the presence of a polypeptide of claim 13 in the sample.
18. The method of claim 17, wherein the compound which binds to the polypeptide is an antibody.
19. A kit comprising a compound which selectively binds to a polypeptide of claim 13 and instructions for use.
20. A method for detecting the presence of a nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5 in a sample comprising: a) contacting the sample with a nucleic acid probe or primer which selectively hybridizes to the nucleic acid molecule; and b) determining whether the nucleic acid probe or primer binds to a nucleic acid molecule in the sample to thereby detect the presence of a nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5 in the sample.
21. The method of claim 20, wherein the sample comprises mRNA molecules and is contacted with a nucleic acid probe.
22. A kit comprising a compound which selectively hybridizes to a nucleic acid molecule of any one of claims 1, 2, 3, 4, or 5 and instructions for use.
23. A method for identifying a compound which binds to a polypeptide of claim 13 comprising: a) contacting the polypeptide, or a cell expressing the polypeptide with a test compound; and b) determining whether the polypeptide binds to the test compound.
24. The method of claim 23, wherein the binding of the test compound to the polypeptide is detected by a method selected from the group consisting of: a) detection of binding by direct detection of test compound/polypeptide binding; b) detection of binding using a competition binding assay; and c) detection ofbinding using an assay for ARP activity.
25. A method for modulating the activity of a polypeptide of claim 13 comprising contacting the polypeptide or a cell expressing the polypeptide with a compound which binds to the polypeptide in a sufficient concentration to modulate the activity of the polypeptide.
26. A method for identifying a compound which modulates the activity of a polypeptide of claim 13 comprising: a) contacting a polypeptide of claim 13 with a test compound; and b) determining the effect of the test compound on the activity of the polypeptide to thereby identify a compound which modulates the activity of the polypeptide
PCT/US1999/022270 1998-09-24 1999-09-24 Apoptosis related protein and uses therefor WO2000017236A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU65005/99A AU6500599A (en) 1998-09-24 1999-09-24 Novel apoptosis related proteins and uses therefor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US16062398A 1998-09-24 1998-09-24
US09/160,623 1998-09-24

Publications (3)

Publication Number Publication Date
WO2000017236A2 true WO2000017236A2 (en) 2000-03-30
WO2000017236A3 WO2000017236A3 (en) 2000-05-25
WO2000017236A8 WO2000017236A8 (en) 2000-07-20

Family

ID=22577668

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/022270 WO2000017236A2 (en) 1998-09-24 1999-09-24 Apoptosis related protein and uses therefor

Country Status (2)

Country Link
AU (1) AU6500599A (en)
WO (1) WO2000017236A2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999014327A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Genes amplified in tumours, antibodies against the proteins coded thereby, and their use in diagnosis and treatment of cancer

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999014327A2 (en) * 1997-09-17 1999-03-25 Genentech, Inc. Genes amplified in tumours, antibodies against the proteins coded thereby, and their use in diagnosis and treatment of cancer

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DATABASE EMBL - EMEST29 [Online] Entry HSAA42749, Acc.no. AA242749, 19 March 1997 (1997-03-19) HILLIER, L. ET AL.: "zr56g02.s1 Soares NhHMPu S1 Homo sapiens cDNA clone 668306 3' similar to TR:G1216486 G1216486 HT protein." XP002132887 *
DATABASE EMBL - EMEST31 [Online] Entry HSZZ12080, Acc.no. AA306943, 18 April 1997 (1997-04-18) ADAMS, M.D. ET AL.: "EST177865 Jurkat T-cells VI Homo sapiens cDNA 5' end similar to similar to adhesive plaque protein matrix." XP002132888 *
DATABASE EMBL - EMROD [Online] Entry CG48852, Acc.no. U48852, 18 March 1996 (1996-03-18) CHEN, H. ET AL.: "Cricetulus griseus HT protein mRNA, complete cds." XP002131426 *

Also Published As

Publication number Publication date
WO2000017236A8 (en) 2000-07-20
AU6500599A (en) 2000-04-10
WO2000017236A3 (en) 2000-05-25

Similar Documents

Publication Publication Date Title
WO2000029577A9 (en) Novel members of the capsaicin/vanilloid receptor family of proteins and uses thereof
US20030232378A1 (en) Novel toll molecules and uses therefor
US6197551B1 (en) Spoil-1 protein and nucleic acid molecules and uses therefor
WO2000005346A1 (en) Novel potassium channel molecules and uses therefor
US6225085B1 (en) LRSG protein and nucleic acid molecules and uses therefor
WO2000053769A1 (en) Novel thioredoxin family active site molecules and uses therefor
WO2002038767A2 (en) G protein-coupled receptor protein and nucleic acid molecules and uses therefor
US6204013B1 (en) MSP-5 nucleic acid molecules and uses therefor
EP1053351A1 (en) Novel molecules of the tnf receptor superfamily and uses therefor
US6756212B1 (en) Isolated proteins and nucleic acid molecules having homology to the NIP2 protein and uses thereof
US7632660B2 (en) Lymphoma associated molecules and uses therefor
WO2001000878A2 (en) A novel vascular endothelial growth factor family member and uses thereof
US20020004193A1 (en) Novel MP-7 protein and nucleic acid molecules and uses therefor
WO2000017236A2 (en) Apoptosis related protein and uses therefor
WO2002010390A2 (en) 57250, a human sugar transporter family member and uses thereof
WO2000026231A1 (en) Lymphoma associated molecules and uses therefor
WO2000043514A2 (en) Human acid sensing ion channel and uses thereof
US20020086982A1 (en) Novel EBI-3-ALT protein and nucleic acid molecules and uses therefor
US20030049727A1 (en) 25658, a novel human calcium channel subunit and uses thereof
WO2002076174A2 (en) Fbh58295fl, a novel human amino acid transporter and uses thereof
WO2001081416A2 (en) Human ion channel and uses thereof

Legal Events

Date Code Title Description
ENP Entry into the national phase in:

Ref country code: AU

Ref document number: 1999 65005

Kind code of ref document: A

Format of ref document f/p: F

AK Designated states

Kind code of ref document: A2

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A3

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: C1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: C1

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

CFP Corrected version of a pamphlet front page

Free format text: REVISED TITLE RECEIVED BY THE INTERNATIONAL BUREAU AFTER COMPLETION OF THE TECHNICAL PREPARATIONS FOR INTERNATIONAL PUBLICATION

122 Ep: pct application non-entry in european phase