EP1226152A2 - Regulation de l'expression genetique au moyen d'agents neuroleptiques - Google Patents

Regulation de l'expression genetique au moyen d'agents neuroleptiques

Info

Publication number
EP1226152A2
EP1226152A2 EP00975448A EP00975448A EP1226152A2 EP 1226152 A2 EP1226152 A2 EP 1226152A2 EP 00975448 A EP00975448 A EP 00975448A EP 00975448 A EP00975448 A EP 00975448A EP 1226152 A2 EP1226152 A2 EP 1226152A2
Authority
EP
European Patent Office
Prior art keywords
seq
polypeptide
polynucleotide
sequence
clz
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP00975448A
Other languages
German (de)
English (en)
Inventor
Elizabeth A. Thomas
J. Gregor Sutcliffe
Thomas M. Pribyl
Brian Hilbush
Karl W. Hasel
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Digital Gene Technologies Inc
Original Assignee
Digital Gene Technologies 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 Digital Gene Technologies Inc filed Critical Digital Gene Technologies Inc
Publication of EP1226152A2 publication Critical patent/EP1226152A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia

Definitions

  • the short-term effects of dopamine antagonists on the brain are well known and include such effects as an increase in dopamine synthesis and catabolism, an increase in the firing rate of dopamine neurons resulting from the inhibition of pre-synaptic dopamine autoreceptors (Grace et al., J. Pharm. Exp. Ther., 238, 1092-1100 (1986), and a potentiation of cyclic AMP formation resulting from the blockade of post-synaptic dopamine receptors (Rupniak et al., Psychopharm., 84, 519-521 (1984)).
  • neuroleptic druss Side effects of neuroleptic druss. In addition to their antipsychotic actions, neuroleptics can cause a series of mild to severe side effects. Some of these side-effects result from the dirty nature of neuroleptic dmgs, including hypotension and tachycardia, which results from alpha-adrenergic receptor blockade, and dry mouth and blurred vision, which results from the blockade of muscarinic acetylcholine receptors. The predominant and most undesirable effects that accompany neuroleptic treatment are the long-lasting motor deficits referred to as extrapyramidal side effects (Marsden et al., Psychol. Med., 10, 55-72 (1980)).
  • neuroleptic drags are characterized by their ability to cause late and long-lasting motor deficits.
  • the distinct temporal discrepancy which exists between dopamine receptor occupancy and the onset of therapeutic and extrapyramidal side effects, suggests that additional molecular changes in the brain occur downstream from dopamine receptor blockade.
  • studies have focused on dopamine-receptor regulation of individual target genes in the striatum and nucleus accumbens.
  • Chronic treatment with neuroleptic dmgs has also been shown to cause changes in the expression of certain neuropeptides and neurotransmitter receptors.
  • both neurotensin and enkephalin are upregulated after chronic (7 - 28 days) treatment with haloperidol, while levels of protachykinin mRNA are decreased (Merchant et al., J. Pharm. Exp. Ther., 271, 460-471 (1994); Delfs et al., J. Neurochem., 63, 777-780 (1994); Angulo et al., Neurosci. Lett. 113, 217-221 (1990)).
  • TOGA Total Gene Expression Analysis
  • the studies have also examined the pattern of expression of neuroleptic-regulated genes in various regions of the brain. Among other things, these studies are useful to determine the genes specifically associated with anti-psychotic activity versus those associated with extrapyramidal side effects, which information advances the development of improved antipsychotic therapies.
  • the identified neuroleptic-regulated molecules are useful in therapeutic and diagnostic applications in the treatment of various neuropsychiatric disorders, such as psychoses, bipolar disorder, and addiction-related behavior. Such molecules are also useful as probes as described by their size, partial nucleotide sequence and characteristic regulation pattern associated with neuroleptic administration.
  • the present invention provides novel polynucleotides and the encoded polypeptides. Moreover, the present invention relates to vectors, host cells, antibodies, and recombinant methods for producing the polynucleotides and the polypeptides.
  • One embodiment of the invention provides an isolated nucleic acid molecule comprising a polynucleotide chosen from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO.T 1, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO:16, SEQ ID NO:17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO: 49, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO
  • an isolated nucleic acid molecule comprising a polynucleotide at least 95% identical to any one of these isolated nucleic acid molecules and an isolated nucleic acid molecule at least ten bases in length that is hybridizable to any one of these isolated nucleic acid molecules under stringent conditions. Any one of these isolated nucleic acid molecules can comprise sequential nucleotide deletions from either the 5'- terminus or the 3 '-terminus. Further provided is a recombinant vector comprising any one of these isolated nucleic acid molecules and a recombinant host cell comprising any one of these isolated nucleic acid molecules. Also provided is the gene corresponding to the cDNA sequence of any one of these isolated nucleic acids.
  • Another embodiment of the invention provides an isolated polypeptide encoded by a polynucleotide chosen from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NO.T 1, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO:14, SEQ ID NO:15, SEQ ID NO.16, SEQ ID NO:17, SEQ ID NO: 18, SEQ ID NO:19, SEQ ID NO: 49, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO: 57, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:62, SEQ ID NO:63, SEQ ID NO.64, SEQ ID NO:65, SEQ ID NO:66,
  • an isolated nucleic acid molecule encoding any of these polypeptides, an isolated nucleic acid molecule encoding a fragment of any of these polypeptides, an isolated nucleic acid molecule encoding a polypeptide epitope of any of these polypeptides, and an isolated nucleic acid encoding a species homologue of any of these polypeptides.
  • Another embodiment of the invention provides an isolated polypeptide of SEQ ID NO: 109.
  • Another embodiment of the invention provides an isolated polypeptide of SEQ ID NO: 110.
  • any one of these polypeptides has biological activity.
  • any one of the isolated polypeptides comprises sequential amino acid deletions from either the C-terminus or the N-terminus.
  • a recombinant host cell that expresses any one of these isolated polypeptides.
  • Yet another embodiment of the invention comprises an isolated antibody that binds specifically to an isolated polypeptide encoded by a polynucleotide chosen from the group consisting of SEQ ID NO:l, SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7, SEQ ID NO:8, SEQ ID NO:9, SEQ ID NO: 10, SEQ ID NOT 1, SEQ ID NO:12, SEQ ID NO:13, SEQ ID NO: 14, SEQ ID NO:15, SEQ ID NO: 16, SEQ ID NO: 17, SEQ ID NO:18, SEQ ID NO:19, SEQ ID NO: 49, SEQ ID NO:50, SEQ ID NO:51, SEQ ID NO:52, SEQ ID NO: 57, SEQ ID NO:58, SEQ ID NO:59, SEQ ID NO:60, SEQ ID NO:61, SEQ ID NO:62, SEQ ID NO:63, SEQ ID NO:64, SEQ ID NO:65, S
  • Yet another embodiment of the invention comprises an isolated antibody that binds specifically to an isolated polypeptide of SEQ ID NO: 109. Yet another embodiment of the invention comprises an isolated antibody that binds specifically to an isolated polypeptide of SEQ ID NO: 110.
  • the isolated antibody can be a monoclonal antibody or a polyclonal antibody.
  • Another embodiment of the invention provides a method for preventing, treating, modulating, or ameliorating a medical condition, such as a neuropsychiatric disorder, comprising administering to a mammalian subject a therapeutically effective amount of a polypeptide of the invention or a polynucleotide of the invention.
  • a method for preventing, treating, modulating or ameliorating schizophrenia is provided.
  • a method for preventing, treating, modulating or ameliorating bipolar disorder is provided.
  • a method for preventing, treating, modulating or ameliorating addiction-related behavior is provided.
  • a further embodiment of the invention provides an isolated antibody that binds specifically to the isolated polypeptide of the invention.
  • a preferred embodiment of the invention provides a method for preventing, treating, modulating, or ameliorating a medical condition, such as a neuropsychiatric disorder, comprising administering to a mammalian subject a therapeutically effective amount of the antibody.
  • a method for preventing, treating, modulating or ameliorating schizophrenia is provided.
  • a method for preventing, treating, modulating or ameliorating bipolar disorders is provided.
  • a method for preventing, treating, modulating or ameliorating addiction-related behavior is provided.
  • An additional embodiment of the invention provides a method of diagnosing a pathological condition or a susceptibility to a pathological condition in a subject.
  • the method comprises determining the presence or absence of a mutation in a polynucleotide of the invention.
  • a pathological condition or a susceptibility to a pathological condition such as a neuropsychiatric disorder, is diagnosed based on the presence or absence of the mutation.
  • a method for diagnosing schizophrenia is provided.
  • a method for diagnosing bipolar disorders is provided.
  • a method for preventing, treating, modulating or ameliorating addiction-related behavior is provided.
  • Yet another embodiment of the invention provides a method of diagnosing a pathological condition or a susceptibility to a pathological condition, such as a neuropsychiatric disorder, in a subject.
  • a pathological condition such as a neuropsychiatric disorder
  • Especially preferred embodiments include methods of diagnosing schizophrenia and bipolar disorders.
  • the method comprises detecting an alteration in expression of a polypeptide encoded by the polynucleotide of the invention, wherein the presence of an alteration in expression of the polypeptide is indicative of the pathological condition or susceptibility to the pathological condition.
  • the alteration in expression can be an increase in the amount of expression or a decrease in the amount of expression.
  • Yet another embodiment of the invention is a method of identifying an activity of an expressed polypeptide in a biological assay.
  • a polypeptide of the invention is expressed in a cell and isolated.
  • the expressed polypeptide is tested for an activity in a biological assay and the activity of the expressed polypeptide is identified based on the test results.
  • DNA molecule suitable for use as a probe for genes regulated in neuropsychiatric disorders chosen from the group consisting of the DNA molecules shown in
  • Figure 1 is a graphical representation of the results of TOGA analysis using a 5' PCR primer with parsing bases AGT A, showing PCR products produced from mRNA extracted from the striatum/nucleus accumbens of mice treated with 7.5 mg/kg of clozapine for the following durations: control (no clozapine), 45 minutes, 7 hours, 5 days, 12 days, and 14 days, where the vertical index line indicates a PCR product of about 106 b.p. that is present in the control sample and enriched in the clozapine-treated samples;
  • Figure 3 is a graphical representation of the results of TOGA analysis using a 5' PCR primer with parsing bases CACC, showing PCR products produced from mRNA extracted from the striatum/nucleus accumbens of mice treated with 7.5 mg/kg of clozapine for the following durations: control (no clozapine), 45 minutes, 7 hours, 5 days, 12 days, and 14 days, where the vertical index line indicates a PCR product of about 201 b.p. that is present in the control sample and increasingly enriched over time in the clozapine-treated samples;
  • Figure 4 shows a Northern Blot analysis of clone CLZ_5 (CACC 201), where an agarose gel containing poly A enriched mRNA from the striatum/nucleus accumbens of mice treated with clozapine as well as size standards was blotted after electrophoresis and probed with radiolabelled CLZ_5.
  • Mice were treated with clozapine (7.5 mg/kg) for the following time durations before mRNA extraction: control (no clozapine), 45 minutes, 7 hours, 5 days, 12 days, and 14 days;
  • Figure 6 is a graphical representation comparing the results of the TOGA analysis of clone CLZ_5 shown in Fig. 3 and the Northern Blot analysis of clone CLZ 5 shown in Figure 4;
  • Figure 11 A-H shows GFAP and apoD co-localization in the striatum (11 A, B, D, E) and optic tract (1 IC, F) of control saline (11 A, B, C) and clozapine-treated animals (1 ID, E, F), with thick arrows designating the co-localization of GFAP and apoD mRNA and thin arrows designating the expression of apoD only;
  • 11G-H shows apoD immunohistochemistry with an anti-human apoD primary antibody (Novocastra, Newcastle, UK) in the optic tract of control saline (11G) and clozapine-treated animals (11H).
  • Figure 13 is a graphical representation of the results of TOGA analysis using a 5' PCR primer with parsing bases TTGT, showing PCR products produced from mRNA extracted from the striatum/nucleus accumbens of mice treated with 7.5 mg/kg clozapine as follows: control (no clozapine), 45 minutes, 7 hours, 5 days, 12 days, and 14 days, where the vertical index line indicates a PCR product of about 266 b.p. that is present in the control sample, is down-regulated within 45 minutes in the clozapine-treated sample, and remains down-regulated for 14 days in the presence of clozapine;
  • Figure 20 shows the sequence of the EST AF006196: Mus musculus metalloprotease-disintegrin MDC15 mRNA, complete eds.
  • Figure 27A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_26, showing the pattern of CLZ 26 mRNA expression in a coronal section of the hemispheres at the level of hippocampal formation (27 A) and coronal section of the hemispheres at the level of striatum (27B) in mouse brain;
  • Figure 28A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_28, showing the pattern of CLZ_28 mRNA expression in a coronal section through the hemispheres at the level of hippocampus (28 A) and coronal section through the posterior region of hemispheres (28B) in mouse brain;
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formulation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer- RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • DNA sequences generated by sequencing reactions can contain sequencing errors.
  • the errors exist as misidentified nucleotides, or as insertions or deletions of nucleotides in the generated DNA sequence.
  • the erroneously inserted or deleted nucleotides cause frame shifts in the reading frames of the predicted amino acid sequence.
  • the predicted amino acid sequence diverges from the actual amino acid sequence, even though the generated DNA sequence may be greater than 99.9% identical to the actual DNA sequence (for example, one base insertion or deletion in an open reading frame of over 1000 bases).
  • polypeptides and the polynucleotides encoding such polypeptides, are contemplated by the present invention.
  • polynucleotides having at least 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity will encode a polypeptide identical to an amino acid sequence contained in the translations of SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • the invention further includes polypeptide variants which show substantial biological activity.
  • variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity.
  • guidance concerning how to make phenotypically silent amino acid substitutions is provided in Bowie, et al., Science, 247:1306-1310 (1990), wherein the authors indicate that there are two main strategies for studying the tolerance of an amino acid sequence to change.
  • variants of the present invention include (i) substitutions with one or more of the non-conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) substitution with one or more of amino acid residues having a substituent group, or (iii) fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), or (iv) fusion of the polypeptide with additional amino acids, such as an IgG Fc fusion region peptide, or leader or secretory sequence, or a sequence facilitating purification.
  • substitutions with one or more of the non-conserved amino acid residues where the substituted amino acid residues may or may not be one encoded by the genetic code
  • substitution with one or more of amino acid residues having a substituent group or fusion of the mature polypeptide with another compound, such as a compound to increase the stability and/or solubility
  • polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as decreased aggregation.
  • aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity (see, e.g., Pinckard et al., Clin. Exp. Immunol., 2:331- 340 (1967); Robbins et al., Diabetes, 36: 838-845 (1987); Cleland et al, Crit. Rev. Therapeutic Drug Carrier Systems, 10:307-377 (1993)).
  • a "polynucleotide fragment” refers to a short polynucleotide having a nucleic acid sequence contained in that shown in SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • the short nucleotide fragments are preferably at least about 15 nt, and more preferably at least about 20 nt, still more preferably at least about 30 nt, and even more preferably, at least about 40 nt in length.
  • a fragment "at least 20 nt in length,” for example, is intended to include 20 or more contiguous bases from the cDNA sequence contained in that shown in SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • These nucleotide fragments are useful as diagnostic probes and primers as discussed herein. Of course, larger fragments (e.g., 50, 150, and more nucleotides) are preferred.
  • Preferred polypeptide fragments include the secreted protein as well as the mature form. Further preferred polypeptide fragments include the secreted protein or the mature form having a continuous series of deleted residues from the amino or the carboxy terminus, or both. For example, any number of amino acids, ranging from 1-60, can be deleted from the amino terminus of either the secreted polypeptide or the mature form. Similarly, any number of amino acids, ranging from 1-30, can be deleted from the carboxy terminus of the secreted protein or mature form. Furthermore, any combination of the above amino and carboxy terminus deletions are preferred. Similarly, polynucleotide fragments encoding these polypeptide fragments are also preferred.
  • polypeptide and polynucleotide fragments characterized by structural or functional domains, such as fragments that comprise alpha-helix and alpha- helix forming regions, beta-sheet and beta-sheet-forming regions, turn and tum-forming regions, coil and coil-forming regions, hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions, substrate binding region, and high antigenic index regions.
  • Polypeptide fragments of the translations of SEQ ID NOs: 1-19; 49-52; 57-72 and 107 falling within conserved domains are specifically contemplated by the present invention.
  • polynucleotide fragments encoding these domains are also contemplated.
  • Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention.
  • the biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
  • epitopes refer to polypeptide fragments having antigenic or immunogenic activity in an animal, especially in a human.
  • a preferred embodiment of the present invention relates to a polypeptide fragment comprising an epitope, as well as the polynucleotide encoding this fragment.
  • a region of a protein molecule to which an antibody can bind is defined as an "antigenic epitope.”
  • an "immunogenic epitope” is defined as a part of a protein that elicits an antibody response (see, e.g., Geysen et al., Proc. Natl. Acad. Sci. USA, 81:3998-4002 (1983)).
  • antibody As used herein, the term "antibody” (Ab) or “monoclonal antibody” (Mab) is meant to include intact molecules as well as antibody fragments (such as, for example, Fab and F(ab')2 fragments) which are capable of specifically binding to protein. Fab and F(ab')2 fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody (Wahl et al., J. Nucl Med., 24:316-325 (1983)). Thus, these fragments are preferred, as well as the products of a FAB or other immunoglobulin expression library. Moreover, antibodies of the present invention include chimeric, single chain, and humanized antibodies.
  • chimeric antibodies e.g., humanized versions of murine monoclonal antibodies.
  • Such humanized antibodies may be prepared by known techniques, and offer the advantage of reduced immunogenicity when the antibodies are administered to humans.
  • a humanized monoclonal antibody comprises the variable region of a murine antibody (or just the antigen binding site thereof) and a constant region derived from a human antibody.
  • a humanized antibody fragment may comprise the antigen binding site of a murine monoclonal antibody and a variable region fragment (lacking the antigen-binding site) derived from a human antibody.
  • Procedures for the production of chimeric and further engineered monoclonal antibodies include those described in Riechmann et al.
  • One method for producing a human antibody comprises immunizing a non-human animal, such as a transgenic mouse, with a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49-52; 57-72 and 107, whereby antibodies directed against the polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49-52; 57-72 and 107 are generated in said animal.
  • Non-human animals such as transgenic mice
  • transgenic mice into which genetic material encoding one or more human immunoglobulin chains has been introduced may be employed.
  • Such transgenic mice may be genetically altered in a variety of ways. The genetic manipulation may result in human immunoglobulin polypeptide chains replacing endogenous immunoglobulin chains in at least some (preferably virtually all) antibodies produced by the animal upon immunization.
  • Antibodies produced by immunizing transgenic animals with a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49- 52; 57-72 and 107 are provided herein.
  • mice in which one or more endogenous immunoglobulin genes are inactivated by various means have been prepared.
  • Human immunoglobulin genes have been introduced into the mice to replace the inactivated mouse genes.
  • Antibodies produced in the animals incorporate human immunoglobulin polypeptide chains encoded by the human genetic material introduced into the animal. Examples of techniques for production and use of such transgenic animals are described in U.S. Patent Nos. 5,814,318; 5,569,825; and 5,545,806, which are incorporated by reference herein.
  • Monoclonal antibodies may be produced by conventional procedures, e.g., by immortalizing spleen cells harvested from the transgenic animal after completion of the immunization schedule.
  • the spleen cells may be fused with myeloma cells to produce hybridomas by conventional procedures.
  • a method for producing a hybridoma cell line comprises immunizing such a transgenic animal with an immunogen comprising at least seven contiguous amino acid residues of a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49-52; 57-72 and 107; harvesting spleen cells from the immunized animal; fusing the harvested spleen cells to a myeloma cell line, thereby generating hybridoma cells; and identifying a hybridoma cell line that produces a monoclonal antibody that binds a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1- 19; 49-52; 57-72 and 107.
  • Such hybridoma cell lines, and mosclonal antibodies produced therefrom, are encompassed by the present invention.
  • Monoclonal antibodies secreted by the hybridoma cell line are purified by conventional techniques.
  • Antibodies may be employed in an in vitro procedure, or administered in vivo to inhibit biological activity induced by a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49-52; 57-72 and 107. Disorders caused or exacerbated (directly or indirectly) by the interaction of such polypeptides of the present invention with cell surface receptors thus may be treated.
  • a therapeutic method involves in vivo administration of a blocking antibody to a mammal in an amount effective for reducing a biological activity induced by a polypeptide translated from a nucleotide sequence chosen from SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • chronic administration of neuroleptics can cause unwanted side effects.
  • Administration of an antibody derived from the identified polynucleotides might block the signaling that causes these side effects.
  • an antibody derived from the identified polynucleotides might selectively block proteins causing motor side effects.
  • domains that can be fused to polypeptides of the present invention include not only heterologous signal sequences, but also other heterologous functional regions.
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • fusion proteins may also be engineered to improve characteristics of the polypeptide of the present invention. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence during purification from the host cell or subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to facilitate handling of polypeptides are familiar and routine techniques in the art.
  • polypeptides of the present invention can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric polypeptides.
  • IgG immunoglobulins
  • fusion proteins facilitate purification and show an increased half-life in vivo.
  • chimeric proteins consisting of the first two domains of the human CD4- polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (see, EP A 394,827; Traunecker et al., Nature, 331 :84-86 (1988)).
  • Fusion proteins having disulfide-linked dimeric structures can also be more efficient in binding and neutralizing other molecules, than the monomeric secreted protein or protein fragment alone (Fountoulakis et al., J. Biochem. 270:3958- 3964 (1995)).
  • EP-A-0 464 533 (Canadian counterpart 2045869) discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • the Fc part in a fusion protein is beneficial in therapy and diagnosis, and thus can result in, for example, improved pharmacokinetic properties (see, e.g., EP-A 0 232 262.)
  • deleting the Fc part after the fusion protein has been expressed, detected, and purified, would be desired.
  • the Fc portion may hinder therapy and diagnosis if the fusion protein is used as an antigen for immunizations.
  • hIL-5 human proteins, such as hIL-5, have been fused with Fc portions for the purpose of high- throughput screening assays to identify antagonists of hIL-5 (see, D. Bennett et al., J Molecular Recognition, 8:52-58 (1995); K. Johanson et al., J. Biol Chem., 270:9459- 9471 (1995)).
  • the polypeptides of the present invention can be fused to marker sequences, such as a peptide which facilitates purification of the fused polypeptide.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein (Proc. Natl. Acad. Sci. USA, 86:821-824 (1989)).
  • HA hemagglutinin protein
  • Other fusion proteins may use the ability of the polypeptides of the present invention to target the delivery of a biologically active peptide. This might include focused delivery of a toxin to tumor cells, or a growth factor to stem cells.
  • any of these above fusions can be engineered using the polynucleotides or the polypeptides of the present invention.
  • Vectors, Host Cells, and Protein Production are engineered using the polynucleotides or the polypeptides of the present invention.
  • the present invention also relates to vectors containing the polynucleotide of the present invention, host cells, and the production of polypeptides by recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral, or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, tip, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, 293, Bowes melanoma cells and plant cells.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, PNHl 6a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc.
  • eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • constmct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology, (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • polypeptide of this invention can be recovered and purified from recombinant cell cultures by well-known methods including ammomum sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention can also be recovered from: products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells.
  • a prokaryotic or eukaryotic host including, for example, bacterial, yeast, higher plant, insect, and mammalian cells.
  • the polypeptides of the present invention may be glycosylated or may be non- glycosylated.
  • polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
  • the polynucleotides of the present invention are useful for chromosome identification. There exists an ongoing need to identify new chromosome markers, since few chromosome marking reagents, based on actual sequence data (repeat polymorphisms), are presently available. Each polynucleotide of the present invention can be used as a chromosome marker.
  • sequences can be mapped to chromosomes by preparing PCR primers
  • Primers can be selected using computer analysis so that primers do not span more than one predicted exon in the genomic DNA. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the SEQ ID NOs: 1-19; 49-52; 57- 72 and 107 will yield an amplified fragment.
  • somatic hybrids provide a rapid method of PCR mapping the polynucleotides to particular chromosomes. Three or more clones can be assigned per day using a single thermal cycler. Moreover, sublocalization of the polynucleotides can be achieved with panels of specific chromosome fragments.
  • Other gene mapping strategies that can be used include in situ hybridization, prescreening with labeled flow- sorted chromosomes, and preselection by hybridization to constmct chromosome specific-cDNA libraries.
  • FISH fluorescence in situ hybridization
  • the polynucleotides can be used individually (to mark a single chromosome or a single site on that chromosome) or in panels (for marking multiple sites and/or multiple chromosomes).
  • Preferred polynucleotides correspond to the noncoding regions of the cDNAs because the coding sequences are more likely conserved within gene families, thus increasing the chance of cross hybridization during chromosomal mapping.
  • Linkage analysis establishes coinheritance between a chromosomal location and presentation of a particular disease .
  • Disease mapping data are found, for example in V. McKusick, Mendelian Inheritance in Man (available on line through Johns Hopkins University Welch Medical Library) Assuming one megabase mapping resolution and one gene per 20 kb, a cDNA precisely localized to a chromosomal region associated with the disease could be one of 50-500 potential causative genes.
  • polynucleotide and the corresponding gene between affected and unaffected individuals can be examined.
  • the polynucleotides of SEQ ID NOs: 1-19; 49-52; 57-72 and 107 can be used for this analysis of individual humans.
  • a polynucleotide can be used to control gene expression through triple helix formation or antisense DNA or RNA. Both methods rely on binding of the polynucleotide to DNA or RNA. For these techniques, preferred polynucleotides are usually 20 to 40 bases in length and complementary to either the region of the gene involved in transcription (see, Lee et al., Nucl. Acids Res., 6:3073 (1979); Cooney et al., Science, 241:456 (1988); and Dervan et al., Science, 251:1360 (1991) for discussion of triple helix formation) or to the mRNA itself (see, Okano, J.
  • Polynucleotides of the present invention are also useful in gene therapy.
  • One goal of gene therapy is to insert a normal gene into an organism having a defective gene, in an effort to correct the genetic defect.
  • the polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner.
  • Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell.
  • the polynucleotides are also useful for identifying individuals from minute biological samples. The United States military, for example, is considering the use of restriction fragment length polymorphism (RFLP) for identification of its personnel.
  • RFLP restriction fragment length polymorphism
  • an individual's genomic DNA is digested with one or more restriction enzymes, and probed on a Southern blot to yield unique bands for identifying personnel.
  • This method does not suffer from the current limitations of "Dog Tags" which can be lost, switched, or stolen, making positive identification difficult.
  • the polynucleotides of the present invention can be used as additional DNA markers for RFLP.
  • polynucleotides of the present invention can also be used as an alternative to
  • RFLP RFLP
  • DNA sequences taken from very small biological samples such as tissues, e.g., hair or skin, or body fluids, e.g., blood, saliva, semen, etc.
  • DNA sequences amplified from polymorphic loci such as DQa class II HLA gene
  • forensic biology to identify individuals (Erlich, H., PCR Technology, Freeman and Co. (1992)).
  • polymorphic loci such as DQa class II HLA gene
  • polymorphic loci such as DQa class II HLA gene
  • reagents capable of identifying the source of a particular tissue. Such need arises, for example, in forensics when presented with tissue of unknown origin.
  • Appropriate reagents can comprise, for example, DNA probes or primers specific to particular tissue prepared from the sequences of the present invention. Panels of such reagents can identify tissue by species and/or by organ type. In a similar fashion, these reagents can be used to screen tissue cultures for contamination.
  • the polynucleotides of the present invention can be used as molecular weight markers on Southern gels, as diagnostic probes for the presence of a specific mRNA in a particular cell type, as a probe to "subtract-out" known sequences in the process of discovering novel polynucleotides, for selecting and making oligomers for attachment to a "gene chip” or other support, to raise anti-DNA antibodies using DNA immunization techniques, and as an antigen to elicit an immune response.
  • polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
  • a polypeptide of the present invention can be used to assay protein levels in a biological sample using antibody-based techniques.
  • protein expression in tissues can be studied with classical immunohistological methods (Jalkanen, M., et al., J. Cell Biol, 101 :976-985 (1985); jalkanen, M., et al., J. Cell . Biol, 105:3087-3096 (1987)).
  • Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine ( I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( U2 In), and technetium ( 99m Tc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase, and radioisotopes, such as iodine ( I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( U2 In), and technetium ( 99m Tc)
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • proteins can also be detected in vivo by imaging.
  • Antibody labels or markers for in vivo imaging of protein include those detectable by X-radiography, NMR or ESR.
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject.
  • suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by labeling of nutrients for the relevant hybridoma.
  • a protein-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, , 12 In, 99m Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously, or intraperitoneally) into the mammal.
  • a radioisotope for example, 131 I, , 12 In, 99m Tc
  • a radio-opaque substance for example, parenterally, subcutaneously, or intraperitoneally
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99m Tc.
  • the labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain the specific protein.
  • In vivo tumor imaging is described in S.W. Burchiel et al., "Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments" (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S.W. Burchiel and B. A. Rhodes, Eds., Masson Publishing Inc. (1982)).
  • the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression of a polypeptide of the present invention in cells or body fluid of an individual; (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed polypeptide gene expression level compared to the standard expression level is indicative of a disorder.
  • Psychiatric disorders and treatment of psychiatric disorders with neuroleptics, including schizophrenia are associated with a dysregulation of neurotransmitter and/or neuropeptide levels that can result in the up- or down regulation of polynucleotides and polypeptides. These changes can be diagnosed or monitored by assaying changes in polypeptide levels in tissue or fluids such as CSF, blook, or in fecal samples.
  • polypeptides of the present invention can be used to treat disease.
  • patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B), to inhibit the activity of a polypeptide (e.g., an oncogene), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth).
  • a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B), to inhibit the activity of a polypeptide (
  • antibodies directed to a polypeptide of the present invention can also be used to treat disease.
  • administration of an antibody directed to a polypeptide of the present invention can bind and reduce overproduction of the polypeptide.
  • administration of an antibody can activate the polypeptide, such as by binding to a polypeptide bound to a membrane (receptor).
  • Polypeptides can also be used as antigens to trigger immune responses.
  • neurotransmitters and neuropeptides modulates many aspects of neuronal function. For example, in schizophrenia overactive neurotransmitter activity is thought to be basis for the psychotic behavior. Administration of an antibody to an overproduced polypeptide can be used to modulate neuronal responses in psychiatric disorders such as schizophrenia.
  • polypeptides of the present invention can be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art. Polypeptides can also be used to raise antibodies, which in turn are used to measure protein expression from a recombinant cell, as a way of assessing transformation of the host cell. Moreover, the polypeptides of the present invention can be used to test the following biological activities.
  • polynucleotides and polypeptides of the present invention can be used in assays to test for one or more biological activities. If these polynucleotides and polypeptides do exhibit activity in a particular assay, it is likely that these molecules may be involved in the diseases associated with the biological activity. Thus, the polynucleotides and polypeptides could be used to treat the associated disease.
  • a polypeptide or polynucleotide of the present invention may be useful in treating deficiencies or disorders of the central nervous system or peripheral nervous system, by activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of neuroblasts, stem cells or glial cells.
  • a polypeptide or polynucleotide of the present invention may be useful in treating deficiencies or disorders of the central nervous system or peripheral nervous system, by activating or inhibiting the mechanisms of synaptic transmission, synthesis, metabolism and inactivation of neural transmitters, neuromodulators and trophic factors, expression and incorporation of enzymes, structural proteins, membrane channels and receptors in neurons and glial cells, or altering neural membrane compositions.
  • a polynucleotide or polypeptide of the present invention can be used as a marker or detector of a particular nervous system disease or disorder.
  • the disorder or disease can be any of Alzheimer's disease, Pick's disease, Binswanger's disease, other senile dementia, Parkinson's disease, parkinsonism, obsessive compulsive disorders, epilepsy, encephalopathy, ischemia, alcohol addiction, drug addiction, schizophrenia, amyotrophic lateral sclerosis, multiple sclerosis, depression, and bipolar manic-depressive disorder.
  • the polypeptide or polynucleotide of the present invention can be used to study circadian variation, aging, or long-term potentiation, the latter affecting the hippocampus. Additionally, particularly with reference to mRNA species occurring in particular structures within the central nervous system, the polypeptide or polynucleotide of the present invention can be used to study brain regions that are known to be involved in complex behaviors, such as learning and memory, emotion, drug addiction, glutamate neurotoxicity, feeding behavior, olfaction, viral infection, vision, and movement disorders.
  • a polypeptide or polynucleotide of the present invention may be useful in treating deficiencies or disorders of the immune system, by activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells.
  • Immune cells develop through a process called hematopoiesis, producing myeloid (platelets, red blood cells, neutrophils, and macrophages) and lymphoid (B and T lymphocytes) cells from pluripotent stem cells.
  • the etiology of these immune deficiencies or disorders may be genetic, somatic, such as cancer or some autoimmune disorders, acquired (e.g., by chemotherapy or toxins), or infectious.
  • a polynucleotide or polypeptide of the present invention can be used as a marker or detector of a particular immune system disease or disorder.
  • a polynucleotide or polypeptide of the present invention may be useful in treating or detecting deficiencies or disorders of hematopoietic cells.
  • a polypeptide or polynucleotide of the present invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat those disorders associated with a decrease in certain (or many) types hematopoietic cells.
  • immunologic deficiency syndromes include, but are not limited to: blood protein disorders (e.g.
  • agammaglobulinemia dysgammaglobulinemia
  • ataxia telangiectasia common variable immunodeficiency
  • Di George's Syndrome HIV infection
  • HTLV-BLV infection leukocyte adhesion deficiency syndrome
  • lymphopenia phagocyte bactericidal dysfunction
  • severe combined immunodeficiency SCIDs
  • Wiskott-Aldrich Disorder anemia, thrombocytopenia, or hemoglobinuria.
  • a polypeptide or polynucleotide of the present invention could also be used to modulate hemostatic (the stopping of bleeding) or thrombolytic activity (clot formation).
  • a polynucleotide or polypeptide of the present invention could be used to treat blood coagulation disorders (e.g., afibrinogenemia, factor deficiencies), blood platelet disorders (e.g. thrombocytopenia), or wounds resulting from trauma, surgery, or other causes.
  • a polynucleotide or polypeptide of the present invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting.
  • a polynucleotide or polypeptide of the present invention may also be useful in treating or detecting autoimmune disorders.
  • Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue. Therefore, the administration of a polypeptide or polynucleotide of the present invention that inhibits an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells or in some ways results in the induction of tolerance, may be an effective therapy in preventing autoimmune disorders.
  • autoimmune disorders examples include, but are not limited to: Addison's Disease, hemolytic anemia, antiphospholipid syndrome, rheumatoid arthritis, dermatitis, allergic encephalomyelitis, glomerulonephritis, Goodpasture's Syndrome, Graves' Disease, Multiple Sclerosis, Myasthenia Gravis, Neuritis, Ophthalmia, Bullous Pemphigoid, Pemphigus, Polyendocrinopathies, Purpura, Reiter's Disease, Stiff-Man Syndrome, Autoimmune Thyroiditis, Systemic Lupus Erythematosus, Autoimmune Pulmonary Inflammation, Guillain-Barre Syndrome, insulin dependent diabetes mellitis, and autoimmune inflammatory eye disease.
  • Schizophrenia has several aspects that suggest an autoimmune component to the disease process.
  • Patients with schizophrenia exhibit immunological abnormalities including hypersecretion of cytokines, presence of antinuclear, anticytoplasmic and antiphospholipid antibodies and a decreased ratio of CD4+/CD8+ cells.
  • allergic reactions and conditions such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated by a polypeptide or polynucleotide of the present invention.
  • these molecules can be used to treat anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
  • a polynucleotide or polypeptide of the present invention may also be used to treat and/or prevent organ rejection or graft-versus-host disease (GVHD).
  • Organ rejection occurs by host immune cell destruction of the transplanted tissue through an immune response.
  • an immune response is also involved in GVHD, but, in this case, the foreign transplanted immune cells destroy the host tissues.
  • the administration of a polypeptide or polynucleotide of the present invention that inhibits an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD.
  • a polypeptide or polynucleotide of the present invention may also be used to modulate inflammation.
  • the polypeptide or polynucleotide may inhibit the proliferation and differentiation of cells involved in an inflammatory response.
  • These molecules can be used to treat inflammatory conditions, both chronic and acute conditions, including inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endo toxin lethality, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, or resulting from over production of cytokines (e.g., TNF or IL-1).
  • infection e.g., septic shock, sepsis, or systemic inflammatory response syndrome (SIRS)
  • ischemia-reperfusion injury e.g., endo toxin lethality, arthritis, complement-
  • a polypeptide or polynucleotide can be used to treat or detect hyperproliferative disorders, including neoplasms.
  • a polypeptide or polynucleotide of the present invention may inhibit the proliferation of the disorder through direct or indirect interactions.
  • a polypeptide or polynucleotide of the present invention may proliferate other cells which can inhibit the hyperproliferative disorder.
  • hyperproliferative disorders can be treated.
  • This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
  • decreasing an immune response may also be a method of treating hyperproliferative disorders, such as a chemotherapeutic agent.
  • hyperproliferative disorders that can be treated or detected by a polynucleotide or polypeptide of the present invention include, but are not limited to neoplasms located in the: abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic region, skin, soft tissue, spleen, thoracic region, and urogenital system.
  • hyperproliferative disorders can also be treated or detected by a polynucleotide or polypeptide of the present invention.
  • hyperproliferative disorders include, but are not limited to: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's Macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
  • a polypeptide or polynucleotide of the present invention can be used to treat or detect infectious agents. For example, by increasing the immune response, particularly increasing the proliferation and differentiation of B and/or T cells, infectious diseases may be treated.
  • the immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
  • the polypeptide or polynucleotide of the present invention may also directly inhibit the infectious agent, without necessarily eliciting an immune response.
  • treatment of patients with a polypeptide or polynucleotide of the present invention might act as a vaccine to trigger a more efficient immune response, altering the course of disease.
  • Virases are one example of an infectious agent that can cause disease or symptoms that can be treated or detected by a polynucleotide or polypeptide of the present invention.
  • viruses include, but are not limited to the following DNA and RNA viral families: Arboviras, Adenoviridae, Arenaviridae, Arteriviras, Birnaviridae, Bunyaviridae, Caliciviridae, Circoviridae, Coronaviridae, Flaviviridae, Hepadnaviridae (Hepatitis), Herpesviridae (such as, Cytomegalovirus, Herpes Simplex, Herpes Zoster), Mononegaviras (e.g., Paramyxoviridae, Morbilliviras, Rhabdoviridae), Orthomyxoviridae (e.g., Influenza), Papovaviridae, Parvoviridae, Picornaviridae,
  • Poxviridae such as Smallpox or Vaccinia
  • Reoviridae e.g., Rotaviras
  • Retroviridae HTLV-I, HTLV-II, Lentiviras
  • Togaviridae e.g., Rubivirus
  • Virases falling within these families can cause a variety of diseases or symptoms, including, but not limited to: arthritis, bronchiollitis, encephalitis, eye infections (e.g., conjunctivitis, keratitis), chronic fatigue syndrome, hepatitis (A, B, C, E, Chronic Active, Delta), meningitis, opportunistic infections (e.g., AIDS), pneumonia, Burkitt's Lymphoma, chickenpox, hemorrhagic fever, Measles, Mumps, Parainfluenza, Rabies, the common cold, Polio, leukemia, Rubella, sexually transmitted diseases, skin diseases (e.g.,
  • a polypeptide or polynucleotide of the present invention can be used to treat or detect any of these symptoms or diseases.
  • bacterial or fungal families can cause the following diseases or symptoms, including, but not limited to: bacteremia, endocarditis, eye infections (conjunctivitis, tuberculosis, uveitis), gingivitis, opportunistic infections (e.g., AIDS related infections), paronychia, prosthesis-related infections, Reiter's Disease, respiratory tract infections, such as Whooping Cough or Empyema, sepsis, Lyme Disease, Cat-Scratch Disease, Dysentery, Paratyphoid Fever, food poisoning, Typhoid, pneumonia, Gonorrhea, meningitis, Chlamydia, Syphilis,
  • a polypeptide or polynucleotide of the present invention can be used to treat or detect any of these symptoms or diseases.
  • parasitic agents causing disease or symptoms that can be treated or detected by a polynucleotide or polypeptide of the present invention include, but not limited to, the following families: Amebiasis, Babesiosis, Coccidiosis, Cryptosporidiosis, Dientamoebiasis, Dourine, Ectoparasitic, Giardiasis, Helminthiasis, Leishmaniasis, Theileriasis, Toxoplasmosis, Trypanosomiasis, and Trichomonas.
  • These parasites can cause a variety of diseases or symptoms, including, but not limited to: Scabies, Trombiculiasis, eye infections, intestinal disease (e.g., dysentery, giardiasis), liver disease, lung disease, opportunistic infections (e.g., AIDS related), Malaria, pregnancy complications, and toxoplasmosis.
  • a polypeptide or polynucleotide of the present invention can be used to treat or detect any of these symptoms or diseases.
  • treatment using a polypeptide or polynucleotide of the present invention could either be by administering an effective amount of a polypeptide to the patient, or by removing cells from the patient, supplying the cells with a polynucleotide of the present invention, and returning the engineered cells to the patient (ex vivo therapy).
  • the polypeptide or polynucleotide of the present invention can be used as an antigen in a vaccine to raise an immune response against infectious disease.
  • a polynucleotide or polypeptide of the present invention can be used to differentiate, proliferate, and attract cells, leading to the regeneration of tissues (see, Science, 276:59-87 (1997)).
  • the regeneration of tissues could be used to repair, replace, or protect tissue damaged by congenital defects, trauma (wounds, bums, incisions, or ulcers), age, disease (e.g. osteoporosis, osteocarthritis, periodontal disease, liver failure), surgery, including cosmetic plastic surgery, fibrosis, reperfusion injury, or systemic cytokine damage.
  • Tissues that could be regenerated using the present invention include organs (e.g., pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth, skeletal or cardiac), vascular (including vascular endothelium), nervous, hematopoietic, and skeletal (bone, cartilage, tendon, and ligament) tissue.
  • organs e.g., pancreas, liver, intestine, kidney, skin, endothelium
  • muscle smooth, skeletal or cardiac
  • vascular including vascular endothelium
  • nervous hematopoietic
  • hematopoietic skeletal tissue
  • skeletal bone, cartilage, tendon, and ligament
  • a polynucleotide or polypeptide of the present invention may increase regeneration of tissues difficult to heal. For example, increased tendon/ligament regeneration would quicken recovery time after damage.
  • a polynucleotide or polypeptide of the present invention could also be used prophylactically in an effort to avoid damage. Specific diseases that could be treated include of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects.
  • tissue regeneration of non-healing wounds includes pressure ulcers, ulcers associated with vascular insufficiency, surgical, and traumatic wounds.
  • nerve and brain tissue could also be regenerated by using a polynucleotide or polypeptide of the present invention to proliferate and differentiate nerve cells.
  • Diseases that could be treated using this method include central and peripheral nervous system diseases, neuropathies, or mechanical and traumatic disorders (e.g., spinal cord disorders, head trauma, cerebrovascular disease, and stroke). Specifically, diseases associated with peripheral nerve injuries, peripheral neuropathy (e.g., resulting from chemotherapy or other medical therapies), localized neuropathies, and central nervous system diseases (e.g., Alzheimer's disease,
  • Parkinson's disease Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome
  • Shy-Drager syndrome could all be treated using the polynucleotide or polypeptide of the present invention.
  • a polynucleotide or polypeptide of the present invention may have chemotaxis activity.
  • a chemotaxic molecule attracts or mobilizes cells (e.g., monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells) to a particular site in the body, such as inflammation, infection, or site of hyperproliferation.
  • the mobilized cells can then fight off and/or heal the particular trauma or abnormality.
  • a polynucleotide or polypeptide of the present invention may increase chemotaxic activity of particular cells. These chemotactic molecules can then be used to treat inflammation, infection, hyperproliferative disorders, or any immune system disorder by increasing the number of cells targeted to a particular location in the body. For example, chemotaxic molecules can be used to treat wounds and other trauma to tissues by attracting immune cells to the injured location. Chemotactic molecules of the present invention can also attract fibroblasts, which can be used to treat wounds. It is also contemplated that a polynucleotide or polypeptide of the present invention may inhibit chemotactic activity. These molecules could also be used to treat disorders. Thus, a polynucleotide or polypeptide of the present invention could be used as an inhibitor of chemotaxis.
  • a polypeptide of the present invention may be used to screen for molecules that bind to the polypeptide or for molecules to which the polypeptide binds.
  • the binding of the polypeptide and the molecule may activate (i.e., an agonist), increase, inhibit (i.e., an antagonist), or decrease activity of the polypeptide or the molecule bound.
  • Examples of such molecules include antibodies, oligonucleotides, proteins (e.g., receptors),or small molecules.
  • the molecule is closely related to the natural ligand of the polypeptide, e.g., a fragment of the ligand, or a natural substrate, a ligand, a structural or functional mimetic (see, Coligan et al., Current Protocols in Immunology, 1(2), Chapter 5 (1991)).
  • the molecule can be closely related to the natural receptor to which the polypeptide binds, or at least, a fragment of the receptor capable of being bound by the polypeptide (e.g., an active site). In either case, the molecule can be rationally designed using known techniques.
  • the screening for these molecules involves producing appropriate cells which express the polypeptide, either as a secreted protein or on the cell membrane.
  • Preferred cells include cells from mammals, yeast, Drosophila, or E. coli. Cells expressing the polypeptide (or cell membrane containing the expressed polypeptide) are then preferably contacted with a test compound potentially containing the molecule to observe binding, stimulation, or inhibition of activity of either the polypeptide or the molecule.
  • the assay may simply test binding of a candidate compound to the polypeptide, wherein binding is detected by a label, or in an assay involving competition with a labeled competitor. Further, the assay may test whether the candidate compound results in a signal generated by binding to the polypeptide.
  • the assay can be carried out using cell-free preparations, polypeptide/molecule affixed to a solid support, chemical libraries, or natural product mixtures.
  • the assay may also simply comprise the steps of mixing a candidate compound with a solution containing a polypeptide, measuring polypeptide/molecule activity or binding, and comparing the polypeptide/molecule activity or binding to a standard.
  • an ELISA assay can measure polypeptide level or activity in a sample (e.g., biological sample) using a monoclonal or polyclonal antibody.
  • the antibody can measure polypeptide level or activity by either binding, directly or indirectly, to the polypeptide or by competing with the polypeptide for a substrate.
  • All of these above assays can be used as diagnostic or prognostic markers.
  • the molecules discovered using these assays can be used to treat disease or to bring about a particular result in a patient (e.g., blood vessel growth) by activating or inhibiting the polypeptide/molecule.
  • the assays can discover agents which may inhibit or enhance the production of the polypeptide from suitably manipulated cells or tissues.
  • the invention includes a method of identifying compounds which bind to a polypeptide of the invention comprising the steps of: (a) incubating a candidate binding compound with a polypeptide of the invention; and (b) determining if binding has occurred.
  • the invention includes a method of identifying agonists/antagonists comprising the steps of: (a) incubating a candidate compound with a polypeptide of the invention, (b) assaying a biological activity, and (c) determining if a biological activity of the polypeptide has been altered.
  • a polypeptide or polynucleotide of the present invention may also increase or decrease the differentiation or proliferation of embryonic stem cells, besides, as discussed above, hematopoietic lineage.
  • a polypeptide or polynucleotide of the present invention may also be used to modulate mammalian characteristics, such as body height, weight, hair color, eye color, skin, percentage of adipose tissue, pigmentation, size, and shape (e.g., cosmetic surgery).
  • a polypeptide or polynucleotide of the present invention may be used to modulate mammalian metabolism affecting catabolism, anabolism, processing, utilization, and storage of energy.
  • a polypeptide or polynucleotide of the present invention may be used to change a mammal's mental state or physical state by influencing biorhythms, circadian rhythms, depression (including depressive disorders), tendency for violence, tolerance for pain, the response to opiates and opioids, tolerance to opiates and opioids, withdrawal from opiates and opioids, reproductive capabilities (preferably by activin or inhibin-like activity), hormonal or endocrine levels, appetite, libido, memory, stress, or other cognitive qualities.
  • a polypeptide or polynucleotide of the present invention may also be used as a food additive or preservative, such as to increase or decrease storage capabilities, fat content, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional components.
  • a polynucleotide of the invention is down-regulated and exacerbates a pathological condition, such as psychosis or other neuropsychiatric disorders
  • the expression of the polynucleotide can be increased or the level of the intact polypeptide product can be increased in order to treat, prevent, ameliorate, or modulate the pathological condition. This can be accomplished by, for example, administering a polynucleotide or polypeptide of the invention to the mammalian subject.
  • a polynucleotide of the invention can be administered to a mammalian subject by a recombinant expression vector comprising the polynucleotide.
  • a mammalian subject can be a human, baboon, chimpanzee, macaque, cow, horse, sheep, pig, horse, dog, cat, rabbit, guinea pig, rat or mouse.
  • the recombinant vector comprises a polynucleotide shown in SEQ ID NOs: 1-19; 49-52; 57-72 and 107 or a polynucleotide which is at least 98% identical to a nucleic acid sequence shown in SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • the recombinant vector comprises a variant polynucleotide that is at least 80%, 90%, or 95% identical to a polynucleotide comprising SEQ ID NOs: 1-19; 49-52; 57-72 and 107.
  • a polynucleotide or recombinant expression vector of the invention can be used to express a polynucleotide in said subject for the treatment of, for example, psychosis or other neuropsychiatric disorder.
  • Expression of a polynucleotide in target cells including but not limited to cells of the striatum and nucleus accumbens, would effect greater production of the encoded polypeptide.
  • the regulation of other genes may be secondarily up- or down-regulated.
  • a naked polynucleotide can be administered to target cells.
  • Polynucleotides and recombinant expression vectors of the invention can be administered as a pharmaceutical composition.
  • Such a composition comprises an effective amount of a polynucleotide or recombinant expression vector, and a pharmaceutically acceptable formulation agent selected for suitability with the mode of administration.
  • Suitable formulation materials preferably are non-toxic to recipients at the concentrations employed and can modify, maintain, or preserve, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, rate of dissolution or release, adsorption, or penetration of the composition. See Remington 's Pharmaceutical Sciences (18 th Ed., A.R. Gennaro, ed., Mack Publishing Company 1990).
  • the dosage regimen for treating a disease with a composition comprising a polynucleotide or expression vector is based on a variety of factors, including the type or severity of the psychosis or other neuropsychiatric disorders, the age, weight, sex, medical condition of the patient, the route of administration, and the particular compound employed. Thus, the dosage regimen may vary widely, but can be determined routinely using standard methods. A typical dosage may range from about 0.1 mg/kg to about 100 mg/kg or more, depending on the factors mentioned above.
  • the cells of a mammalian subject may be transfected in vivo, ex vivo, or in vitro.
  • Administration of a polynucleotide or a recombinant vector containing a polynucleotide to a target cell in vivo may be accomplished using any of a variety of techniques well known to those skilled in the art.
  • U.S. Patent No. 5,672,344 describes an in vivo viral-mediated gene transfer system involving a recombinant neurotrophic HSV-1 vector.
  • compositions of polynucleotides and recombinant vectors can be transfected in vivo by oral, buccal, parenteral, rectal, or topical administration as well as by inhalation spray.
  • a polynucleotide of the invention is up-regulated and exacerbates a pathological condition in a mammalian subject, such as psychosis or other neuropsychiatric disorders
  • the expression of the polynucleotide can be blocked or reduced or the level of the intact polypeptide product can be reduced in order to treat, prevent, ameliorate, or modulate the pathological condition.
  • This can be accomplished by, for example, the use of antisense oligonucleotides or ribozymes.
  • drags or antibodies that bind to and inactivate the polypeptide product can be used.
  • Antisense oligonucleotides can be deoxyribonucleotides, ribonucleotides, or a combination of both. Oligonucleotides can be synthesized manually or by an automated synthesizer, by covalently linking the 5' end of one nucleotide with the 3' end of another nucleotide with non-phosphodiester internucleotide linkages such alkylphosphonates, phosphorothioates, phosphorodithioates, alkylphosphonothioates, alkylphosphonates, phosphoramidates, phosphate esters, carbamates, acetamidate, carboxymethyl esters, carbonates, and phosphate triesters.
  • Modifications of gene expression can be obtained by designing antisense oligonucleotides which will form duplexes to the control, 5', or regulatory regions of a gene of the invention. Oligonucleotides derived from the transcription initiation site, e.g., between positions -10 and +10 from the start site, are preferred. Similarly, inhibition can be achieved using "triple helix" base-pairing methodology. Triple helix pairing is useful because it causes inhibition of the ability of the double helix to open sufficiently for the binding of polymerases, transcription factors, or chaperons.
  • An antisense oligonucleotide also can be designed to block translation of mRNA by preventing the transcript from binding to ribosomes. Precise complementarity is not required for successful complex formation between an antisense oligonucleotide and the complementary sequence of a polynucleotide.
  • Antisense oligonucleotides which comprise, for example, 2, 3, 4, or 5 or more stretches of contiguous nucleotides which are precisely complementary to a polynucleotide, each separated by a stretch of contiguous nucleotides which are not complementary to adjacent nucleotides, can provide sufficient targeting specificity for mRNA.
  • each stretch of complementary contiguous nucleotides is at least 4, 5, 6, 7, or 8 or more nucleotides in length.
  • Non-complementary intervening sequences are preferably 1 , 2, 3, or 4 nucleotides in length.
  • One skilled in the art can easily use the calculated melting point of an antisense-sense pair to determine the degree of mismatching which will be tolerated between a particular antisense oligonucleotide and a particular polynucleotide sequence.
  • a first biological sample from a patient suspected of having a pathological condition is obtained along with a second sample from a suitable comparable control source.
  • a biological sample can comprise saliva, blood, cerebrospinal fluid, amniotic fluid, urine, feces, or tissue, such as gastrointestinal tissue.
  • a suitable control source can be obtained from one or more mammalian subjects that do not have the pathological condition.
  • the average concentrations and distribution of a polynucleotide or polypeptide of the invention can be determined from biological samples taken from a representative population of mammalian subjects, wherein the mammalian subjects are the same species as the subject from which the test sample was obtained.
  • a reagent is typically affixed to a solid matrix by adso ⁇ tion from an aqueous medium although other modes of affixation applicable to proteins and polypeptides can be used that are well known to those skilled in the art. Exemplary adso ⁇ tion methods are described herein. Useful solid matrices are also well known in the art.
  • mice Male C57B1/6J mice (20-28 g) were housed in groups of four on a standard 12/12 hour light-dark cycle with ad libitum access to standard laboratory chow and tap water. For the experimental paradigms, mice were divided into groups of 25 and subjected to the following treatments: Control groups: Mice received a single injection of sterile saline (0.1 ml volume), or no injection, and were sacrificed after 45 minutes.
  • the method yields Digital Sequence Tags (DSTs), that is, polynucleotides that are expressed sequence tags of the 3' end of mRNAs. DSTs that showed changes in relative levels as a result of clozapine treatment were selected for further study. The intensities of the laser-induced fluorescence of the labeled PCR products were compared across samples isolated from the striatum/nucleus accumbens of mice treated with clozapine for 45 minutes, 7 hours, 5 days, 12 days, or 14 days.
  • DSTs Digital Sequence Tags
  • Each biotinylated double stranded cDNA sample was cleaved with the restriction endonuclease MspL which recognizes the sequence CCGG.
  • the resulting fragments of cDNA corresponding to the 3' region of the starting mRNA were then isolated by capture of the biotinylated cDNA fragments on a streptavidin-coated substrate.
  • Suitable streptavidin-coated substrates include microtitre plates, PCR tubes, polystyrene beads, paramagnetic polymer beads and paramagnetic porous glass particles.
  • a preferred streptavidin-coated substrate is a suspension of paramagnetic polymer beads (Dynal, Inc., Lake Success, NY).
  • the cDNA fragment product was released by digestion with Notl, which cleaves at an 8-nucleotide sequence within the anchor primers but rarely within the mRNA-derived portion of the cDNAs.
  • Notl which cleaves at an 8-nucleotide sequence within the anchor primers but rarely within the mRNA-derived portion of the cDNAs.
  • the 3' Mspl-Notl fragments which are of uniform length for each mRNA species, were directionally ligated into Clal- Notl- cleaved plasmid pBC SK + (Stratagene, La Jolla, CA) in an antisense orientation with respect to the vector's T3 promoter, and the product used to transform Escherichia coli SURE cells (Stratagene).
  • each of the cRNA preparations was processed in a three-step fashion.
  • 250ng of cRNA was converted to first-strand cDNA using the 5' RT primer (A-G-G-T-C-G-A-C-G-G-T-A-T-C-G-G, (SEQ ID NO: 21).
  • Table 3 contains primers generated from each of the cloned DSTs used in such studies.
  • the TOGA PCR product was sequenced using a modification of a direct sequencing methodology (Innis et al., Proc. Nat'l. Acad. Sci., 85: 9436-9440 (1988)).
  • PCR was performed using the following program: 94°C, 4 minutes and 25 cycles of 94°C, 20 seconds; 65°C, 20 seconds; 72°C, 20 seconds; and 72°C 4 minutes.
  • the resulting amplified adapted PCR product was gel purified as described above.
  • the purified ds-extended PCR product was sequenced using a standard protocol for ABI 3700 sequencing. Briefly, triplicate reactions in forward and reverse orientation (6 total reactions) were prepared, each reaction containing 5 ⁇ l of gel purified ds- extended N5 PCR product as template.
  • the 3' sequencing primer was the sequence 5' GGT GGC GGC CGC AGG AAT TTT TTT TTT TTT TT 3', (SEQ ED NO: 93).
  • PCR was performed using the following thermal cycling program: 96°C, 2 minutes and 29 cycles of 96°C, 15 seconds; 50°C, 15 seconds; 60°C, 4 minutes.
  • oligonucleotides were synthesized with the sequence G-A-T-C- G-A-A-T-C extended at the 3' end with a partial Mspl site (C-G-G) and an additional 18 nucleotides adjacent to the partial Mspl site from the sequence determined by direct sequencing.
  • the 5' PCR primers were paired with the fluorescent labeled universal 3' PCR primer (SEQ ID NO: 23) in PCR reactions with the Nl TOGA PCR reaction product as template. The lengths of these PCR products were compared to the length of the PCR products of interest. Table 3 contains the sequences of the primers used in these studies.
  • CLZ_17 (SEQ ID NO: 49); CLZ_26, (SEQ ID NO: 50); CLZ 28, (SEQ ID NO: 51); and CLZ_ 58 (SEQ ID NO: 52) the sequences listed for the TOGA PCR products were derived from candidate matches to sequences present in available Genbank, EST, or proprietary databases. Table 4 lists the candidate matches for each by accession number of the Genbank entry or by the accession numbers of a set of computer-assembled ESTs used to create a consensus sequence.
  • PCR primers were designed with the sequence G-A-T- C-G-A-A-T-C extended at the 3' end with a partial Mspl site (C-G-G), and an additional 18 nucleotides adjacent to the terminal Mspl site in the candidate match sequence.
  • Each extended primer is combined with the fluorescent labeled universal 3' PCR primer (SEQ ID NO: 23) in a PCR reaction with the product of the first TOGA PCR reaction (Nl reaction products) as the template.
  • the PCR products obtained using an extended primer and the universal 3' primer were compared to products obtained using the original TOGA PCR primers.
  • oligonucleotides were synthesized co ⁇ esponding to the 5' PCR primer in the second PCR step for each candidate extended at the 3' end with an additional 12-15 nucleotides from the cloned sequence.
  • the 5' PCR primer was G-A-T-C-G-A-A-T-C-C-G-G-C-A-C-C-T- A-C-T-G-G-A-T-C-C-T-G-G (SEQ ID NO: 29).
  • This 5' PCR primer were paired with the fluorescently labeled 3' PCR primer (SEQ ID NO: 23) in PCRs using the cDNA produced in the first PCR reaction as substrate.
  • RNA transcripts were fractionated by electrophoresis on a 1.5% agarose gel containing formaldehyde, transfe ⁇ ed to a biotrans membrane by the method of Thomas (Thomas, P. S., Proc. Natl. Acad. Sci., 77,5201-5215 (1980)), and prehybridized for 30 minutes in Expresshyb (Clonetech).
  • a 160 bp insert of CLZ_5 (25- 100 ng) was labeled with [ ⁇ - 32 P]-d CTP by oligonucleotide labeling to specific activities of approximately 5xl0 8 cpm/ ⁇ g, added to the prehybridization solution and incubated for 1 hour. Filters were washed to high stringency (0.2 X SSC) (1 X SSC: 0.015 M NaCl and 0.0015 M Na citrate) at 68°C then exposed to Kodak X-AR film (Eastman Kodak, Rochester, NY) for up to 1 week. Densitrometry analysis on Northern blots was performed by ImageQuant software.
  • a 900 bp mRNA was detected in control and clozapine- treated mice which corresponds with the apoD gene.
  • the apoD mRNA expression is progressively up-regulated with clozapine treatment over the two-week time course. It is possible that clozapine may mediate its antipsychotic effect through the regulation of apoD. Alternatively, apoD may be co-regulated by clozapine, in parallel with the mechanism of the clozapine therapeutic effects, and can serve as an indicator of clozapine bioactive levels. Shown in Fig.
  • Figure 6 is a graphical representation comparing the results of the clozapine treatment TOGA analysis of clone CLZ 5 (CACC 201) shown in Fig. 4 and the clozapine treatment Northern Blot analysis of clone CLZ_5 shown in Figure 5.
  • the Northern Blot was imaged using a phosphoimager to determine the amount of apoD mRNA in each clozapine-treated sample relative to the amount of mRNA in the control sample.
  • the clozapine treatment TOGA analysis shows co ⁇ elation with the clozapine treatment Northern Blot analysis.
  • Figure 7A-C shows an in situ hybridization analysis, demonstrating the apoD expression in mouse brain.
  • the in situ hybridization was performed on free-floating sections (25 ⁇ M thick) as described (Thomas et al., J. Neurosci. Res., 52, 118-124 (1998)). Coronal sections were hybridized at 55°C for 16 hours with an S-labeled, single-stranded 160 bp antisense cRNA probe of CLZ 5 at IO 7 cpm/ml. The probe was synthesized from the 3 '-ended cDNA TOGA clone CLZ-5 using the Maxiscript Transcription Kit (Ambion, Austin, TX).
  • Fig. 7A shows CLZ-5 (apoD) mRNA expression in mouse anterior brain
  • 7B shows apoD mRNA expression in midbrain
  • 7C shows apoD expression in posterior brain.
  • apoD is expressed by astroglial cells, pial cells, perivascular fibroblasts and scattered neurons. This is consistent with previous studies examining the expression of apoD in mice, rabbits and humans (Yoshida et al., DNA and Cell Biology, 15, 873-882 (1996); Provost et al., J. Lipid Res., 32, 1959-1970 (1991); Nava ⁇ o et al., Neurosci. Lett., 254, 17-20 (1998).
  • Figure 8A-I presents an in situ hybridization analysis, showing clone CLZ_5 (apoD) mRNA expression in mouse anterior (8 A-C), mid (8D-F), and posterior (8G-I) brain following saline treatment (top row) or clozapine treatment (7.5 mg/kg) for 5 days (middle row) and 14 days (bottom row), using previously described methods.
  • Figure 9A-H shows a darkfield photomicrograph demonstrating upregulated apoD mRNA expression in various brain regions, including the co ⁇ us callosum (cc, Fig. 9A, E); caudate putamen (CPu, Fig. 9B, 7F); anterior commissure (aca, Fig. 9C, 9G); and globus pallidus (GP, Fig. 9D, 9H).
  • In situ hybridizations were perfomed as described above, using an antisense 35 S-labeled apoD riboprobe on brains from control (Fig. 9A-D) and clozapine-treated (Fig. 9E-H) animals.
  • the observed upregulation of apoD was due to an increase in the amount of apoD expressed per cell.
  • Figure IOA B shows a darkfield photomicrograph demonstrating upregulated apoD mRNA expression in the internal capsule (ic).
  • Figure IOC D shows a brightfield view of the optic tract (opt) demonstrating upregulation of apoD expression in oligodendrocytes.
  • In situ hybridizations were perfomed as described above, using an antisense 35 S-labeled apoD riboprobe on brains from control (IOA, C) and clozapine- treated (10B, D) animals.
  • the cells prominantly expressing apoD in the optic tract have a box-like mo ⁇ hology and are lined up in a serial a ⁇ ay, presumably along axonal tracts.
  • White matter tracts comprise nerve fiber bundles connecting different regions of the brain.
  • the predominant cells in these regions are astrocytes and oligodendrocytes, both of which have been shown to express apoD (Boyles et al., J Lipid Res 31:2243-2256 (1990); Nava ⁇ o et al, Neurosci Lett 254:17-20 (1995); Provost et al., J Lipid Res 32 (1991)).
  • apoD riboprobe 35 S-labeled apoD riboprobe in combination with either an antibody specific for an astrocyte marker, glial fibrillary acidic protein (GFAP), or an antibody specific for an oligodendrocyte marker, 2', 3'- cyclic nucleotide 3'-phosphodiesterase (CNP) (Boehringer Mannheim, Germany).
  • GFAP glial fibrillary acidic protein
  • CNP 2', 3'- cyclic nucleotide 3'-phosphodiesterase
  • the immunoreaction was detected with Vectastain ABC TM kit (Vector Laboratory, Inc., Burlingame, CA) according to the manufacturer's instructions.
  • Fig. 11 shows sections of striatum and optic tract in control and clozapine-treated animals. Thick arrows indicate the co-localization of GFAP and apoD, while thin arrows indicate the expression of apoD alone.
  • Fig. 11A, B shows that in untreated striatum, many GFAP -positive cells in both gray and white matter regions are positive for apoD.
  • Fig. 1 ID, E shows that in brain from clozapine-treated animals, an increase in the amount of apoD was observed in a small subset of GFAP-positive cells in the striatum.
  • Fig. 1 IC shows GFAP and apoD co-localization in the optic tract in control
  • Fig. 1 IG H shows apoD immunohistochemistry with an anti-human apoD primary antibody (Novocastra, Newcastle, UK) in the optic tract of control saline (1 IG) and clozapine-treated animals (11H).
  • cytoplasmic RNA from glial cell cultures were elect ⁇ ophoresed on a 1.5% agarose gel containing formaldehyde, blotted, and probed as previously described.
  • apoD mRNA levels were down-regulated in mixed glial cell cultures treated with clozapine (both 100 nM and 1 ⁇ M) for 1 week, suggesting that perhaps neurons and glia display different mechanisms for apoD regulation.
  • TOGA methodology, Northern blot analyses, and in situ hybridization studies have demonstrated an increase in apoD mRNA expression in both white and gray matter regions of mouse brain in response to chronic clozapine administration.
  • Colocalization studies, combining in situ hybridization and imunohistochemistry methods have revealed that apoD mRNA levels are increased in both neurons and glial cells with clozapine administration. The evidence indicates that the glial cells responsible for the most dramatic increases in apoD expression are primarily oligodendrocytes, but a subset of astrocytes also have increased apoD expression after clozapine treatment.
  • TOGA, Northern blot and in situ hybridization analyses showed that apoD expression was not affected by haloperidol treatment.
  • apoD is regulated by chronic antipsychotic drag administration
  • studies using schizophrenic and bipolar human subjects showed that apoD expression is increased in the prefrontal cortex of such patients.
  • the combined results suggest that apoD is a marker for neuropathology associated with psychiatric disorders and therefore can be used to target abnormalities in specific anatomical brain regions.
  • ApoD was initially identified as a constituent of plasma high-density lipoproteins (HDLs), which also contain phospholipids, cholesterol and fatty acids (McConathy et al., Fed. Eur. Biochem. Soc. Lett, 37: 178 (1973)).
  • HDLs plasma high-density lipoproteins
  • apoD is thought to play a role in reverse cholesterol transport, the removal of excess cholesterol from tissues to the liver for catabolism (Oram et al., J. Lipid. Res., 37: (1996)).
  • apoD is major protein component in cyst fluid from women with human breast cystic disease (Balbin et al., Biochem.
  • apoD is expressed primarily in glial cells, pial cells, perivascular cells, and some neuronal populations (Nava ⁇ o et al., Neurosci.
  • ApoD has also been shown to bind arachidonic acid Morais-Cabral et al., FEBSLett., 366: 53 (1995)) implicating it in functions associated with cell membrane remodeling and prostaglandin synthesis.
  • a process that involves massive membrane degradation and lipid release apoD concentrations are increased 500-fold (Boyles et al., J. Biol. Chem., 265: 17805 (1990)).
  • apoD may play an important role in psychotic disease. It is widely believed that imbalances in basal ganglia circuitry contribute to psychotic behaviors and that blockade of specific receptors in these regions is responsible for neuroleptic action. The neuronal increases in apoD mRNA expression observed in neurons of the striatum and globus pallidus are consistent with this hypothesis.
  • the internal capsule consists of massive nerve fibers connecting the thalamus to the cortex and is an area of convergence for the fiber tracts running transversely through the striatum.
  • the thalamus is a relay station for virtually all information passing to the cortex and coordinated cortico-thalamic activity is essential for normal consciousness. Recent theories have associated psychotic behavior with disraptions in cortico-thalamic oscillations.
  • An upregulation of apoD expression in the internal capsule may play a role in restoring the proper balance of neuronal communication.
  • HDLs plasma high-density lipoproteins
  • LDL and VLDL plasma lipoproteins
  • HDLs protect against cardiovascular disease by removing excess cholesterol from cells of arterial walls. This removal involves the direct interaction of HDL lipoproteins with plasma membrane domains and subsequent transport to the liver for catabolism (Oram, et al., J. Lipid Res., 37, 2473-2491 (1996)).
  • apoD is synthesized and secreted by cultured astrocytes, which secretion has been shown to increase in the presence of cholesterol derivatives (Patel, et al., Neuroreport 6, 653-657 (1995)). Further, it has also been demonstrated that apoD levels are increased in Niemann Pick Disease, type C, which is associated with elevated levels of cholesterol. These studies provide evidence of a functionally significant role for apoD in cholesterol transport in the CNS.
  • Membrane phospholipids act as precursors in numerous signaling systems (e.g., inositol phosphates, arachidonic acid, platelet activation factors, and eicosaniods) and comprise the membrane environment for neurotransmitter-mediated signal transduction.
  • signaling systems e.g., inositol phosphates, arachidonic acid, platelet activation factors, and eicosaniods
  • altered membrane phospholipid metabolism could have significant consequences for neuronal communication, resulting in behavioral abnormalities. Alterations in plasma membrane structure and function may result from the altered content and distribution of membrane lipids and fatty acids, such as arachidonic acid.
  • Arachidonic acid is released by the action of numerous phospholipase enzymes, primarily phospholipase A2, and is a substrate for prostglandins and leukotriene synthesis. While the molecular mechanisms underlying abnormalities in the complex system of phospolipid biochemistry are not known, several groups have demonstrated an increase in phospholipase A2 activity in the plasma and brains of schizophrenic patients (Gattaz et al., Biol Psychiatry., 22, 421-426 (1987); Ross et al., Arch. Gen. Psychiatry., 54, 487-494 (1997); Ross et al., Brain Research, 821, 407-413 (1999)).
  • phospholipase A2 levels have been shown to be decreased after neuroleptic therapy (Gattaz et al., Biol. Psychiatry, 22, 421-426 (1987)).
  • Other molecular candidates implicated in psychotic disease include phospholipase C enzymes, diacyl glycerol kinases, and inositol phosphates (Ho ⁇ obin et al., Prostaglandins, Leukotrienes and Essential Fatty Acids, 60, 141-167 (1999)).
  • apoD has been shown to specifically bind arachidonic acid.
  • ApoD is an atypical apolipoprotein in that it does not share sequence homology with other apolipoproteins (Weech et al., Prog. Lipid Res., 30, 259-266 (1991)) but, rather, is a member of the lipocalin superfamily of proteins, which function in the transport of small hydrophobic molecules, including sterols, steroid hormones, and arachidonic acid (Balbin et al., Biochem. J, 271, 803-807 (1990); Dilley et al., Breast Cancer Res.
  • apoD can affect fatty acid composition, cholesterol levels and membrane phospholipids, all of which will affect plasma membrane composition and structure. Also, since apoD specifically binds cholesterol, arachidonic acid and other lipids, alterations in the levels of apoD can affect lipid metabolism and signal transduction by affecting substrate availability for these pathways.
  • apoD may have a chromosomal linkage with schizophrenia.
  • the chromosomal location of apoD is 3q26.
  • Genetic studies have implicated a potential association between schizophrenia and chromosome 3q, however the linkage is relatively inconsistent (reviewed by Maier, et al., Curr. Opin. Psych., 11, 19-25 (1998)).
  • a serotonin sub-type such as 5HT 2a and 5HT 2c may provide a pharmacological mechanism for clozapine's effect on apoD expression.
  • Preliminary results demonstrate that treatment with ketanserin and mesulergine, 5HT 2a / 2c and 5HT c - selective antagonists respectively, results in an apparent upregulation of apoD mRNA expression in mouse brain.
  • the striatum expresses a number of 5HT receptor subtypes, including the 5HT 2c , which subtype may mediate clozapine's effect on apoD expression.
  • cultured glial cells or astrocytes do not appear to express 5HT 2c receptors.
  • the downregulation observed in these cells may reflect actions at a different 5HT subtype, such as 5HT 2a , or a different receptor.
  • 5HT 2a a different receptor
  • ketanserin has been associated with a decrease in total cholesterol levels and an upregulation of another apolipoprotein, apo Al (Loschiavo, et al., Int. J. Clin. Pharmacol. Ther. Toxicol, 28, 455-457 (1990)).
  • apo Al apolipoprotein
  • the similar effects observed by both ketanserin and clozapine suggest that they may be working through the same receptor subtype(s).
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in Example 1.
  • the same experimental paradigm used in Example 1 for clozapine treatment was used for the various analyses described below. Briefly, in the clozapine studies, the control group mice received a single injection of sterile saline (0.1 ml volume), or no injection, and were sacrificed after 45 minutes. The mice subjected to acute clozapine treatment were given a single intraperitoneal injection of clozapine (7.5 mg/kg) and sacrificed after 45 minutes or 7 hours, as described in Example 1. The mice subjected to chronic clozapine treatment received daily subcutaneous injections of clozapine (7.5 mg/kg) for 5 days, 12 days or 14 days.
  • the mRNA was prepared according to the method described in Example 2.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in Example 1 and divided into the following groups:
  • mice were subcutaneusly implanted with one placebo pellet upon halothane anaesthesia;
  • mice received a mo ⁇ hine intraperitoneal injection of 10 mg/kg; 3) a chronic or tolerant group, in which mice were rendered drag-tolerant and dependent by means of subcutaneous implantation of a single pellet containing 75 mg of mo ⁇ hine free base for 3 days; and
  • mice rendered tolerant to mo ⁇ hine were injected intraperitoneally with naltrexone 1 mg/kg.
  • Animals were sacrificed in their cages with CO 2 at 72 hours after placebo or mo ⁇ hine pellet implantation, or 4 hours after single injection of mo ⁇ hine, or 4 hours after administration of naltrexone to mo ⁇ hine-tolerant mice. Their brains were rapidly removed.
  • the striatum, including the nucleus accumbens, and block of tissues containing the amygdala complex were dissected under microscope and collected in ice-cold RNA extraction buffer.
  • the TOGA data shown in Figures 13 and 14 were generated with a 5' -PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-T-T-G-T; SEQ ID NO: 26) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • Figs. 13 and 14 show PCR products produced from mRNA isolated from the striatum/nucleus accumbens of mice treated with clozapine (Fig. 13) or mo ⁇ hine (Fig. 14).
  • the vertical index line indicates a PCR product of about 266 b.p.
  • the vertical index line indicates a PCR product of about 266 b.p. that is present in control cells, and whose expression differentially regulated in control striatum (PS), acutely treated striatum (AS), withdrawal striatum (WS), control amygdala (PA), acutely treated amygdala (AA), chronically treated amygdala (TA), and withdrawal amygdala (WA).
  • PS control striatum
  • AS acutely treated striatum
  • WS withdrawal striatum
  • PA control amygdala
  • AA acutely treated amygdala
  • TA chronically treated amygdala
  • WA withdrawal amygdala
  • the expression of CLZ_40 product is greater in striatum than in amygdala.
  • CLZ_40 displays chronic-specific or withdrawal-specific regulation in both of these brain regions.
  • CLZ_40 is downregulated in withdrawal striatum but not acutely treated striatum.
  • CLZ_40 is slightly upregulated in acutely treated amygdala and increasingly upregulated in chronically treated amygdala and withdrawal amygdala.
  • Northern Blot analysis was performed using mRNA extracted from the striatum/nucleus accumbens of control mice and clozapine-treated mice. Briefly, an agarose gel containing 2 ⁇ g of poly A enriched mRNA as well as size standards was electrophoresed on a 1.5% agarose gel containing formaldehyde, transferred to a biotrans membrane, and prehybridized for 30 minutes in Expresshyb (Clonetech).
  • Figure 16 is a graphical representation comparing the results of the clozapine treatment TOGA analysis of clone CLZ_40 shown in Fig. 13 and the clozapine treatment Northern Blot analysis of clone CLZ_40 shown in Figure 15.
  • the Northern Blot was imaged using a phosphoimager to determine the amount of CLZ 40 mRNA in each clozapine-treated sample relative to the amount of mRNA in the control sample.
  • the clozapine treatment TOGA analysis shows co ⁇ elation with the clozapine treatment Northern Blot analysis.
  • Figure 17A-B is an in situ hybridization analysis, demonstrating CLZ 40 mRNA expression in the mouse brain. In situ hybridization was performed on free-floating sections (25 ⁇ M thick). Coronal sections were hybridized at 55°C for 16 hour with an
  • CLZ 40 (SEQ ID NO: 12) is of unknown identity.
  • the CLZ_40 DST has been PCR amplified and the extended sequence clone of CLZ 40 (SEQ ID NO: 13) matches an EST in the GenBank database (AI509550) as shown in Table 4.
  • the observation that CLZ_40 is down-regulated with clozapine treatment suggests a potential association with the therapeutic effects of clozapine.
  • its highly unique gene expression pattern is like no other gene identified to date, and its presence in the nucleus accumbens may implicate CLZ 40 in a number of functional roles associated with this structure, namely limbic/mental behavior and addiction.
  • Addiction to opiates and other drags of abuse is a chronic disease of the brain, most likely resulting from molecular and cellular adaptations of specific neurons to repeated exposure to opiates (Leshner, A., Science, 278, 45-47 (1997)).
  • An important neural substrate implicated in the opioid reinforcement and addiction is the mesolimbic system, notably the nucleus accumbens (Everitt, et al, Ann. NY. Acad. Sci., 877, 412-438 (1999)). All highly addictive drags, such as opiates, cocaine and amphetamines, produce adaptations in the neural circuitry of the nucleus accumbens, but the precise relationships are unknown.
  • CLZ 40 is a likely candidate for involvement in such mechanisms due to its specific expression in the nucleus accumbens. Elucidation of the biology underlying psychoses and addiction is key to understanding the underlying causes of such disorders and may lead to the development of more effective treatments, including anti-addiction medications.
  • the hippocampal system has long been associated with learning and memory, including forms of conditional associative learning (Sziklas, et al., Hippocampus, 8, 131-137 (1998)), which is the form of learning associated with addiction (Di Chiara, et al., Ann. N.Y. Acad. Sci., 877, 461-85 (1999)).
  • conditional associative learning Sziklas, et al., Hippocampus, 8, 131-137 (1998)
  • the expression of CLZ 40 in the hippocampus suggests that this gene may provide a link with such learning processes.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses described below.
  • FIG. 18 shows PCR products produced from mRNA isolated from the striatum/nucleus accumbens of mice treated with clozapine for various lengths of time as described in Example 1.
  • the vertical index line indicates a PCR product of about 89 b.p. that is present in control cells, and whose expression in the striatum/nucleus accumbens of mice treated with clozapine is differntially regulated with acute treatment versus chronic treatment.
  • CLZ_34 is upregulated with clozapine treatment at 45 minutes and 7 hours, but decreases to control level by day 5 and remains at about control level for as long as 12 days, showing a slight increase at day 14.
  • In situ analysis performed using CLZ_34 as a probe revealed that CLZ_34 is expressed ubiquitously throughout the brain (data not shown).
  • CLZ_34 co ⁇ esponds with GenBank sequence UO8262, which is identified as a rat N-methyl-D-aspartate receptor/NMD AR1 -2a subunit (NMDAR1).
  • NMDAR1 receptor is a glutamate receptor involved in the processes underlying learning and memory.
  • numerous studies show that blockade of glutamate actions by noncompetitive (e.g. MK801 and dextrometho ⁇ han) and competitive (e.g.
  • NMDA receptor antagonists blocks or reduces the development of mo ⁇ hine tolerance following long term opiate administration (Trajillo et al., Science, 251, 85-87, (1991); Elliott et al., Pain, 56, 69-75 (1994); Wiesenfeld-Hallin, Z., Neuropsychopharm., 13, 347-56 (1995)).
  • CLZ_34 which has high homology with an NMDA receptor is interesting in view of the ability of NMDA antagonists to block the development of tolerance to opioids.
  • Figure 19 shows the consensus sequence from the computer generated assembly of the following 4 sequences AI415388: Soares mouse p3NMF19.5 Mus musculus cDNA clone IMAGE:350746 3', mRNA sequence; AI841003: UI-M-AMO- ado-e-04-O-UI.sl NIH_BMAP_MAM Mus musculus cDNA clone UI-M-AMO-ado-e- 04-0-UI 3*, mRNA sequence; AI413353: Soares mouse embryo NbME13.5 14.5 Mus musculus cDNA EMAGE:356159 3', mRNA sequence; AI425991 : Soares mouse embryo NbME13.5 14.5 Mus musculus cDNA IMAGE:426077 3', mRNA sequence. (SEQ ID NO: 53)
  • Figure 20 shows the sequence of the EST AF006196: Mus musculus metalloprotease-disintegrin MDC 15 mRNA, complete eds. (SEQ ID NO: 54)
  • Figure 21 shows the consensus sequence from the computer generated assembly of the following 3 sequences: C86593: Mus musculus fertilized egg cDNA 3'-end sequence, clone J0229E09 3', mRNA sequence; AI428410: Life Tech mouse embryo 13 5dpc 10666014 Mus musculus cDNA clone EMAGE:553802 3', mRNA sequence; AI561814: Stratagene mouse skin (#937313) Mus musculus cDNA clone IMAGE: 1227449 3', mRNA sequence. (SEQ ID NO: 55).
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-A-C-G-G; SEQ ID NO:96) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5%> acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • an agarose gel containing 2 ⁇ g of poly A enriched mRNA as well as size standards was electrophoresed on a 1.5% agarose gel containing formaldehyde, transfe ⁇ ed to a biotrans membrane, and prehybridized for 30 minutes in Expresshyb (Clonetech).
  • a CLZ_44 insert (25-100 ng) was labeled with [ ⁇ - 32 P]-d CTP by o oligonucleotide labeling to specific activities of approximately 5x10 cpm/ ⁇ g and added to the prehybridization solution and incubated 1 hour.
  • FIG. 22 is a graphical representation of the described northern blot analyses.
  • CLZ 44 was up-regulated with haloperidol and ketanserin, but not clozapine. This suggests that both dopamines D2 and 5HT 2A/2C receptors are involved in CLZ 44 expression regulation.
  • the lack of effect of clozapine may indicate that antagonism at other receptors (i.e. 5HT 3 , D4, DI) may override the effects of D2, 5HT 2 receptors.
  • Example 1 Male C57B1/6J mice (20-28 g) were housed as previously described in Example 1. The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-T-G-C-A; SEQ ID NO: 97) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • CLZ 38 is an oligodendrocyte-specific protein mRNA.
  • northern blot analyses were performed to determine the pattern of expression in the striatum/nucleus accumbens of control miceand mice treated with clozapine for 45 minutes, 7 hours, 5 days, and 2 weeks ( Figure 23).
  • CLZ_38 insert 25-100 ng was labeled with [ ⁇ - 32 P]-d CTP by oligonucleotide labeling to specific activities of approximately 5xl0 8 cpm/ ⁇ g and added to the prehybridization solution and incubated 1 hour. Filters were washed to high stringency (0.2 X SSC) (1 X SSC: 0.015 M NaCl and 0.0015 M Na citrate) at
  • Figure 23 is a graphical representation of the described northern blot analyses.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'-
  • the probe was synthesized from the 3 '-ended cDNA TOGA clone using the
  • High stringency washes were carried out at 55°C for 2 hours in 0.5 X SSC/50% formamide/0.01 M ⁇ -mercaptoethanol, and then at 68°C for 1 hour in 0.1 X SSC/0.01 M ⁇ -mercaptoethanol/0.5% sarkosyl.
  • Slices were mounted onto gelatin-coated slides and dehydrated with ethanol and chloroform before autoradiography. Slides were exposed for 1-4 days to Kodak X-AR film and then dipped in Ilford K-5 emulsion. After 4 weeks, slides were developed with Kodak D 19 developer, fixed, and counterstained with Richardson's blue stain.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • In situ hybridization was performed on free-floating sections (25 ⁇ M thick) taken from control mice and mice treated with 7.5 mg/kg clozapine for 2 weeks. Coronal sections were hybridized at 55°C for 16 hour with an 35 S-labeled, single- stranded antisense cRNA probe of CLZ_17 at IO 7 cpm ml. The probe was synthesized from the 3 '-ended cDNA TOGA clone using the Maxiscript Transcription Kit (Ambion, Austin, TX). Excess probe was removed by washing as previously described in Example 8. Slices were mounted onto gelatin-coated slides and dehydrated with ethanol and chloroform before autoradiography. Slides were exposed for 1 -4 days to Kodak X-AR film and then dipped in Ilford K-5 emulsion. After 4 weeks, slides were developed with Kodak D19 developer, fixed, and counterstained with Richardson's blue stain.
  • Figure 25A-B shows an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_17, showing the pattern of CLZ 17 mRNA expression in a coronal sections from posterior (25A) and anterior (25B) regions of mouse brain. As shown, CLZ_17 mRNA is expressed in the cortex, hippocampus, striatum, and amygdala.
  • Example 1 Male C57B1/6J mice (20-28 g) were housed as previously described in Example 1.
  • the same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-G-C-A; SEQ ID NO: 100) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • In situ hybridization was performed on free-floating sections (25 ⁇ M thick) obtained from comtrol mica nd mice treated with 7.5 mg/kg clozapine for 2 weeks. Coronal sections were hyb ⁇ dized at 55°C for 16 hour with an S-labeled, single- stranded antisense cRNA probe of CLZ 24 at IO 7 cprn/ml. The probe was synthesized from the 3 '-ended cDNA TOGA clone using the Maxiscript Transcription Kit (Ambion, Austin, TX). Excess probe was removed by washing as previously described in Example 8. Slices were mounted onto gelatin-coated slides and dehydrated with ethanol and chloroform before autoradiography.
  • Figure 26A-B shows an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ 24, showing the pattern of CLZ_24 mRNA expression in a coronal section through the hemispheres (26A) and cross section through the brainstem (26B) in mouse brain. As shown, CLZ_24 mRNA is ubiquitously expressed in the cortex.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'-
  • Figure 27A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_26, showing the pattern of CLZ 26 mRNA expression in a coronal section of the hemispheres at the level of hippocampal formation (27 A) and coronal section of the hemispheres at the level of striatum (27B) in mouse brain.
  • CLZ_26 mRNA is ubiquitously expressed in the cortex.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-G-T-A; SEQ ID NO: 102) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • Figure 28A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_28, showing the pattern of CLZ_28 mRNA expression in a coronal section through the hemispheres at the level of hippocampus (28 A) and coronal section through the posterior region of hemispheres (28B) in mouse brain.
  • CLZ 28 mRNA is expressed ubiquitously in the cortex.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'-
  • Figure 29A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_3, showing the pattern of CLZ 3 mRNA expression in a coronal section through the hemispheres at level of hippocampus
  • mice Male C57B1 6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-T-A-T-T; SEQ ED NO: 103) paired with the "universal" 3' primer (SEQ ED NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • Figure 30A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_34, showing the pattern of CLZ_34 mRNA expression in a coronal section through the hemispheres at the level of hippocampus (30A) and cross section through the midbrain (30B) in mouse brain. As shown in Figure 30A and B, CLZ 34 mRNA is ubiquitously expressed.
  • mice Male C57B1/6J mice (20-28 g) were housed as previously described in
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'-
  • Figure 31A-C is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ_43, showing the pattern of CLZ_43 mRNA expression in coronal sections of the hemispheres showing in the cortex, and intense lebelling in the striatum (31 A-C) in mouse brain. Comparison with brain sections obtained from control mice showed that CLZ_43 expression is increased approximately 10-fold by chronic treatment (2 weeks) with clozapine.
  • the open reading frame of the 1717 b.p. clone encodes a 385 amino acid peptide (SEQ ID NO: 108, SEQ ID NO: 109).
  • the following methods were used to isolate the 1717 b.p. cDNA clone.
  • the target pool was a cDNA plasmid library created from adult human brain RNA.
  • the oligonucleotide sequence used for hybridization was 5' - AAC AAG TCC GTC CTG GCA TGG-3' (SEQ ID NO:88).
  • the clone was isolated using the methods prescribed by the manufacturer of the GeneTrapper kit (Life Technologies, Inc.). Capture oligonucleotide were prepared by end-labeling the oligonucleotide with biotin-14- dCTP using terminal deoxynucloetidyl transferase. The cDNA plasmid pool was converted from double-stranded cDNA to single-stranded cDNA through the specific action of Genell protein and exonuclease III. The single-stranded cDNA pool was combined with the end-labelled oligonucleotide and hybridization was allowed to occur at room temperature for 30 minutes. The reaction was then mixed with strepavidin-coated magnetic beads.
  • the single-stranded cDNA plasmids that hybridized to the oligonucleotide were purified using a magnet to retain the magnetic beads in the reaction tube while all of the unbound components were washed away.
  • the single-stranded plasmid DNA was released from the oligonucleotide and repaired back into a double-stranded plasmid using a fresh sample of the capture oligonucleotide and DNA polymerase.
  • the repaired plasmids were transformed into bacteria and plated on an agar plate. The following day, bacterial colonies were individually picked and grown overnight. Plasmid DNA was prepared from these mini-preparations and subjected to sequence analysis.
  • Example 1 The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses.
  • the TOGA data was generated with a 5'- PCR primer (C-G-A-C-G-G-T-A-T-C-G-G-A-C-G-G; SEQ ID NO: 105) paired with the "universal" 3' primer (SEQ ID NO: 23) labeled with 6-carboxyfluorescein (6FAM, ABI) at the 5' terminus.
  • PCR reaction products were resolved by gel electrophoresis on 4.5% acrylamide gels and fluorescence data acquired on ABI377 automated sequencers. Data were analyzed using GeneScan software (Perkin-Elmer).
  • Figure 32A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ 44, showing the pattern of CLZ_44 mRNA expression in a coronal section showing labelling in the hippocampus, hypothalamus, and temporal cortex (32A) and coronal section showing cortical labelling (32B) in mouse brain.
  • Example 1 Male C57B1/6J mice (20-28 g) were housed as previously described in Example 1. The same experimental paradigm used in Example 1 for clozapine treatment was used for the TOGA analyses. The TOGA data was generated with a 5'-
  • Figure 33A-B is an in situ hybridization analysis using an antisense cRNA probe directed against the 3' end of CLZ 64, showing the pattern of CLZ 64 mRNA expression in different coronal sections of the hemispheres in mouse brain. As shown in Figure 33A and B, CLZ_64 mRNA is ubiquitously expressed.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Psychiatry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Hydrogenated Pyridines (AREA)

Abstract

L'invention concerne des polynucléotides, des polypeptides, des trousses et des méthodes liés à des gènes exprimés dans les système nerveux central et régulés par des neuroleptiques.
EP00975448A 1999-10-26 2000-10-26 Regulation de l'expression genetique au moyen d'agents neuroleptiques Withdrawn EP1226152A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16137999P 1999-10-26 1999-10-26
US161379P 1999-10-26
PCT/US2000/029690 WO2001030972A2 (fr) 1999-10-26 2000-10-26 Regulation de l'expression genetique au moyen d'agents neuroleptiques

Publications (1)

Publication Number Publication Date
EP1226152A2 true EP1226152A2 (fr) 2002-07-31

Family

ID=22580950

Family Applications (1)

Application Number Title Priority Date Filing Date
EP00975448A Withdrawn EP1226152A2 (fr) 1999-10-26 2000-10-26 Regulation de l'expression genetique au moyen d'agents neuroleptiques

Country Status (6)

Country Link
EP (1) EP1226152A2 (fr)
JP (1) JP2003533970A (fr)
AU (1) AU1349801A (fr)
CA (1) CA2389110A1 (fr)
MX (1) MXPA02004241A (fr)
WO (1) WO2001030972A2 (fr)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6960439B2 (en) 1999-06-28 2005-11-01 Source Precision Medicine, Inc. Identification, monitoring and treatment of disease and characterization of biological condition using gene expression profiles
AU2002318112B2 (en) 2001-04-10 2007-12-06 Agensys, Inc. Nucleic acids and corresponding proteins useful in the detection and treatment of various cancers
KR20040064275A (ko) 2001-11-09 2004-07-16 소스 프리시전 메디슨, 인코포레이티드 유전자 발현 프로파일을 이용한 질병의 동정, 모니터링,치료 및 생물학적 상태의 확인

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO0130972A2 *

Also Published As

Publication number Publication date
MXPA02004241A (es) 2002-10-17
WO2001030972A2 (fr) 2001-05-03
CA2389110A1 (fr) 2001-05-03
AU1349801A (en) 2001-05-08
JP2003533970A (ja) 2003-11-18
WO2001030972A3 (fr) 2002-05-10

Similar Documents

Publication Publication Date Title
US20080146505A1 (en) 47 Human Secreted Proteins
JP2002502589A (ja) 45個のヒト分泌タンパク質
CA2398255A1 (fr) Polynucleotides semblables a bcl-2, polypeptides et anticorps
US20090305991A1 (en) 33 Human Secreted Proteins
JP2002501738A (ja) 67個のヒト分泌タンパク質
JP2002506625A (ja) サイトカインレセプター共通γ鎖様
CA2394753A1 (fr) Polynucleotides et polypeptides de la protein thyrosine phosphatase humaine, et anticorps
US20070190612A1 (en) 31 Human Secreted Proteins
US20070010664A1 (en) Gene expression in the central nervous system regulated by neuroleptic agents
JP2002506875A (ja) 第18p染色体関連障害の診断及び治療のための方法及び組成物
WO2001030972A2 (fr) Regulation de l'expression genetique au moyen d'agents neuroleptiques
WO2001029179A2 (fr) Regulation de l'expression genetique par des neuromodulateurs
JP2001517093A (ja) 成長因子様タンパク質をコードするポリヌクレオチド
WO2002026936A2 (fr) Expression genetique dans le systeme nerveux central regule par des neuroleptiques
US20060142233A1 (en) Gene expression and genetic changes implicated in alcoholism
WO2001034770A2 (fr) Expression genetique modulee par l'activation de microglies ou de macrophages
CA2395054A1 (fr) Polynucleotides, polypeptides et anticorps humains
CA2309677A1 (fr) Nouveaux gene et proteine exprimes dans les tissus nerveux et pancreatiques
WO2001012647A1 (fr) Expression genique modulee en tumorigenese de l'ataxie-telangiectasie
WO2002031114A2 (fr) Expression genique module dans l'ileite
EP1250431A2 (fr) Modulation de l'expression genique dans la formation de lesions atherosclereuses grasses
WO2002042420A2 (fr) Modulation de l'expression de genes lors de l'hyperplasie de l'intima de l'artere carotide
WO2002031116A2 (fr) Expression genique modulee dans des maladies enteriques inflammatoires spontanees
WO2002081726A2 (fr) Modulation de l'expression genetique dans des modeles genetiques murins de l'atherosclerose
WO2003057825A2 (fr) Modeles d'expression genique en reponse a des agents neuroleptiques atypiques et typiques du systeme nerveux central

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20020523

AK Designated contracting states

Kind code of ref document: A2

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

AX Request for extension of the european patent

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

RIN1 Information on inventor provided before grant (corrected)

Inventor name: THOMAS, ELIZABETH, A.

Inventor name: HILBUSH, BRIAN

Inventor name: PRIBYL, THOMAS, M.

Inventor name: SUTCLIFFE, J., GREGOR

Inventor name: HASEL, KARL, W.

19U Interruption of proceedings before grant

Effective date: 20040407

19W Proceedings resumed before grant after interruption of proceedings

Effective date: 20041102

19W Proceedings resumed before grant after interruption of proceedings

Effective date: 20210201

PUAJ Public notification under rule 129 epc

Free format text: ORIGINAL CODE: 0009425

32PN Public notification

Free format text: COMMUNICATION PURSUANT TO RULE 142 EPC (RESUMPTION OF PROCEEDINGS UNDER RULE 142(2) EPC DATED 04.09.2020)

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

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

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

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

18D Application deemed to be withdrawn

Effective date: 20210803