Compositions and methods for diagnosing and treating neuropsychiatric disorders

Abstract
The present invention provides methods for diagnosing mental disorders (e.g., psychotic disorders such as schizophrenia and mood disorders such as major depression disorder and bipolar disorder). The invention also provides methods of identifying modulators of such mental disorders as well as methods of using these modulators to treat patients suffering from such mental disorders.
Description
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT

Not applicable.


BACKGROUND OF THE INVENTION

Psychotic disorders such as schizophrenia and mood disorders such as major depression and bipolar disorder are a major public health problem, affecting a significant portion of the adult population of the United States each year. While it has been hypothesized that mental disorders, including psychotic disorders such as schizophrenia as well as mood disorders such as major depression and bipolar disorder have genetic roots, little progress has been made in identifying gene sequences and gene products that play a role in causing these disorders, as is true for many diseases with a complex genetic origin (see, e.g., Burmeister, Biol. Psychiatry 45:522-532 (1999)). Relying on the discovery that certain genes expressed in particular brain pathways and regions are likely involved in the development of mental disorders, the present invention provides methods for diagnosis and treatment of mental disorders such as schizophrenia, as well as methods for identifying compounds effective in treating mental disorders.


BRIEF SUMMARY OF THE INVENTION

In order to further understand the neurobiology of psychotic disorders such as schizophrenia, the inventors of the present application have used DNA microarrays to study expression profiles of human post-mortem brains from patients diagnosed with schizophrenia. The work has focused on six brain regions that are pathways or circuits involved in schizophrenia: the anterior cingulate cortex (AnCg), dorsolateral prefrontal cortex (DLPFC), cerebellar cortex (CB), superior temporal gyrus (STG), parietal cortex (PC), and nucleus accumbens (nAcc).


The present invention demonstrates differential expression of genes in selected regions of brains of patients suffering from schizophrenia in comparison with normal control subjects. These genes include the transcripts listed in Table 1; the genes listed in Table 2 which are differentially expressed in the AnCg using Affymetrix chips and using brains with no agonal factors; the genes listed in Table 3 which are differentially expressed in the DLPFC using Affymetrix chips and using brains with no agonal factors; and the genes listed in Table 4 which are significantly dysregulated in both lymphoblastic and brain tissues.


In addition, the present invention identifies genes which are not differentially regulated in brain tissue but which are differentially regulated in lymphocytes of schizophrenic patients (Table 5). Also provided is a list (Table 6) of single nucleotide polymorphic markers which are related to aspartylglucosaminuria (AGA), a gene which is dysregulated in both brain and lymphocytes of schizophrenic patients. Tables 7 and 8 show genes that are dysregulated in schizophrenia, major depression, and bipolar disorder.


The present invention also provides genes that are differentially expressed in the amygdala in patients diagnosed with major depression disorder, bipolar disorder, and/or schizophrenia (Tables 9-13).


The present invention also provides lithium responsive genes that are differentially expressed in the amygdale of lithium treated bipolar subjects and lithium treated non-human primates (Table 14).


The present invention also provides validation of a variant version of PSPHL with an insertion deletion mutation as a useful diagnostic tool to distinguish patients with bipolar disorder among patients presenting with depression, or for diagnosis of bipolar disorder.


Genes that are differentially expressed in neuropsychiatric disorders are useful in diagnosing psychotic and mood disorders, e.g., providing SNPs, biomarkers, diagnostic probe sets for PCR and chip assays, and antigens and antibodies for immunoassays such as ELISA and immunohistochemical assays. Differential expression by brain region similarly is a useful diagnostic and therapeutic tool, as psychotic and mood disorders primarily affect certain brain regions that are part of circuits or pathways involved in the disorder. Imaging brain endogenous gene expression with sequence-specific antisense radiopharmaceuticals and novel aptamer-based probes is a powerful diagnostic tool. Those probes can be detected using both fluorescent- and radio-labels that can be used in conjunction with single-photon emission computed tomography (SPECT) or positron emission tomography (PET) imaging modalities. The identification of genes, proteins, and biochemical assays involved in psychotic and mood disorders also provides the means for drug discovery for anti-psychotic therapeutics, such as small molecules, siRNA, and antibodies.


This invention thus provides methods for determining whether a subject has or is predisposed for a mental disorder. The invention also provides methods of providing a prognosis and for monitoring disease progression and treatment. Furthermore, the present invention provides nucleic acid and protein targets for assays for drugs for the treatment of mental disorders.


In one aspect, the methods comprise the steps of: (i) obtaining a biological sample from a subject; (ii) contacting the sample with a reagent that selectively associates with a polynucleotide or polypeptide encoded by a nucleic acid that hybridizes under stringent conditions to a nucleotide sequence listed in Tables 1-14; and (iii) detecting the level of reagent that selectively associates with the sample, thereby determining whether the subject has or is predisposed for a mental disorder.


In some embodiments, the reagent is an antibody. In some embodiments, the reagent is a nucleic acid. In some embodiments, the reagent associates with a polynucleotide. In some embodiments, the reagent associates with a polypeptide. In some embodiments, the polynucleotide comprises a nucleotide sequence listed in Tables 1-14. In some embodiments, the polypeptide comprises an amino acid sequence of a gene listed in Tables 1-14. In some embodiments, the level of reagent that associates with the sample is different (i.e., higher or lower) from a level associated with humans without a mental disorder. In some embodiments, the biological sample is obtained from lymphocytes, amniotic fluid, spinal fluid, or saliva. In some embodiments, the mental disorder is a mood disorder. In some embodiments, the mental disorder is a psychotic disorder such as schizophrenia.


The invention also provides methods of identifying a compound for treatment of a mental disorder. In some embodiments, the methods comprises the steps of: (i) contacting the compound with a polypeptide, which is encoded by a polynucleotide that hybridizes under stringent conditions to a nucleic acid comprising a nucleotide sequence of Tables 1-14; and (ii) determining the functional effect of the compound upon the polypeptide, thereby identifying a compound for treatment of a mental disorder, e.g., schizophrenia.


In some embodiments, the contacting step is performed in vitro. In some embodiment, the polypeptide comprises an amino acid sequence of a gene listed in Tables 1-14. In some embodiments, the polypeptide is expressed in a cell or biological sample, and the cell or biological sample is contacted with the compound. In some embodiments, the methods further comprise administering the compound to an animal and determining the effect on the animal, e.g., an invertebrate, a vertebrate, or a mammal. In some embodiments, the determining step comprises testing the animal's mental function.


In some embodiments, the methods comprise the steps of (i) contacting the compound to a cell, the cell comprising a polynucleotide that hybridizes under stringent conditions to a nucleotide sequence of Tables 1-14; and (ii) selecting a compound that modulates expression of the polynucleotide, thereby identifying a compound for treatment of a mental disorder. In some embodiments, the polynucleotide comprises a nucleotide sequence listed in Tables 1-14. In some embodiment, the expression of the polynucleotide is enhanced. In some embodiments, the expression of the polynucleotide is decreased. In some embodiments, the methods further comprise administering the compound to an animal and determining the effect on the animal. In some embodiments, the determining step comprises testing the animal's mental function. In some embodiments, the mental disorder is a mood disorder or a psychotic disorder. In some embodiments, the psychotic disorder is schizophrenia. In some embodiment,s the mood disorder is major depression disorder or bipolar disorder.


The invention also provides methods of treating a mental disorder in a subject. In some embodiments, the methods comprise the step of administering to the subject a therapeutically effective amount of a compound identified using the methods described above. In some embodiments, the mental disorder is a mood disorder or a psychotic disorder. In some embodiments, the compound is a small organic molecule, an antibody, an antisense molecule, an aptamer, an siRNA molecule, or a peptide.


The invention also provides methods of treating mental disorder in a subject, comprising the step of administering to the subject a therapeutically effective amount of a polypeptide, which is encoded by a polynucleotide that hybridizes under stringent conditions to a nucleic acid of Tables 1-14. In some embodiments, the polypeptide comprises an amino acid sequence encoded by a gene sequence listed in Tables 1-14. In some embodiments, the mental disorder is a mood disorder or a psychotic disorder.


The invention also provides methods of treating mental disorder in a subject, comprising the step of administering to the subject a therapeutically effective amount of a polynucleotide, which hybridizes under stringent conditions to a nucleic acid of Tables 1-14. In some embodiments, the mental disorder is a mood disorder or a psychotic disorder. In some embodiments, the psychotic disorder is schizophrenia. In some embodiments, the mood disorder is a bipolar disorder or major depression.


BRIEF DESCRIPTION OF THE DRAWINGS

Table 1: Table 1 lists genes that are differentially expressed in schizophrenic versus control patients in each of the 6 brain regions. For every gene (row) listed: the UniGene ID, GenBank Accession # (“Acc”), Gene Symbol, Chromosome # (“Chr”), and direction of change (up or down) in expression levels are listed in successive columns. The last column provides the name of the differentially expressed gene and related information, where available.


Table 2: Table 2 shows gene ontology (GO) terms enriched in AnCg. Probe sets that showed GO term enrichment and met default FDR correction threshold criteria (http://brainarray.mhri.med.umich.edu/Brainarray/) are listed here. Under each enriched GO term, individual genes are listed in the rows. Information related to the LocusLink ID #, UniGene ID #, Gene Symbol and Gene Description is provided in successive columns.


Table 3: Table 3 shows GO terms enriched in DLPFC. Probe sets that showed GO-term enrichment and met default FDR correction threshold criteria (http://brainarray.mhri.med.umich.edu/Brainarray/) are listed here. Under each enriched GO term, individual genes are listed in the rows. A LocusLink ID #, UniGene ID #, Gene Symbol and Gene Description are indicated in successive columns.


Table 4: Table 4 lists 84 genes which are significantly dysregulated in both lymphoblastic and brain tissues.


Table 5. Table 5 lists 16 genes which are significantly dysregulated in lymphoblasts only. These genes were significant by microarray and the amount and direction of change was confirmed using Q-PCR. Seven of these genes (bold in Accession Number column) exhibited statistically significant dysregulation when examined by Q-PCR and evaluated using the two-tailed t-test.


Table 6. Table 6 lists 11 single nucleotide polymorphic markers correlated with aspartylglucosaminuria (AGA) gene expression which is dysregulated in both brain and lymphocytes of individuals with schizophrenia (see Table 4, infra). The regression p-values of genotype with lymphocyte gene expression are shown in the last column.


Table 7: Table 7 lists genes involved in mood disorders and psychotic disorders.


Table 8: Table 8 lists genes involved in mood disorders and psychotic disorders.


Tables 9-13: Tables 9-13 list gene differentially expressed in the amygdala that are involved in mood disorders and psychotic disorders.


Table 14: Table 14 lists lithium responsive genes expressed in the amygdala.


DEFINITIONS

A “mental disorder” or “mental illness” or “mental disease” or “psychiatric or neuropsychiatric disease or illness or disorder” refers to mood disorders (e.g., major depression, mania, and bipolar disorders), psychotic disorders (e.g., schizophrenia, schizoaffective disorder, schizophreniform disorder, delusional disorder, brief psychotic disorder, and shared psychotic disorder), personality disorders, anxiety disorders (e.g., obsessive-compulsive disorder) as well as other mental disorders such as substance-related disorders, childhood disorders, dementia, autistic disorder, adjustment disorder, delirium, multi-infarct dementia, and Tourette's disorder as described in Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, (DSM IV). Typically, such disorders have a complex genetic and/or a biochemical component.


“A psychotic disorder” refers to a condition that affects the mind, resulting in at least some loss of contact with reality. Symptoms of a psychotic disorder include, e.g., hallucinations, changed behavior that is not based on reality, delusions and the like. See, e.g., DSM IV. Schizophrenia, schizoaffective disorder, schizophreniform disorder, delusional disorder, brief psychotic disorder, substance-induced psychotic disorder, and shared psychotic disorder are examples of psychotic disorders.


“Schizophrenia” refers to a psychotic disorder involving a withdrawal from reality by an individual. Symptoms comprise for at least a part of a month two or more of the following symptoms: delusions (only one symptom is required if a delusion is bizarre, such as being abducted in a space ship from the sun); hallucinations (only one symptom is required if hallucinations are of at least two voices talking to one another or of a voice that keeps up a running commentary on the patient's thoughts or actions); disorganized speech (e.g., frequent derailment or incoherence); grossly disorganized or catatonic behavior; or negative symptoms, i.e., affective flattening, alogia, or avolition. Schizophrenia encompasses disorders such as, e.g., schizoaffective disorders. Diagnosis of schizophrenia is described in, e.g., Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition (DSM IV). Types of schizophrenia include, e.g., paranoid, disorganized, catatonic, undifferentiated, and residual.


A “mood disorder” refers to disruption of feeling tone or emotional state experienced by an individual for an extensive period of time. Mood disorders include major depression disorder (i.e., unipolar disorder), mania, dysphoria, bipolar disorder, dysthymia, cyclothymia and many others. See, e.g., DSM IV.


“Major depression disorder,” “major depressive disorder,” or “unipolar disorder” refers to a mood disorder involving any of the following symptoms: persistent sad, anxious, or “empty” mood; feelings of hopelessness or pessimism; feelings of guilt, worthlessness, or helplessness; loss of interest or pleasure in hobbies and activities that were once enjoyed, including sex; decreased energy, fatigue, being “slowed down”; difficulty concentrating, remembering, or making decisions; insomnia, early-morning awakening, or oversleeping; appetite and/or weight loss or overeating and weight gain; thoughts of death or suicide or suicide attempts; restlessness or irritability; or persistent physical symptoms that do not respond to treatment, such as headaches, digestive disorders, and chronic pain. Various subtypes of depression are described in, e.g., DSM IV.


“Bipolar disorder” is a mood disorder characterized by alternating periods of extreme moods. A person with bipolar disorder experiences cycling of moods that usually swing from being overly elated or irritable (mania) to sad and hopeless (depression) and then back again, with periods of normal mood in between. Diagnosis of bipolar disorder is described in, e.g., DSM IV. Bipolar disorders include bipolar disorder I (mania with or without major depression) and bipolar disorder II (hypomania with major depression), see, e.g., DSM IV.


Anxiety disorders, learning and memory disorders or cognitive disorders are described in DSM IV. Anxiety disorders display co-morbidity with depression, and learning and memory disorders display co-morbidity with schizophrenia.


An “agonist” refers to an agent that binds to a polypeptide or polynucleotide of the invention, stimulates, increases, activates, facilitates, enhances activation, sensitizes or up regulates the activity or expression of a polypeptide or polynucleotide of the invention.


An “antagonist” refers to an agent that inhibits expression of a polypeptide or polynucleotide of the invention or binds to, partially or totally blocks stimulation, decreases, prevents, delays activation, inactivates, desensitizes, or down regulates the activity of a polypeptide or polynucleotide of the invention.


“Inhibitors,” “activators,” and “modulators” of expression or of activity are used to refer to inhibitory, activating, or modulating molecules, respectively, identified using in vitro and in vivo assays for expression or activity, e.g., ligands, agonists, antagonists, and their homologs and mimetics. The term “modulator” includes inhibitors and activators. Inhibitors are agents that, e.g., inhibit expression of a polypeptide or polynucleotide of the invention or bind to, partially or totally block stimulation or enzymatic activity, decrease, prevent, delay activation, inactivate, desensitize, or down regulate the activity of a polypeptide or polynucleotide of the invention, e.g., antagonists. Activators are agents that, e.g., induce or activate the expression of a polypeptide or polynucleotide of the invention or bind to, stimulate, increase, open, activate, facilitate, enhance activation or enzymatic activity, sensitize or up regulate the activity of a polypeptide or polynucleotide of the invention, e.g., agonists. Modulators include naturally occurring and synthetic ligands, antagonists, agonists, small chemical molecules and the like. Assays to identify inhibitors and activators include, e.g., applying putative modulator compounds to cells, in the presence or absence of a polypeptide or polynucleotide of the invention and then determining the functional effects on a polypeptide or polynucleotide of the invention activity. Samples or assays comprising a polypeptide or polynucleotide of the invention that are treated with a potential activator, inhibitor, or modulator are compared to control samples without the inhibitor, activator, or modulator to examine the extent of effect. Control samples (untreated with modulators) are assigned a relative activity value of 100%. Inhibition is achieved when the activity value of a polypeptide or polynucleotide of the invention relative to the control is about 80%, optionally 50% or 25-1%. Activation is achieved when the activity value of a polypeptide or polynucleotide of the invention relative to the control is 110%, optionally 150%, optionally 200-500%, or 1000-3000% higher.


The term “test compound” or “drug candidate” or “modulator” or grammatical equivalents as used herein describes any molecule, either naturally occurring or synthetic, e.g., protein, oligopeptide (e.g., from about 5 to about 25 amino acids in length, preferably from about 10 to 20 or 12 to 18 amino acids in length, preferably 12, 15, or 18 amino acids in length), small organic molecule, polysaccharide, lipid, fatty acid, polynucleotide, RNAi, oligonucleotide, etc. The test compound can be in the form of a library of test compounds, such as a combinatorial or randomized library that provides a sufficient range of diversity. Test compounds are optionally linked to a fusion partner, e.g., targeting compounds, rescue compounds, dimerization compounds, stabilizing compounds, addressable compounds, and other functional moieties. Conventionally, new chemical entities with useful properties are generated by identifying a test compound (called a “lead compound”) with some desirable property or activity, e.g., inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds. Often, high throughput screening (HTS) methods are employed for such an analysis.


A “small organic molecule” refers to an organic molecule, either naturally occurring or synthetic, that has a molecular weight of more than about 50 Daltons and less than about 2500 Daltons, preferably less than about 2000 Daltons, preferably between about 100 to about 1000 Daltons, more preferably between about 200 to about 500 Daltons. An “siRNA” or “RNAi” refers to a nucleic acid that forms a double stranded RNA, which double stranded RNA has the ability to reduce or inhibit expression of a gene or target gene when the siRNA expressed in the same cell as the gene or target gene. “siRNA” or “RNAi” thus refers to the double stranded RNA formed by the complementary strands. The complementary portions of the siRNA that hybridize to form the double stranded molecule typically have substantial or complete identity. In one embodiment, an siRNA refers to a nucleic acid that has substantial or complete identity to a target gene and forms a double stranded siRNA. Typically, the siRNA is at least about 15-50 nucleotides in length (e.g., each complementary sequence of the double stranded siRNA is 15-50 nucleotides in length, and the double stranded siRNA is about 15-50 base pairs in length, preferable about preferably about 20-30 base nucleotides, preferably about 20-25 or about 24-29 nucleotides in length, e.g., 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length.


“Determining the functional effect” refers to assaying for a compound that increases or decreases a parameter that is indirectly or directly under the influence of a polynucleotide or polypeptide of the invention (such as a polynucleotide of Tables 1-6 or a polypeptide encoded by a gene of Tables 1-6), e.g., measuring physical and chemical or phenotypic effects. Such functional effects can be measured by any means known to those skilled in the art, e.g., changes in spectroscopic (e.g., fluorescence, absorbance, refractive index), hydrodynamic (e.g., shape), chromatographic, or solubility properties for the protein; measuring inducible markers or transcriptional activation of the protein; measuring binding activity or binding assays, e.g. binding to antibodies; measuring changes in ligand binding affinity; measurement of calcium influx; measurement of the accumulation of an enzymatic product of a polypeptide of the invention or depletion of an substrate; measurement of changes in protein levels of a polypeptide of the invention; measurement of RNA stability; G-protein binding; GPCR phosphorylation or dephosphorylation; signal transduction, e.g., receptor-ligand interactions, second messenger concentrations (e.g., cAMP, IP3, or intracellular Ca2+); identification of downstream or reporter gene expression (CAT, luciferase, β-gal, GFP and the like), e.g., via chemiluminescence, fluorescence, colorimetric reactions, antibody binding, inducible markers, and ligand binding assays.


Samples or assays comprising a nucleic acid or protein disclosed herein that are treated with a potential activator, inhibitor, or modulator are compared to control samples without the inhibitor, activator, or modulator to examine the extent of inhibition. Control samples (untreated with inhibitors) are assigned a relative protein activity value of 100%. Inhibition is achieved when the activity value relative to the control is about 80%, preferably 50%, more preferably 25-0%. Activation is achieved when the activity value relative to the control (untreated with activators) is 110%, more preferably 150%, more preferably 200-500% (i.e., two to five fold higher relative to the control), more preferably 1000-3000% higher.


“Biological sample” includes sections of tissues such as biopsy and autopsy samples, and frozen sections taken for histologic purposes. Such samples include blood, serum, lymphocytes, spinal fluid, sputum, tissue, lysed cells, brain biopsy, cultured cells, e.g., primary cultures, explants, and transformed cells, stool, urine, etc. A biological sample is typically obtained from a eukaryotic organism, most preferably a mammal such as a primate, e.g., chimpanzee or human; cow; dog; cat; a rodent, e.g., guinea pig, rat, mouse; rabbit; or a bird; reptile; or fish. Biological samples can be used to examine nucleic acids and proteins.


“Antibody” refers to a polypeptide substantially encoded by an immunoglobulin gene or immunoglobulin genes, or fragments thereof which specifically bind and recognize an analyte (antigen). The recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.


An exemplary immunoglobulin (antibody) structural unit comprises a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kD) and one “heavy” chain (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms variable light chain (VL) and variable heavy chain (VH) refer to these light and heavy chains respectively.


Antibodies exist, e.g., as intact immunoglobulins or as a number of well-characterized fragments produced by digestion with various peptidases. Thus, for example, pepsin digests an antibody below the disulfide linkages in the hinge region to produce F(ab)′2, a dimer of Fab which itself is a light chain joined to VH-CH1 by a disulfide bond. The F(ab)′2 may be reduced under mild conditions to break the disulfide linkage in the hinge region, thereby converting the F(ab)′2 dimer into an Fab′ monomer. The Fab′ monomer is essentially an Fab with part of the hinge region (see, Paul (Ed.) Fundamental Immunology, Third Edition, Raven Press, NY (1993)). While various antibody fragments are defined in terms of the digestion of an intact antibody, one of skill will appreciate that such fragments may be synthesized de novo either chemically or by utilizing recombinant DNA methodology. Thus, the term antibody, as used herein, also includes antibody fragments either produced by the modification of whole antibodies or those synthesized de novo using recombinant DNA methodologies (e.g., single chain Fv).


The terms “peptidomimetic” and “mimetic” refer to a synthetic chemical compound that has substantially the same structural and functional characteristics of the polynucleotides, polypeptides, antagonists or agonists of the invention. Peptide analogs are commonly used in the pharmaceutical industry as non-peptide drugs with properties analogous to those of the template peptide. These types of non-peptide compound are termed “peptide mimetics” or “peptidomimetics” (Fauchere, Adv. Drug Res. 15:29 (1986); Veber and Freidinger TINS p. 392 (1985); and Evans et al., J. Med. Chem. 30:1229 (1987), which are incorporated herein by reference). Peptide mimetics that are structurally similar to therapeutically useful peptides may be used to produce an equivalent or enhanced therapeutic or prophylactic effect. Generally, peptidomimetics are structurally similar to a paradigm polypeptide (i.e., a polypeptide that has a biological or pharmacological activity), such as a CCX CKR, but have one or more peptide linkages optionally replaced by a linkage selected from the group consisting of, e.g., —CH2NH—, —CH2S—, —CH2—CH2—, —CH═CH— (cis and trans), —COCH2—, —CH(OH)CH2—, and —CH2SO—. The mimetic can be either entirely composed of synthetic, non-natural analogues of amino acids, or, is a chimeric molecule of partly natural peptide amino acids and partly non-natural analogs of amino acids. The mimetic can also incorporate any amount of natural amino acid conservative substitutions as long as such substitutions also do not substantially alter the mimetic's structure and/or activity. For example, a mimetic composition is within the scope of the invention if it is capable of carrying out the binding or enzymatic activities of a polypeptide or polynucleotide of the invention or inhibiting or increasing the enzymatic activity or expression of a polypeptide or polynucleotide of the invention.


The term “gene” means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).


The term “isolated,” when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is essentially free of other cellular components with which it is associated in the natural state. It is preferably in a homogeneous state although it can be in either a dry or aqueous solution. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein that is the predominant species present in a preparation is substantially purified. In particular, an isolated gene is separated from open reading frames that flank the gene and encode a protein other than the gene of interest. The term “purified” denotes that a nucleic acid or protein gives rise to essentially one band in an electrophoretic gel. Particularly, it means that the nucleic acid or protein is at least 85% pure, more preferably at least 95% pure, and most preferably at least 99% pure.


The term “nucleic acid” or “polynucleotide” refers to deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides that have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e.g., degenerate codon substitutions), alleles, orthologs, SNPs, haplotypes, and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); and Cassol et al. (1992); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)). The term nucleic acid is used interchangeably with gene, cDNA, and mRNA encoded by a gene.


The terms “polypeptide,” “peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymers. As used herein, the terms encompass amino acid chains of any length, including full-length proteins (i.e., antigens), wherein the amino acid residues are linked by covalent peptide bonds.


The term “amino acid” refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, γ-carboxyglutamate, and O-phosphoserine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. “Amino acid mimetics” refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.


Amino acids may be referred to herein by either the commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.


“Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids that encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein that encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of skill will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid that encodes a polypeptide is implicit in each described sequence.


As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.


The following eight groups each contain amino acids that are conservative substitutions for one another:


1) Alanine (A), Glycine (G);


2) Aspartic acid (D), Glutamic acid (E);


3) Asparagine (N), Glutamine (Q);


4) Arginine (R), Lysine (K);


5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);


6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);


7) Serine (S), Threonine (T); and


8) Cysteine (C), Methionine (M)


(see, e.g., Creighton, Proteins (1984)).


“Percentage of sequence identity” is determined by comparing two optimally aligned sequences over a comparison window, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) as compared to the reference sequence (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid base or amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the window of comparison and multiplying the result by 100 to yield the percentage of sequence identity.


The terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 60% identity, optionally 65%, 70%, 75%, 80%, 85%, 90%, or 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms or by manual alignment and visual inspection. Such sequences are then said to be “substantially identical.” This definition also refers to the complement of a test sequence. Optionally, the identity exists over a region that is at least about 50 nucleotides in length, or more preferably over a region that is 100 to 500 or 1000 or more nucleotides in length.


For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.


A “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are well known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l. Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Ausubel et al., Current Protocols in Molecular Biology (1995 supplement)).


An example of an algorithm that is suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nuc. Acids Res. 25:3389-3402, and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al., supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are extended in both directions along each sequence for as far as the cumulative alignment score can be increased. Cumulative scores are calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) or 10, M=5, N=−4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915) alignments (B) of 50, expectation (E) of 10, M=5, N=−4, and a comparison of both strands.


The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, more preferably less than about 0.01, and most preferably less than about 0.001.


An indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the antibodies raised against the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent conditions, as described below. Yet another indication that two nucleic acid sequences are substantially identical is that the same primers can be used to amplify the sequence.


The phrase “selectively (or specifically) hybridizes to” refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (e.g., total cellular or library DNA or RNA).


The phrase “stringent hybridization conditions” refers to conditions under which a probe will hybridize to its target subsequence, typically in a complex mixture of nucleic acid, but to no other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Probes, “Overview of principles of hybridization and the strategy of nucleic acid assays” (1993). Generally, stringent conditions are selected to be about 5-10° C. lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength pH. The Tm is the temperature (under defined ionic strength, pH, and nucleic concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at Tm, 50% of the probes are occupied at equilibrium). Stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 30° C. for short probes (e.g., 10 to 50 nucleotides) and at least about 60° C. for long probes (e.g., greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. For selective or specific hybridization, a positive signal is at least two times background, optionally 10 times background hybridization. Exemplary stringent hybridization conditions can be as following: 50% formamide, 5×SSC, and 1% SDS, incubating at 42° C., or 5×SSC, 1% SDS, incubating at 65° C., with wash in 0.2×SSC, and 0.1% SDS at 65° C. Such washes can be performed for 5, 15, 30, 60, 120, or more minutes. Nucleic acids that hybridize to the genes listed in Tables 1-6 are encompassed by the invention.


Nucleic acids that do not hybridize to each other under stringent conditions are still substantially identical if the polypeptides that they encode are substantially identical. This occurs, for example, when a copy of a nucleic acid is created using the maximum codon degeneracy permitted by the genetic code. In such cases, the nucleic acids typically hybridize under moderately stringent hybridization conditions. Exemplary “moderately stringent hybridization conditions” include a hybridization in a buffer of 40% formamide, 1 M NaCl, 1% SDS at 37° C., and a wash in 1×SSC at 45° C. Such washes can be performed for 5, 15, 30, 60, 120, or more minutes. A positive hybridization is at least twice background. Those of ordinary skill will readily recognize that alternative hybridization and wash conditions can be utilized to provide conditions of similar stringency.


For PCR, a temperature of about 36° C. is typical for low stringency amplification, although annealing temperatures may vary between about 32° C. and 48° C. depending on primer length. For high stringency PCR amplification, a temperature of about 62° C. is typical, although high stringency annealing temperatures can range from about 50° C. to about 65° C., depending on the primer length and specificity. Typical cycle conditions for both high and low stringency amplifications include a denaturation phase of 90° C.-95° C. for 30 sec-2 min., an annealing phase lasting 30 sec.-2 min., and an extension phase of about 72° C. for 1-2 min. Protocols and guidelines for low and high stringency amplification reactions are provided, e.g., in Innis et al., PCR Protocols, A Guide to Methods and Applications (1990).


The phrase “a nucleic acid sequence encoding” refers to a nucleic acid that contains sequence information for a structural RNA such as rRNA, a tRNA, or the primary amino acid sequence of a specific protein or peptide, or a binding site for a trans-acting regulatory agent. This phrase specifically encompasses degenerate codons (i.e., different codons which encode a single amino acid) of the native sequence or sequences which may be introduced to conform with codon preference in a specific host cell.


The term “recombinant” when used with reference, e.g., to a cell, or nucleic acid, protein, or vector, indicates that the cell, nucleic acid, protein or vector, has been modified by the introduction of a heterologous nucleic acid or protein or the alteration of a native nucleic acid or protein, or that the cell is derived from a cell so modified. Thus, for example, recombinant cells express genes that are not found within the native (nonrecombinant) form of the cell or express native genes that are otherwise abnormally expressed, under-expressed or not expressed at all.


The term “heterologous” when used with reference to portions of a nucleic acid indicates that the nucleic acid comprises two or more subsequences that are not found in the same relationship to each other in nature. For instance, the nucleic acid is typically recombinantly produced, having two or more sequences from unrelated genes arranged to make a new functional nucleic acid, e.g., a promoter from one source and a coding region from another source. Similarly, a heterologous protein indicates that the protein comprises two or more subsequences that are not found in the same relationship to each other in nature (e.g., a fusion protein).


An “expression vector” is a nucleic acid construct, generated recombinantly or synthetically, with a series of specified nucleic acid elements that permit transcription of a particular nucleic acid in a host cell. The expression vector can be part of a plasmid, virus, or nucleic acid fragment. Typically, the expression vector includes a nucleic acid to be transcribed operably linked to a promoter.


The phrase “specifically (or selectively) binds to an antibody” or “specifically (or selectively) immunoreactive with”, when referring to a protein or peptide, refers to a binding reaction which is determinative of the presence of the protein in the presence of a heterogeneous population of proteins and other biologics. Thus, under designated immunoassay conditions, the specified antibodies bind to a particular protein and do not bind in a significant amount to other proteins present in the sample. Specific binding to an antibody under such conditions may require an antibody that is selected for its specificity for a particular protein. For example, antibodies raised against a protein having an amino acid sequence encoded by any of the polynucleotides of the invention can be selected to obtain antibodies specifically immunoreactive with that protein and not with other proteins, except for polymorphic variants. A variety of immunoassay formats may be used to select antibodies specifically immunoreactive with a particular protein. For example, solid-phase ELISA immunoassays, Western blots, or immunohistochemistry are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See, Harlow and Lane Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, NY (1988) for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity. Typically, a specific or selective reaction will be at least twice the background signal or noise and more typically more than 10 to 100 times background.


One who is “predisposed for a mental disorder” as used herein means a person who has an inclination or a higher likelihood of developing a mental disorder when compared to an average person in the general population.







DETAILED DESCRIPTION OF THE INVENTION

I. Introduction


To understand the complex genetic basis of mental disorders, the present invention provides studies that have been conducted to investigate the expression patterns of genes that are differentially expressed specifically in central nervous system of subjects with psychotic and mood disorders. The large spectrum of symptoms associated with mental disorders is a reflection of the complex genetic basis and complex gene expression patterns in patients with mental disorders. Different combinations of the genes disclosed herein can be responsible for one or more mental disorders. Furthermore, brain pathways or circuits as well as subcellular pathways are important for understanding the development and diagnosis of mental disorders. The selected brain regions described herein (anterior cingulate cortex (AnCg), dorsolateral prefrontal cortex (DLPFC), cerebellar cortex (CB), entorhinal cortex (ERC), superior temporal gyrus (STG), parietal cortex (PC), nucleus accumbens (nAcc), ventral thalamus (VThal), medial thalamus (MThal), amygdala (AMY) and/or the hippocampus (HC)) are implicated in the clinical symptoms of mental disorders such as psychotic and mood disorders. Brain imaging studies focusing on particular brain regions, cytoarchitectural changes in brain regions, expression of key neurotransmittors or related molecules in brain regions, and subcellular pathways in brain regions all contribute to the development of mental disorders, and thus are an important consideration in the diagnosis and therapeutic uses described herein.


The present invention demonstrates the altered expression (either higher or lower) of the genes of Tables 1-14 at the mRNA or protein level in various regions of the brain (e.g., Tables 1-4 and 6) or lymphocytes (e.g., Tables 4-6) of patients with mental disorders (e.g., schizophrenia, MDD and BPD) in comparison with normal individuals. This invention thus provides methods for diagnosis of mental disorders, e.g., schizophrenia, MDD and BPD, and the like, and other mental disorders by detecting the level of a transcript or translation product of the genes listed in Tables 1-14 as well as their corresponding biochemical pathways. The chromosomal location of such genes can be used to discover other genes in the region that are linked to development of a particular disorder. Table 6 of the invention also provides single nucleotide polymorphic markers which are related (in cis or trans) to regulatory sites associated with AGA.


The invention further provides methods of identifying a compound useful for the treatment of such disorders by selecting compounds that modulates the functional effect of the translation products or the expression of the transcripts described herein. The invention also provides for methods of treating patients with such mental disorders, e.g., by administering the compounds of the invention or by gene therapy. Therapeutic compounds include antibodies, peptides, antisense molecules, siRNA, and small organic molecules.


The genes and the polypeptides that they encode, which are associated with psychotic and mood disorders, are useful for facilitating the design and development of various molecular diagnostic tools such as GeneChips™ containing probe sets specific for all or selected mental disorders, including but not limited to psychotic and mood disorders, and as an ante-and/or post-natal diagnostic tool for screening newborns in concert with genetic counseling. Other diagnostic applications include evaluation of disease susceptibility, prognosis, and monitoring of disease or treatment process, as well as providing individualized medicine via predictive drug profiling systems, e.g., by correlating specific genomic motifs with the clinical response of a patient to individual drugs. In addition, the present invention is useful for multiplex SNP or haplotype profiling, including but not limited to the identification of pharmacogenetic targets at the gene, mRNA, protein, and pathway level (see, e.g, Basile V S, Masellis M, Potkin S G, Kennedy J L. Pharmacogenomics in schizophrenia: the quest for individualized therapy. Hum Mol Genet. 2002 Oct. 1; 11(20):2517-30). Profiling of splice variants is also useful for diagnostic and therapeutic applications. Diagnostic kits are contemplated by the present invention, and include arrays, nanoparticles, and magnetic beads. Marker combinations can provide useful diagnosis. Brain expression patterns, regions, pathways, and circuits can be used for in vivo imaging and diagnosis.


The genes and the polypeptides that they encode, described herein, as also useful as drug targets for the development of therapeutic drugs for the treatment or prevention of mental disorders including, but not limited to, psychotic and mood disorders. Mental disorders have a high co-morbidity with other neurological disorders, such as Parkinson's disease or Alzheimer's. Therefore, the present invention can be used for diagnosis and treatment of patients with multiple disease states that include a mental disorder such as a psychotic disorder.


Antipsychotic medicines are in general equally effect for the treatment of schizophrenia, but act by different mechanisms. The similar effectiveness of the drugs for treatment of schizophrenia suggests that they act through a yet as unidentified common pathway. As demonstrated by the results shown herein, these drugs regulate a common gene, and/or a common group of genes as well as a unique set of genes.


The genes listed herein can be used to provide a differential diagnosis or prognosis of mood and psychotic disorders. In some cases, differentially expressed genes can be used to predict and treat particular symptoms or outcomes, such as suicide attempt. The therapeutic agents described herein can be used in combination with known therapeutics. Nucleic acid therapeutics can be delivered using adenoviral vectors, while peptides, nucleic acids, and other therapeutic molecules can be delivered using nanoparticles and translocation peptides. Orally available peptides can be made using D-amino acids or pegylation, and serum half life can be extended using albumin conjugation and the like.


II. General Recombinant Nucleic Acid Methods for Use with the Invention


In numerous embodiments of the present invention, polynucleotides of the invention will be isolated and cloned using recombinant methods. Such polynucleotides include, e.g., those listed in Tables 1-14, which can be used for, e.g., protein expression or during the generation of variants, derivatives, expression cassettes, to monitor gene expression, for the isolation or detection of sequences of the invention in different species, for diagnostic purposes in a patient, e.g., to detect mutations or to detect expression levels of nucleic acids or polypeptides of the invention. In some embodiments, the sequences of the invention are operably linked to a heterologous promoter. In one embodiment, the nucleic acids of the invention are from any mammal, including, in particular, e.g., a human, a mouse, a rat, a primate, etc.


A. General Recombinant Nucleic Acids Methods


This invention relies on routine techniques in the field of recombinant genetics. Basic texts disclosing the general methods of use in this invention include Sambrook et al., Molecular Cloning, A Laboratory Manual (3rd ed. 2001); Kriegler, Gene Transfer and Expression: A Laboratory Manual (1990); and Current Protocols in Molecular Biology (Ausubel et al., eds., 1994)).


For nucleic acids, sizes are given in either kilobases (kb) or base pairs (bp). These are estimates derived from agarose or acrylamide gel electrophoresis, from sequenced nucleic acids, or from published DNA sequences. For proteins, sizes are given in kilodaltons (kDa) or amino acid residue numbers. Proteins sizes are estimated from gel electrophoresis, from sequenced proteins, from derived amino acid sequences, or from published protein sequences.


Oligonucleotides that are not commercially available can be chemically synthesized according to the solid phase phosphoramidite triester method first described by Beaucage & Caruthers, Tetrahedron Letts. 22:1859-1862 (1981), using an automated synthesizer, as described in Van Devanter et. al., Nucleic Acids Res. 12:6159-6168 (1984). Purification of oligonucleotides is by either native acrylamide gel electrophoresis or by anion-exchange HPLC as described in Pearson & Reanier, J. Chrom. 255:137-149 (1983).


The sequence of the cloned genes and synthetic oligonucleotides can be verified after cloning using, e.g., the chain termination method for sequencing double-stranded templates of Wallace et al., Gene 16:21-26 (1981).


B. Cloning Methods for the Isolation of Nucleotide Sequences Encoding Desired Proteins


In general, the nucleic acids encoding the subject proteins are cloned from DNA sequence libraries that are made to encode cDNA or genomic DNA. The particular sequences can be located by hybridizing with an oligonucleotide probe, the sequence of which can be derived from the sequences of the genes and/or SNPs listed in Tables 1-6, which provide a reference for PCR primers and defines suitable regions for isolating specific probes. Alternatively, where the sequence is cloned into an expression library, the expressed recombinant protein can be detected immunologically with antisera or purified antibodies made against a polypeptide comprising an amino acid sequence encoded by a gene listed in Tables 1-14.


Methods for making and screening genomic and cDNA libraries are well known to those of skill in the art (see, e.g., Gubler and Hoffman Gene 25:263-269 (1983); Benton and Davis Science, 196:180-182 (1977); and Sambrook, supra). Brain cells are an example of suitable cells to isolate RNA and cDNA sequences of the invention.


Briefly, to make the cDNA library, one should choose a source that is rich in mRNA. The mRNA can then be made into cDNA, ligated into a recombinant vector, and transfected into a recombinant host for propagation, screening and cloning. For a genomic library, the DNA is extracted from a suitable tissue and either mechanically sheared or enzymatically digested to yield fragments of preferably about 5-100 kb. The fragments are then separated by gradient centrifugation from undesired sizes and are constructed in bacteriophage lambda vectors. These vectors and phage are packaged in vitro, and the recombinant phages are analyzed by plaque hybridization. Colony hybridization is carried out as generally described in Grunstein et al., Proc. Natl. Acad. Sci. USA., 72:3961-3965 (1975).


An alternative method combines the use of synthetic oligonucleotide primers with polymerase extension on an mRNA or DNA template. Suitable primers can be designed from specific sequences of the invention. This polymerase chain reaction (PCR) method amplifies the nucleic acids encoding the protein of interest directly from mRNA, cDNA, genomic libraries or cDNA libraries. Restriction endonuclease sites can be incorporated into the primers. Polymerase chain reaction or other in vitro amplification methods may also be useful, for example, to clone nucleic acids encoding specific proteins and express said proteins, to synthesize nucleic acids that will be used as probes for detecting the presence of mRNA encoding a polypeptide of the invention in physiological samples, for nucleic acid sequencing, or for other purposes (see, U.S. Pat. Nos. 4,683,195 and 4,683,202). Genes amplified by a PCR reaction can be purified from agarose gels and cloned into an appropriate vector.


Appropriate primers and probes for identifying polynucleotides of the invention from mammalian tissues can be derived from the sequences provided herein. For a general overview of PCR, see, Innis et al. PCR Protocols: A Guide to Methods and Applications, Academic Press, San Diego (1990).


Synthetic oligonucleotides can be used to construct genes. This is done using a series of overlapping oligonucleotides, usually 40-120 bp in length, representing both the sense and anti-sense strands of the gene. These DNA fragments are then annealed, ligated and cloned.


A gene encoding a polypeptide of the invention can be cloned using intermediate vectors before transformation into mammalian cells for expression. These intermediate vectors are typically prokaryote vectors or shuttle vectors. The proteins can be expressed in either prokaryotes, using standard methods well known to those of skill in the art, or eukaryotes as described infra.


III. Purification of Proteins of the Invention


Either naturally occurring or recombinant polypeptides of the invention can be purified for use in functional assays. Naturally occurring polypeptides, e.g., polypeptides encoded by genes listed in Tables 1-14, can be purified, for example, from mouse or human tissue such as brain or any other source of an ortholog. Recombinant polypeptides can be purified from any suitable expression system.


The polypeptides of the invention may be purified to substantial purity by standard techniques, including selective precipitation with such substances as ammonium sulfate; column chromatography, immunopurification methods, and others (see, e.g., Scopes, Protein Purification: Principles and Practice (1982); U.S. Pat. No. 4,673,641; Ausubel et al., supra; and Sambrook et al., supra).


A number of procedures can be employed when recombinant polypeptides are purified. For example, proteins having established molecular adhesion properties can be reversible fused to polypeptides of the invention. With the appropriate ligand, the polypeptides can be selectively adsorbed to a purification column and then freed from the column in a relatively pure form. The fused protein is then removed by enzymatic activity. Finally the polypeptide can be purified using immunoaffinity columns.


A. Purification of Proteins from Recombinant Bacteria


When recombinant proteins are expressed by the transformed bacteria in large amounts, typically after promoter induction, although expression can be constitutive, the proteins may form insoluble aggregates. There are several protocols that are suitable for purification of protein inclusion bodies. For example, purification of aggregate proteins (hereinafter referred to as inclusion bodies) typically involves the extraction, separation and/or purification of inclusion bodies by disruption of bacterial cells typically, but not limited to, by incubation in a buffer of about 100-150 μg/ml lysozyme and 0.1% Nonidet P40, a non-ionic detergent. The cell suspension can be ground using a Polytron grinder (Brinkman Instruments, Westbury, N.Y.). Alternatively, the cells can be sonicated on ice. Alternate methods of lysing bacteria are described in Ausubel et al. and Sambrook et al., both supra, and will be apparent to those of skill in the art.


The cell suspension is generally centrifuged and the pellet containing the inclusion bodies resuspended in buffer which does not dissolve but washes the inclusion bodies, e.g., 20 mM Tris-HCl (pH 7.2), 1 mM EDTA, 150 mM NaCl and 2% Triton-X 100, a non-ionic detergent. It may be necessary to repeat the wash step to remove as much cellular debris as possible. The remaining pellet of inclusion bodies may be resuspended in an appropriate buffer (e.g., 20 mM sodium phosphate, pH 6.8, 150 mM NaCl). Other appropriate buffers will be apparent to those of skill in the art.


Following the washing step, the inclusion bodies are solubilized by the addition of a solvent that is both a strong hydrogen acceptor and a strong hydrogen donor (or a combination of solvents each having one of these properties). The proteins that formed the inclusion bodies may then be renatured by dilution or dialysis with a compatible buffer. Suitable solvents include, but are not limited to, urea (from about 4 M to about 8 M), formamide (at least about 80%, volume/volume basis), and guanidine hydrochloride (from about 4 M to about 8 M). Some solvents that are capable of solubilizing aggregate-forming proteins, such as SDS (sodium dodecyl sulfate) and 70% formic acid, are inappropriate for use in this procedure due to the possibility of irreversible denaturation of the proteins, accompanied by a lack of immunogenicity and/or activity. Although guanidine hydrochloride and similar agents are denaturants, this denaturation is not irreversible and renaturation may occur upon removal (by dialysis, for example) or dilution of the denaturant, allowing re-formation of the immunologically and/or biologically active protein of interest. After solubilization, the protein can be separated from other bacterial proteins by standard separation techniques.


Alternatively, it is possible to purify proteins from bacteria periplasm. Where the protein is exported into the periplasm of the bacteria, the periplasmic fraction of the bacteria can be isolated by cold osmotic shock in addition to other methods known to those of skill in the art (see, Ausubel et al., supra). To isolate recombinant proteins from the periplasm, the bacterial cells are centrifuiged to form a pellet. The pellet is resuspended in a buffer containing 20% sucrose. To lyse the cells, the bacteria are centrifuged and the pellet is resuspended in ice-cold 5 mM MgSO4 and kept in an ice bath for approximately 10 minutes. The cell suspension is centrifuged and the supernatant decanted and saved. The recombinant proteins present in the supernatant can be separated from the host proteins by standard separation techniques well known to those of skill in the art.


B. Standard Protein Separation Techniques for Purifying Proteins


1. Solubility Fractionation


Often as an initial step, and if the protein mixture is complex, an initial salt fractionation can separate many of the unwanted host cell proteins (or proteins derived from the cell culture media) from the recombinant protein of interest. The preferred salt is ammonium sulfate. Ammonium sulfate precipitates proteins by effectively reducing the amount of water in the protein mixture. Proteins then precipitate on the basis of their solubility. The more hydrophobic a protein is, the more likely it is to precipitate at lower ammonium sulfate concentrations. A typical protocol is to add saturated ammonium sulfate to a protein solution so that the resultant ammonium sulfate concentration is between 20-30%. This will precipitate the most hydrophobic proteins. The precipitate is discarded (unless the protein of interest is hydrophobic) and ammonium sulfate is added to the supernatant to a concentration known to precipitate the protein of interest. The precipitate is then solubilized in buffer and the excess salt removed if necessary, through either dialysis or diafiltration. Other methods that rely on solubility of proteins, such as cold ethanol precipitation, are well known to those of skill in the art and can be used to fractionate complex protein mixtures.


2. Size Differential Filtration


Based on a calculated molecular weight, a protein of greater and lesser size can be isolated using ultrafiltration through membranes of different pore sizes (for example, Amicon or Millipore membranes). As a first step, the protein mixture is ultrafiltered through a membrane with a pore size that has a lower molecular weight cut-off than the molecular weight of the protein of interest. The retentate of the ultrafiltration is then ultrafiltered against a membrane with a molecular cut off greater than the molecular weight of the protein of interest. The recombinant protein will pass through the membrane into the filtrate. The filtrate can then be chromatographed as described below.


3. Column Chromatography


The proteins of interest can also be separated from other proteins on the basis of their size, net surface charge, hydrophobicity and affinity for ligands. In addition, antibodies raised against proteins can be conjugated to column matrices and the proteins immunopurified. All of these methods are well known in the art.


It will be apparent to one of skill that chromatographic techniques can be performed at any scale and using equipment from many different manufacturers (e.g., Pharmacia Biotech).


IV. Detection of Gene Expression


Those of skill in the art will recognize that detection of expression of polynucleotides of the invention has many uses. For example, as discussed herein, detection of the level of polypeptides or polynucleotides of the invention in a patient is useful for diagnosing mood disorders or psychotic disorder or a predisposition for a mood disorder or psychotic disorder. Moreover, detection of gene expression is useful to identify modulators of expression of the polypeptides or polynucleotides of the invention.


A variety of methods of specific DNA and RNA measurement using nucleic acid hybridization techniques are known to those of skill in the art (see, Sambrook, supra). Some methods involve an electrophoretic separation (e.g., Southern blot for detecting DNA, and Northern blot for detecting RNA), but measurement of DNA and RNA can also be carried out in the absence of electrophoretic separation (e.g., by dot blot). Southern blot of genomic DNA (e.g., from a human) can be used for screening for restriction fragment length polymorphism (RFLP) to detect the presence of a genetic disorder affecting a polypeptide of the invention.


The selection of a nucleic acid hybridization format is not critical. A variety of nucleic acid hybridization formats are known to those skilled in the art. For example, common formats include sandwich assays and competition or displacement assays. Hybridization techniques are generally described in Hames and Higgins Nucleic Acid Hybridization, A Practical Approach, IRL Press (1985); Gall and Pardue, Proc. Natl. Acad. Sci. U.S.A., 63:378-383 (1969); and John et al. Nature, 223:582-587 (1969).


Detection of a hybridization complex may require the binding of a signal-generating complex to a duplex of target and probe polynucleotides or nucleic acids. Typically, such binding occurs through ligand and anti-ligand interactions as between a ligand-conjugated probe and an anti-ligand conjugated with a signal. The binding of the signal generation complex is also readily amenable to accelerations by exposure to ultrasonic energy.


The label may also allow indirect detection of the hybridization complex. For example, where the label is a hapten or antigen, the sample can be detected by using antibodies. In these systems, a signal is generated by attaching fluorescent or enzyme molecules to the antibodies or in some cases, by attachment to a radioactive label (see, e.g., Tijssen, “Practice and Theory of Enzyme Immunoassays,” Laboratory Techniques in Biochemistry and Molecular Biology, Burdon and van Knippenberg Eds., Elsevier (1985), pp. 9-20).


The probes are typically labeled either directly, as with isotopes, chromophores, lumiphores, chromogens, or indirectly, such as with biotin, to which a streptavidin complex may later bind. Thus, the detectable labels used in the assays of the present invention can be primary labels (where the label comprises an element that is detected directly or that produces a directly detectable element) or secondary labels (where the detected label binds to a primary label, e.g., as is common in immunological labeling). Typically, labeled signal nucleic acids are used to detect hybridization. Complementary nucleic acids or signal nucleic acids may be labeled by any one of several methods typically used to detect the presence of hybridized polynucleotides. The most common method of detection is the use of autoradiography with 3H, 125I, 35S, 14C, or 32P-labeled probes or the like.


Other labels include, e.g., ligands that bind to labeled antibodies, fluorophores, chemiluminescent agents, enzymes, and antibodies which can serve as specific binding pair members for a labeled ligand. An introduction to labels, labeling procedures and detection of labels is found in Polak and Van Noorden Introduction to Immunocytochemistry, 2nd ed., Springer Verlag, NY (1997); and in Haugland Handbook of Fluorescent Probes and Research Chemicals, a combined handbook and catalogue Published by Molecular Probes, Inc. (1996).


In general, a detector which monitors a particular probe or probe combination is used to detect the detection reagent label. Typical detectors include spectrophotometers, phototubes and photodiodes, microscopes, scintillation counters, cameras, film and the like, as well as combinations thereof. Examples of suitable detectors are widely available from a variety of commercial sources known to persons of skill in the art. Commonly, an optical image of a substrate comprising bound labeling moieties is digitized for subsequent computer analysis.


Most typically, the amount of RNA is measured by quantifying the amount of label fixed to the solid support by binding of the detection reagent. Typically, the presence of a modulator during incubation will increase or decrease the amount of label fixed to the solid support relative to a control incubation which does not comprise the modulator, or as compared to a baseline established for a particular reaction type. Means of detecting and quantifying labels are well known to those of skill in the art.


In preferred embodiments, the target nucleic acid or the probe is immobilized on a solid support. Solid supports suitable for use in the assays of the invention are known to those of skill in the art. As used herein, a solid support is a matrix of material in a substantially fixed arrangement.


A variety of automated solid-phase assay techniques are also appropriate. For instance, very large scale immobilized polymer arrays (VLSIPS™), available from Affymetrix, Inc. (Santa Clara, Calif.) can be used to detect changes in expression levels of a plurality of genes involved in the same regulatory pathways simultaneously. See, Tijssen, supra., Fodor et al. (1991) Science, 251: 767-777; Sheldon et al. (1993) Clinical Chemistry 39(4): 718-719, and Kozal et al. (1996) Nature Medicine 2(7): 753-759.


Detection can be accomplished, for example, by using a labeled detection moiety that binds specifically to duplex nucleic acids (e.g., an antibody that is specific for RNA-DNA duplexes). One preferred example uses an antibody that recognizes DNA-RNA heteroduplexes in which the antibody is linked to an enzyme (typically by recombinant or covalent chemical bonding). The antibody is detected when the enzyme reacts with its substrate, producing a detectable product. Coutlee et al. (1989) Analytical Biochemistry 181:153-162; Bogulavski (1986) et al. J. Immunol. Methods 89:123-130; Prooijen-Knegt (1982) Exp. Cell Res. 141:397-407; Rudkin (1976) Nature 265:472-473, Stollar (1970) Proc. Nat'l Acad. Sci. USA 65:993-1000; Ballard (1982) Mol. Immunol. 19:793-799; Pisetsky and Caster (1982) Mol. Immunol. 19:645-650; Viscidi et al. (1988) J. Clin. Microbial. 41:199-209; and Kiney et al. (1989) J. Clin. Microbiol. 27:6-12 describe antibodies to RNA duplexes, including homo and heteroduplexes. Kits comprising antibodies specific for DNA:RNA hybrids are available, e.g., from Digene Diagnostics, Inc. (Beltsville, Md.).


In addition to available antibodies, one of skill in the art can easily make antibodies specific for nucleic acid duplexes using existing techniques, or modify those antibodies that are commercially or publicly available. In addition to the art referenced above, general methods for producing polyclonal and monoclonal antibodies are known to those of skill in the art (see, e.g., Paul (3rd ed.) Fundamental Immunology Raven Press, Ltd., NY (1993); Coligan Current Protocols in Immunology Wiley/Greene, NY (1991); Harlow and Lane Antibodies: A Laboratory Manual Cold Spring Harbor Press, NY (1988); Stites et al. (eds.) Basic and Clinical Immunology (4th ed.) Lange Medical Publications, Los Altos, Calif., and references cited therein; Goding Monoclonal Antibodies: Principles and Practice (2d ed.) Academic Press, New York, N.Y., (1986); and Kohler and Milstein Nature 256: 495-497 (1975)). Other suitable techniques for antibody preparation include selection of libraries of recombinant antibodies in phage or similar vectors (see, Huse et al. Science 246:1275-1281 (1989); and Ward et al. Nature 341:544-546 (1989)). Specific monoclonal and polyclonal antibodies and antisera will usually bind with a KD of at least about 0.1 μM, preferably at least about 0.01 μM or better, and most typically and preferably, 0.001 μM or better.


The nucleic acids used in this invention can be either positive or negative probes. Positive probes bind to their targets and the presence of duplex formation is evidence of the presence of the target. Negative probes fail to bind to the suspect target and the absence of duplex formation is evidence of the presence of the target. For example, the use of a wild type specific nucleic acid probe or PCR primers may serve as a negative probe in an assay sample where only the nucleotide sequence of interest is present.


The sensitivity of the hybridization assays may be enhanced through use of a nucleic acid amplification system that multiplies the target nucleic acid being detected. Examples of such systems include the polymerase chain reaction (PCR) system, in particular RT-PCR or real time PCR, and the ligase chain reaction (LCR) system. Other methods recently described in the art are the nucleic acid sequence based amplification (NASBA, Cangene, Mississauga, Ontario) and Q Beta Replicase systems. These systems can be used to directly identify mutants where the PCR or LCR primers are designed to be extended or ligated only when a selected sequence is present. Alternatively, the selected sequences can be generally amplified using, for example, nonspecific PCR primers and the amplified target region later probed for a specific sequence indicative of a mutation.


An alternative means for determining the level of expression of the nucleic acids of the present invention is in situ hybridization. In situ hybridization assays are well known and are generally described in Angerer et al., Methods Enzymol. 152:649-660 (1987). In an in situ hybridization assay, cells or tissue, preferentially human cells or tissue from a selected brain region, are fixed to a solid support, typically a glass slide. If DNA is to be probed, the cells are denatured with heat or alkali. The cells are then contacted with a hybridization solution at a moderate temperature to permit annealing of specific probes that are labeled. The probes are preferably labeled with radioisotopes or fluorescent reporters.


V. Immunological Detection of the Polypeptides of the Invention


In addition to the detection of polynucleotide expression using nucleic acid hybridization technology, one can also use immunoassays to detect polypeptides of the invention. Immunoassays can be used to qualitatively or quantitatively analyze polypeptides. A general overview of the applicable technology can be found in Harlow & Lane, Antibodies: A Laboratory Manual (1988).


A. Antibodies to Target Polypeptides or Other Immunogens


Methods for producing therapeutic and diagnostic polyclonal and monoclonal antibodies that react specifically with a protein of interest or other immunogen are known to those of skill in the art (see, e.g., Coligan, supra; and Harlow and Lane, supra; Stites et al., supra and references cited therein; Goding, supra; and Kohler and Milstein Nature, 256:495-497 (1975)). Such techniques include antibody preparation by selection of antibodies from libraries of recombinant antibodies in phage or similar vectors (see, Huse et al., supra; and Ward et al., supra). For example, in order to produce antisera for use in an immunoassay, the protein of interest or an antigenic fragment thereof, is isolated as described herein. For example, a recombinant protein is produced in a transformed cell line. An inbred strain of mice or rabbits is immunized with the protein using a standard adjuvant, such as Freund's adjuvant, and a standard immunization protocol. Alternatively, a synthetic peptide derived from the sequences disclosed herein and conjugated to a carrier protein can be used as an immunogen.


Polyclonal sera are collected and titered against the immunogen in an immunoassay, for example, a solid phase immunoassay with the immunogen immobilized on a solid support. Polyclonal antisera with a titer of 104 or greater are selected and tested for their cross-reactivity against unrelated proteins or even other homologous proteins from other organisms, using a competitive binding immunoassay. Specific monoclonal and polyclonal antibodies and antisera will usually bind with a KD of at least about 0.1 mM, more usually at least about 1 μM, preferably at least about 0.1 μM or better, and most preferably, 0.01 μM or better.


A number of proteins of the invention comprising immunogens may be used to produce antibodies specifically or selectively reactive with the proteins of interest. Recombinant protein is the preferred immunogen for the production of monoclonal or polyclonal antibodies. Naturally occurring proteins, such as one comprising an amino acid sequence encoded by a gene listed in Tables 1-14, may also be used either in pure or impure form. Synthetic peptides made using the protein sequences described herein may also be used as an immunogen for the production of antibodies to the protein. Recombinant protein can be expressed in eukaryotic or prokaryotic cells and purified as generally described supra. The product is then injected into an animal capable of producing antibodies. Either monoclonal or polyclonal antibodies may be generated for subsequent use in immunoassays to measure the protein.


Methods of production of polyclonal antibodies are known to those of skill in the art. In brief, an immunogen, preferably a purified protein, is mixed with an adjuvant and animals are immunized. The animal's immune response to the immunogen preparation is monitored by taking test bleeds and determining the titer of reactivity to the polypeptide of interest. When appropriately high titers of antibody to the immunogen are obtained, blood is collected from the animal and antisera are prepared. Further fractionation of the antisera to enrich for antibodies reactive to the protein can be done if desired (see, Harlow and Lane, supra).


Monoclonal antibodies may be obtained using various techniques familiar to those of skill in the art. Typically, spleen cells from an animal immunized with a desired antigen are immortalized, commonly by fusion with a myeloma cell (see, Kohler and Milstein, Eur. J. Immunol. 6:511-519 (1976)). Alternative methods of immortalization include, e.g., transformation with Epstein Barr Virus, oncogenes, or retroviruses, or other methods well known in the art. Colonies arising from single immortalized cells are screened for production of antibodies of the desired specificity and affinity for the antigen, and yield of the monoclonal antibodies produced by such cells may be enhanced by various techniques, including injection into the peritoneal cavity of a vertebrate host. Alternatively, one may isolate DNA sequences which encode a monoclonal antibody or a binding fragment thereof by screening a DNA library from human B cells according to the general protocol outlined by Huse et al., supra.


Once target protein specific antibodies are available, the protein can be measured by a variety of immunoassay methods with qualitative and quantitative results available to the clinician. For a review of immunological and immunoassay procedures in general see, Stites, supra. Moreover, the immunoassays of the present invention can be performed in any of several configurations, which are reviewed extensively in Maggio Enzyme Immunoassay, CRC Press, Boca Raton, Fla. (1980); Tijssen, supra; and Harlow and Lane, supra.


Immunoassays to measure target proteins in a human sample may use a polyclonal antiserum that was raised to the protein (e.g., one has an amino acid sequence encoded by a gene listed in Table 1-14) or a fragment thereof. This antiserum is selected to have low cross-reactivity against different proteins and any such cross-reactivity is removed by immunoabsorption prior to use in the immunoassay.


B. Immunological Binding Assays


In a preferred embodiment, a protein of interest is detected and/or quantified using any of a number of well-known immunological binding assays (see, e.g., U.S. Pat. Nos. 4,366,241; 4,376,110; 4,517,288; and 4,837,168). For a review of the general immunoassays, see also Asai Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. NY (1993); Stites, supra. Immunological binding assays (or immunoassays) typically utilize a “capture agent” to specifically bind to and often immobilize the analyte (in this case a polypeptide of the present invention or antigenic subsequences thereof). The capture agent is a moiety that specifically binds to the analyte. In a preferred embodiment, the capture agent is an antibody that specifically binds, for example, a polypeptide of the invention. The antibody may be produced by any of a number of means well known to those of skill in the art and as described above.


Immunoassays also often utilize a labeling agent to specifically bind to and label the binding complex formed by the capture agent and the analyte. The labeling agent may itself be one of the moieties comprising the antibody/analyte complex. Alternatively, the labeling agent may be a third moiety, such as another antibody, that specifically binds to the antibody/protein complex.


In a preferred embodiment, the labeling agent is a second antibody bearing a label. Alternatively, the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. The second antibody can be modified with a detectable moiety, such as biotin, to which a third labeled molecule can specifically bind, such as enzyme-labeled streptavidin.


Other proteins capable of specifically binding immunoglobulin constant regions, such as protein A or protein G, can also be used as the label agents. These proteins are normal constituents of the cell walls of streptococcal bacteria. They exhibit a strong non-immunogenic reactivity with immunoglobulin constant regions from a variety of species (see, generally, Kronval, et al. J. Immunol., 111: 1401-1406 (1973); and Akerstrom, et al. J. Immunol., 135:2589-2542 (1985)).


Throughout the assays, incubation and/or washing steps may be required after each combination of reagents. Incubation steps can vary from about 5 seconds to several hours, preferably from about 5 minutes to about 24 hours. The incubation time will depend upon the assay format, analyte, volume of solution, concentrations, and the like. Usually, the assays will be carried out at ambient temperature, although they can be conducted over a range of temperatures, such as 10° C. to 40° C.


1. Non-Competitive Assay Formats


Immunoassays for detecting proteins of interest from tissue samples may be either competitive or noncompetitive. Noncompetitive immunoassays are assays in which the amount of captured analyte (in this case the protein) is directly measured. In one preferred “sandwich” assay, for example, the capture agent (e.g., antibodies specific for a polypeptide encoded by a gene listed in Tables 1-14) can be bound directly to a solid substrate where it is immobilized. These immobilized antibodies then capture the polypeptide present in the test sample. The polypeptide thus immobilized is then bound by a labeling agent, such as a second antibody bearing a label. Alternatively, the second antibody may lack a label, but it may, in turn, be bound by a labeled third antibody specific to antibodies of the species from which the second antibody is derived. The second can be modified with a detectable moiety, such as biotin, to which a third labeled molecule can specifically bind, such as enzyme-labeled streptavidin.


2. Competitive Assay Formats


In competitive assays, the amount of analyte (such as a polypeptide encoded by a gene listed in Table 1-14) present in the sample is measured indirectly by measuring the amount of an added (exogenous) analyte displaced (or competed away) from a capture agent (e.g., an antibody specific for the analyte) by the analyte present in the sample. In one competitive assay, a known amount of, in this case, the protein of interest is added to the sample and the sample is then contacted with a capture agent, in this case an antibody that specifically binds to a polypeptide of the invention. The amount of immunogen bound to the antibody is inversely proportional to the concentration of immunogen present in the sample. In a particularly preferred embodiment, the antibody is immobilized on a solid substrate. For example, the amount of the polypeptide bound to the antibody may be determined either by measuring the amount of subject protein present in a protein/antibody complex or, alternatively, by measuring the amount of remaining uncomplexed protein. The amount of protein may be detected by providing a labeled protein molecule.


Immunoassays in the competitive binding format can be used for cross-reactivity determinations. For example, a protein of interest can be immobilized on a solid support. Proteins are added to the assay which compete with the binding of the antisera to the immobilized antigen. The ability of the above proteins to compete with the binding of the antisera to the immobilized protein is compared to that of the protein of interest. The percent cross-reactivity for the above proteins is calculated, using standard calculations. Those antisera with less than 10% cross-reactivity with each of the proteins listed above are selected and pooled. The cross-reacting antibodies are optionally removed from the pooled antisera by immunoabsorption with the considered proteins, e.g., distantly related homologs.


The immunoabsorbed and pooled antisera are then used in a competitive binding immunoassay as described above to compare a second protein, thought to be perhaps a protein of the present invention, to the immunogen protein. In order to make this comparison, the two proteins are each assayed at a wide range of concentrations and the amount of each protein required to inhibit 50% of the binding of the antisera to the immobilized protein is determined. If the amount of the second protein required is less than 10 times the amount of the protein partially encoded by a sequence herein that is required, then the second protein is said to specifically bind to an antibody generated to an immunogen consisting of the target protein.


3. Other Assay Formats


In a particularly preferred embodiment, western blot (immunoblot) analysis is used to detect and quantify the presence of a polypeptide of the invention in the sample. The technique generally comprises separating sample proteins by gel electrophoresis on the basis of molecular weight, transferring the separated proteins to a suitable solid support (such as, e.g., a nitrocellulose filter, a nylon filter, or a derivatized nylon filter) and incubating the sample with the antibodies that specifically bind the protein of interest. For example, the antibodies specifically bind to a polypeptide of interest on the solid support. These antibodies may be directly labeled or alternatively may be subsequently detected using labeled antibodies (e.g., labeled sheep anti-mouse antibodies) that specifically bind to the antibodies against the protein of interest.


Other assay formats include liposome immunoassays (LIA), which use liposomes designed to bind specific molecules (e.g., antibodies) and release encapsulated reagents or markers. The released chemicals are then detected according to standard techniques (see, Monroe et al. (1986) Amer. Clin. Prod. Rev. 5:34-41).


4. Labels


The particular label or detectable group used in the assay is not a critical aspect of the invention, as long as it does not significantly interfere with the specific binding of the antibody used in the assay. The detectable group can be any material having a detectable physical or chemical property. Such detectable labels have been well developed in the field of immunoassays and, in general, most labels useful in such methods can be applied to the present invention. Thus, a label is any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Useful labels in the present invention include magnetic beads (e.g., Dynabeads™), fluorescent dyes (e.g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 125I, 35S, 14C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads.


The label may be coupled directly or indirectly to the desired component of the assay according to methods well known in the art. As indicated above, a wide variety of labels may be used, with the choice of label depending on the sensitivity required, the ease of conjugation with the compound, stability requirements, available instrumentation, and disposal provisions.


Non-radioactive labels are often attached by indirect means. The molecules can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorescent compound. A variety of enzymes and fluorescent compounds can be used with the methods of the present invention and are well-known to those of skill in the art (for a review of various labeling or signal producing systems which may be used, see, e.g., U.S. Pat. No. 4,391,904).


Means of detecting labels are well known to those of skill in the art. Thus, for example, where the label is a radioactive label, means for detection include a scintillation counter or photographic film as in autoradiography. Where the label is a fluorescent label, it may be detected by exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence. The fluorescence may be detected visually, by means of photographic film, by the use of electronic detectors such as charge-coupled devices (CCDs) or photomultipliers and the like. Similarly, enzymatic labels may be detected by providing the appropriate substrates for the enzyme and detecting the resulting reaction product. Finally simple colorimetric labels may be detected directly by observing the color associated with the label. Thus, in various dipstick assays, conjugated gold often appears pink, while various conjugated beads appear the color of the bead.


Some assay formats do not require the use of labeled components. For instance, agglutination assays can be used to detect the presence of the target antibodies. In this case, antigen-coated particles are agglutinated by samples comprising the target antibodies. In this format, none of the components need to be labeled and the presence of the target antibody is detected by simple visual inspection.


VI. Screening for Modulators of Polypeptides and Polynucleotides of the Invention


Modulators of polypeptides or polynucleotides of the invention, i.e. agonists or antagonists of their activity or modulators of polypeptide or polynucleotide expression, are useful for treating a number of human diseases, including mood disorders or psychotic disorders. Administration of agonists, antagonists or other agents that modulate expression of the polynucleotides or polypeptides of the invention can be used to treat patients with mood disorders or psychotic disorders.


A. Screening Methods


A number of different screening protocols can be utilized to identify agents that modulate the level of expression or activity of polypeptides and polynucleotides of the invention in cells, particularly mammalian cells, and especially human cells. In general terms, the screening methods involve screening a plurality of agents to identify an agent that modulates the polypeptide activity by binding to a polypeptide of the invention, modulating inhibitor binding to the polypeptide or activating expression of the polypeptide or polynucleotide, for example.


1. Binding Assays


Preliminary screens can be conducted by screening for agents capable of binding to a polypeptide of the invention, as at least some of the agents so identified are likely modulators of polypeptide activity. The binding assays usually involve contacting a polypeptide of the invention with one or more test agents and allowing sufficient time for the protein and test agents to form a binding complex. Any binding complexes formed can be detected using any of a number of established analytical techniques. Protein binding assays include, but are not limited to, methods that measure co-precipitation, co-migration on non-denaturing SDS-polyacrylamide gels, and co-migration on Western blots (see, e.g., Bennet and Yamamura, (1985) “Neurotransmitter, Hormone or Drug Receptor Binding Methods,” in Neurotransmitter Receptor Binding (Yamamura, H. I., et al, eds.), pp. 61-89. The protein utilized in such assays can be naturally expressed, cloned or synthesized.


Binding assays are also useful, e.g., for identifying endogenous proteins that interact with a polypeptide of the invention. For example, antibodies, receptors or other molecules that bind a polypeptide of the invention can be identified in binding assays.


2. Expression Assays


Certain screening methods involve screening for a compound that up or down-regulates the expression of a polypeptide or polynucleotide of the invention. Such methods generally involve conducting cell-based assays in which test compounds are contacted with one or more cells expressing a polypeptide or polynucleotide of the invention and then detecting an increase or decrease in expression (either transcript, translation product, or catalytic product). Some assays are performed with peripheral cells, or other cells, that express an endogenous polypeptide or polynucleotide of the invention.


Polypeptide or polynucleotide expression can be detected in a number of different ways. As described infra, the expression level of a polynucleotide of the invention in a cell can be determined by probing the mRNA expressed in a cell with a probe that specifically hybridizes with a transcript (or complementary nucleic acid derived therefrom) of a polynucleotide of the invention. Probing can be conducted by lysing the cells and conducting Northern blots or without lysing the cells using in situ-hybridization techniques. Alternatively, a polypeptide of the invention can be detected using immunological methods in which a cell lysate is probed with antibodies that specifically bind to a polypeptide of the invention.


Other cell-based assays are reporter assays conducted with cells that do not express a polypeptide or polynucleotide of the invention. Certain of these assays are conducted with a heterologous nucleic acid construct that includes a promoter of a polynucleotide of the invention that is operably linked to a reporter gene that encodes a detectable product. A number of different reporter genes can be utilized. Some reporters are inherently detectable. An example of such a reporter is green fluorescent protein that emits fluorescence that can be detected with a fluorescence detector. Other reporters generate a detectable product. Often such reporters are enzymes. Exemplary enzyme reporters include, but are not limited to, β-glucuronidase, chloramphenicol acetyl transferase (CAT); Alton and Vapnek (1979) Nature 282:864-869), luciferase, β-galactosidase, green fluorescent protein (GFP) and alkaline phosphatase (Toh, et al. (1980) Eur. J. Biochem. 182:231-238; and Hall et al. (1983) J. Mol. Appl. Gen. 2:101).


In these assays, cells harboring the reporter construct are contacted with a test compound. A test compound that either activates the promoter by binding to it or triggers a cascade that produces a molecule that activates the promoter causes expression of the detectable reporter. Certain other reporter assays are conducted with cells that harbor a heterologous construct that includes a transcriptional control element that activates expression of a polynucleotide of the invention and a reporter operably linked thereto. Here, too, an agent that binds to the transcriptional control element to activate expression of the reporter or that triggers the formation of an agent that binds to the transcriptional control element to activate reporter expression, can be identified by the generation of signal associated with reporter expression.


The level of expression or activity can be compared to a baseline value. As indicated above, the baseline value can be a value for a control sample or a statistical value that is representative of expression levels for a control population (e.g., healthy individuals not having or at risk for mood disorders or psychotic disorders). Expression levels can also be determined for cells that do not express a polynucleotide of the invention as a negative control. Such cells generally are otherwise substantially genetically the same as the test cells.


A variety of different types of cells can be utilized in the reporter assays. Cells that express an endogenous polypeptide or polynucleotide of the invention include, e.g., brain cells, including cells from the cerebellum, anterior cingulate cortex, or dorsolateral prefrontal cortex. Cells that do not endogenously express polynucleotides of the invention can be prokaryotic, but are preferably eukaryotic. The eukaryotic cells can be any of the cells typically utilized in generating cells that harbor recombinant nucleic acid constructs. Exemplary eukaryotic cells include, but are not limited to, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cell lines and stem cells, e.g., neural stem cells.


Various controls can be conducted to ensure that an observed activity is authentic including running parallel reactions with cells that lack the reporter construct or by not contacting a cell harboring the reporter construct with test compound. Compounds can also be further validated as described below.


3. Catalytic Activity


Catalytic activity of polypeptides of the invention can be determined by measuring the production of enzymatic products or by measuring the consumption of substrates. Activity refers to either the rate of catalysis or the ability to the polypeptide to bind (Km) the substrate or release the catalytic product (Kd).


Analysis of the activity of polypeptides of the invention are performed according to general biochemical analyses. Such assays include cell-based assays as well as in vitro assays involving purified or partially purified polypeptides or crude cell lysates. The assays generally involve providing a known quantity of substrate and quantifying product as a function of time.


4. Validation


Agents that are initially identified by any of the foregoing screening methods can be further tested to validate the apparent activity. Preferably such studies are conducted with suitable animal models. The basic format of such methods involves administering a lead compound identified during an initial screen to an animal that serves as a model for humans and then determining if expression or activity of a polynucleotide or polypeptide of the invention is in fact upregulated. The animal models utilized in validation studies generally are mammals of any kind. Specific examples of suitable animals include, but are not limited to, primates, mice, and rats.


5. Animal Models


Animal models of mental disorders also find use in screening for modulators. In one embodiment, rat models of schizophrenia or other mental disorder, such as depression, are used for screening. In one embodiment, invertebrate models such as Drosophila models can be used, screening for modulators of Drosophila orthologs of the human genes disclosed herein. In another embodiment, transgenic animal technology including gene knockout technology, for example as a result of homologous recombination with an appropriate gene targeting vector, or gene overexpression, will result in the absence, decreased or increased expression of a polynucleotide or polypeptide of the invention. The same technology can also be applied to make knockout cells. When desired, tissue-specific expression or knockout of a polynucleotide or polypeptide of the invention may be necessary. Transgenic animals generated by such methods find use as animal models of mental disorder and are useful in screening for modulators of mental disorder.


Knockout cells and transgenic mice can be made by insertion of a marker gene or other heterologous gene into an endogenous gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting an endogenous polynucleotide of the invention with a mutated version of the polynucleotide, or by mutating an endogenous polynucleotide, e.g., by exposure to carcinogens.


For development of appropriate stem cells, a DNA construct is introduced into the nuclei of embryonic stem cells. Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells partially derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, e.g., Capecchi et al., Science 244:1288 (1989)). Chimeric targeted mice can be derived according to Hogan et al., Manipulating the Mouse Embryo: A Laboratory Manual, Cold Spring Harbor Laboratory (1988) and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed., IRL Press, Washington, D.C., (1987).


B. Modulators of Polypeptides or Polynucleotides of the Invention


The agents tested as modulators of the polypeptides or polynucleotides of the invention can be any small chemical compound, or a biological entity, such as a protein, sugar, nucleic acid or lipid. Alternatively, modulators can be genetically altered versions of a polypeptide or polynucleotide of the invention. Typically, test compounds will be small chemical molecules and peptides. Essentially any chemical compound can be used as a potential modulator or ligand in the assays of the invention, although most often compounds that can be dissolved in aqueous or organic (especially DMSO-based) solutions are used. The assays are designed to screen large chemical libraries by automating the assay steps and providing compounds from any convenient source to assays, which are typically run in parallel (e.g., in microtiter formats on microtiter plates in robotic assays). It will be appreciated that there are many suppliers of chemical compounds, including Sigma (St. Louis, Mo.), Aldrich (St. Louis, Mo.), Sigma-Aldrich (St. Louis, Mo.), Fluka Chemika-Biochemica Analytika (Buchs, Switzerland) and the like. Modulators also include agents designed to reduce the level of mRNA of the invention (e.g. antisense molecules, ribozymes, DNAzymes and the like) or the level of translation from an mRNA.


In one preferred embodiment, high throughput screening methods involve providing a combinatorial chemical or peptide library containing a large number of potential therapeutic compounds (potential modulator or ligand compounds). Such “combinatorial chemical libraries” or “ligand libraries” are then screened in one or more assays, as described herein, to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds” or can themselves be used as potential or actual therapeutics.


A combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis, by combining a number of chemical “building blocks” such as reagents. For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks (amino acids) in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.


Preparation and screening of combinatorial chemical libraries is well known to those of skill in the art. Such combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Pat. No. 5,010,175, Furka, Int. J. Pept. Prot. Res. 37:487-493 (1991) and Houghton et al., Nature 354:84-88 (1991)). Other chemistries for generating chemical diversity libraries can also be used. Such chemistries include, but are not limited to: peptoids (e.g., PCT Publication No. WO 91/19735), encoded peptides (e.g., PCT Publication WO 93/20242), random bio-oligomers (e.g., PCT Publication No. WO 92/00091), benzodiazepines (e.g., U.S. Pat. No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., Proc. Nat. Acad. Sci. USA 90:6909-6913 (1993)), vinylogous polypeptides (Hagihara et al., J. Amer. Chem. Soc. 114:6568 (1992)), nonpeptidal peptidomimetics with glucose scaffolding (Hirschmann et al., J. Amer. Chem. Soc. 114:9217-9218 (1992)), analogous organic syntheses of small compound libraries (Chen et al., J. Amer. Chem. Soc. 116:2661 (1994)), oligocarbamates (Cho et al., Science 261:1303 (1993)), and/or peptidyl phosphonates (Campbell et al., J. Org. Chem. 59:658 (1994)), nucleic acid libraries (see Ausubel, Berger and Sambrook, all supra), peptide nucleic acid libraries (see, e.g., U.S. Pat. No. 5,539,083), antibody libraries (see, e.g., Vaughn et al., Nature Biotechnology, 14(3):309-314 (1996) and PCT/US96/10287), carbohydrate libraries (see, e.g., Liang et al., Science, 274:1520-1522 (1996) and U.S. Pat. No. 5,593,853), small organic molecule libraries (see, e.g., benzodiazepines, Baum C&EN, January 18, page 33 (1993); isoprenoids, U.S. Pat. No. 5,569,588; thiazolidinones and metathiazanones, U.S. Pat. No. 5,549,974; pyrrolidines, U.S. Pat. Nos. 5,525,735 and 5,519,134; morpholino compounds, U.S. Pat. No. 5,506,337; benzodiazepines, U.S. Pat. No. 5,288,514, and the like).


Devices for the preparation of combinatorial libraries are commercially available (see, e.g., 357 MPS, 390 MPS, Advanced Chem Tech, Louisville Ky.; Symphony, Rainin, Woburn, Mass.; 433A Applied Biosystems, Foster City, Calif.; 9050 Plus, Millipore, Bedford, Mass.). In addition, numerous combinatorial libraries are themselves commercially available (see, e.g., ComGenex, Princeton, N.J.; Tripos, Inc., St. Louis, Mo.; 3D Pharmaceuticals, Exton, Pa.; Martek Biosciences, Columbia, Md., etc.).


C. Solid State and Soluble High Throughput Assays


In the high throughput assays of the invention, it is possible to screen up to several thousand different modulators or ligands in a single day. In particular, each well of a microtiter plate can be used to run a separate assay against a selected potential modulator, or, if concentration or incubation time effects are to be observed, every 5-10 wells can test a single modulator. Thus, a single standard microtiter plate can assay about 100 (e.g., 96) modulators. If 1536 well plates are used, then a single plate can easily assay from about 100 to about 1500 different compounds. It is possible to assay several different plates per day; assay screens for up to about 6,000-20,000 different compounds are possible using the integrated systems of the invention. More recently, microfluidic approaches to reagent manipulation have been developed.


The molecule of interest can be bound to the solid state component, directly or indirectly, via covalent or non-covalent linkage, e.g., via a tag. The tag can be any of a variety of components. In general, a molecule that binds the tag (a tag binder) is fixed to a solid support, and the tagged molecule of interest is attached to the solid support by interaction of the tag and the tag binder.


A number of tags and tag binders can be used, based upon known molecular interactions well described in the literature. For example, where a tag has a natural binder, for example, biotin, protein A, or protein G, it can be used in conjunction with appropriate tag binders (avidin, streptavidin, neutravidin, the Fc region of an immunoglobulin, etc.). Antibodies to molecules with natural binders such as biotin are also widely available and appropriate tag binders (see, SIGMA Immunochemicals 1998 catalogue SIGMA, St. Louis Mo.).


Similarly, any haptenic or antigenic compound can be used in combination with an appropriate antibody to form a tag/tag binder pair. Thousands of specific antibodies are commercially available and many additional antibodies are described in the literature. For example, in one common configuration, the tag is a first antibody and the tag binder is a second antibody which recognizes the first antibody. In addition to antibody-antigen interactions, receptor-ligand interactions are also appropriate as tag and tag-binder pairs, such as agonists and antagonists of cell membrane receptors (e.g., cell receptor-ligand interactions such as transferrin, c-kit, viral receptor ligands, cytokine receptors, chemokine receptors, interleukin receptors, immunoglobulin receptors and antibodies, the cadherin family, the integrin family, the selectin family, and the like; see, e.g., Pigott & Power, The Adhesion Molecule Facts Book I (1993)). Similarly, toxins and venoms, viral epitopes, hormones (e.g., opiates, steroids, etc.), intracellular receptors (e.g., which mediate the effects of various small ligands, including steroids, thyroid hormone, retinoids and vitamin D; peptides), drugs, lectins, sugars, nucleic acids (both linear and cyclic polymer configurations), oligosaccharides, proteins, phospholipids and antibodies can all interact with various cell receptors.


Synthetic polymers, such as polyurethanes, polyesters, polycarbonates, polyureas, polyamides, polyethyleneimines, polyarylene sulfides, polysiloxanes, polyimides, and polyacetates can also form an appropriate tag or tag binder. Many other tag/tag binder pairs are also useful in assay systems described herein, as would be apparent to one of skill upon review of this disclosure.


Common linkers such as peptides, polyethers, and the like can also serve as tags, and include polypeptide sequences, such as poly-Gly sequences of between about 5 and 200 amino acids. Such flexible linkers are known to those of skill in the art. For example, poly(ethelyne glycol) linkers are available from Shearwater Polymers, Inc., Huntsville, Ala. These linkers optionally have amide linkages, sulfhydryl linkages, or heterofunctional linkages.


Tag binders are fixed to solid substrates using any of a variety of methods currently available. Solid substrates are commonly derivatized or functionalized by exposing all or a portion of the substrate to a chemical reagent which fixes a chemical group to the surface which is reactive with a portion of the tag binder. For example, groups which are suitable for attachment to a longer chain portion would include amines, hydroxyl, thiol, and carboxyl groups. Aminoalkylsilanes and hydroxyalkylsilanes can be used to functionalize a variety of surfaces, such as glass surfaces. The construction of such solid phase biopolymer arrays is well described in the literature (see, e.g., Merrifield, J. Am. Chem. Soc. 85:2149-2154 (1963) (describing solid phase synthesis of, e.g., peptides); Geysen et al., J Immun. Meth. 102:259-274 (1987) (describing synthesis of solid phase components on pins); Frank and Doring, Tetrahedron 44:60316040 (1988) (describing synthesis of various peptide sequences on cellulose disks); Fodor et al., Science, 251:767-777 (1991); Sheldon et al., Clinical Chemistry 39(4):718-719 (1993); and Kozal et al., Nature Medicine 2(7):753759 (1996) (all describing arrays of biopolymers fixed to solid substrates). Non-chemical approaches for fixing tag binders to substrates include other common methods, such as heat, cross-linking by UV radiation, and the like.


The invention provides in vitro assays for identifying, in a high throughput format, compounds that can modulate the expression or activity of the polynucleotides or polypeptides of the invention. In a preferred embodiment, the methods of the invention include such a control reaction. For each of the assay formats described, “no modulator” control reactions that do not include a modulator provide a background level of binding activity.


In some assays it will be desirable to have positive controls to ensure that the components of the assays are working properly. At least two types of positive controls are appropriate. First, a known activator of a polynucleotide or polypeptide of the invention can be incubated with one sample of the assay, and the resulting increase in signal resulting from an increased expression level or activity of polynucleotide or polypeptide determined according to the methods herein. Second, a known inhibitor of a polynucleotide or polypeptide of the invention can be added, and the resulting decrease in signal for the expression or activity can be similarly detected.


D. Computer-Based Assays


Yet another assay for compounds that modulate the activity of a polypeptide or polynucleotide of the invention involves computer assisted drug design, in which a computer system is used to generate a three-dimensional structure of the polypeptide or polynucleotide based on the structural information encoded by its amino acid or nucleotide sequence. The input sequence interacts directly and actively with a pre-established algorithm in a computer program to yield secondary, tertiary, and quaternary structural models of the molecule. Similar analyses can be performed on potential receptors or binding partners of the polypeptides or polynucleotides of the invention. The models of the protein or nucleotide structure are then examined to identify regions of the structure that have the ability to bind, e.g., a polypeptide or polynucleotide of the invention. These regions are then used to identify polypeptides that bind to a polypeptide or polynucleotide of the invention.


The three-dimensional structural model of a protein is generated by entering protein amino acid sequences of at least 10 amino acid residues or corresponding nucleic acid sequences encoding a potential receptor into the computer system. The amino acid sequences encoded by the nucleic acid sequences provided herein represent the primary sequences or subsequences of the proteins, which encode the structural information of the proteins. At least 10 residues of an amino acid sequence (or a nucleotide sequence encoding 10 amino acids) are entered into the computer system from computer keyboards, computer readable substrates that include, but are not limited to, electronic storage media (e.g., magnetic diskettes, tapes, cartridges, and chips), optical media (e.g., CD ROM), information distributed by internet sites, and by RAM. The three-dimensional structural model of the protein is then generated by the interaction of the amino acid sequence and the computer system, using software known to those of skill in the art.


The amino acid sequence represents a primary structure that encodes the information necessary to form the secondary, tertiary, and quaternary structure of the protein of interest. The software looks at certain parameters encoded by the primary sequence to generate the structural model. These parameters are referred to as “energy terms,” and primarily include electrostatic potentials, hydrophobic potentials, solvent accessible surfaces, and hydrogen bonding. Secondary energy terms include van der Waals potentials. Biological molecules form the structures that minimize the energy terms in a cumulative fashion. The computer program is therefore using these terms encoded by the primary structure or amino acid sequence to create the secondary structural model.


The tertiary structure of the protein encoded by the secondary structure is then formed on the basis of the energy terms of the secondary structure. The user at this point can enter additional variables such as whether the protein is membrane bound or soluble, its location in the body, and its cellular location, e.g., cytoplasmic, surface, or nuclear. These variables along with the energy terms of the secondary structure are used to form the model of the tertiary structure. In modeling the tertiary structure, the computer program matches hydrophobic faces of secondary structure with like, and hydrophilic faces of secondary structure with like.


Once the structure has been generated, potential ligand binding regions are identified by the computer system. Three-dimensional structures for potential ligands are generated by entering amino acid or nucleotide sequences or chemical formulas of compounds, as described above. The three-dimensional structure of the potential ligand is then compared to that of a polypeptide or polynucleotide of the invention to identify binding sites of the polypeptide or polynucleotide of the invention. Binding affinity between the protein and ligands is determined using energy terms to determine which ligands have an enhanced probability of binding to the protein.


Computer systems are also used to screen for mutations, polymorphic variants, alleles and interspecies homologs of genes encoding a polypeptide or polynucleotide of the invention. Such mutations can be associated with disease states or genetic traits and can be used for diagnosis. As described above, GeneChip™ and related technology can also be used to screen for mutations, polymorphic variants, alleles and interspecies homologs. Once the variants are identified, diagnostic assays can be used to identify patients having such mutated genes. Identification of the mutated a polypeptide or polynucleotide of the invention involves receiving input of a first amino acid sequence of a polypeptide of the invention (or of a first nucleic acid sequence encoding a polypeptide of the invention), e.g., any amino acid sequence having at least 60%, optionally at least 70% or 85%, identity with the amino acid sequence of interest, or conservatively modified versions thereof. The sequence is entered into the computer system as described above. The first nucleic acid or amino acid sequence is then compared to a second nucleic acid or amino acid sequence that has substantial identity to the first sequence. The second sequence is entered into the computer system in the manner described above. Once the first and second sequences are compared, nucleotide or amino acid differences between the sequences are identified. Such sequences can represent allelic differences in various polynucleotides, including SNPs and/or haplotypes, of the invention, and mutations associated with disease states and genetic traits.


VII. Compositions, Kits and Integrated Systems


The invention provides compositions, kits and integrated systems for practicing the assays described herein using polypeptides or polynucleotides of the invention, antibodies specific for polypeptides or polynucleotides of the invention, etc.


The invention provides assay compositions for use in solid phase assays; such compositions can include, for example, one or more polynucleotides or polypeptides of the invention immobilized on a solid support, and a labeling reagent. In each case, the assay compositions can also include additional reagents that are desirable for hybridization. Modulators of expression or activity of polynucleotides or polypeptides of the invention can also be included in the assay compositions.


The invention also provides kits for carrying out the therapeutic and diagnostic assays of the invention. The kits typically include a probe that comprises an antibody that specifically binds to polypeptides or polynucleotides of the invention, and a label for detecting the presence of the probe. The kits may include several polynucleotide sequences encoding polypeptides of the invention. Kits can include any of the compositions noted above, and optionally further include additional components such as instructions to practice a high-throughput method of assaying for an effect on expression of the genes encoding the polypeptides of the invention, or on activity of the polypeptides of the invention, one or more containers or compartments (e.g., to hold the probe, labels, or the like), a control modulator of the expression or activity of polypeptides of the invention, a robotic armature for mixing kit components or the like.


The invention also provides integrated systems for high-throughput screening of potential modulators for an effect on the expression or activity of the polypeptides of the invention. The systems typically include a robotic armature which transfers fluid from a source to a destination, a controller which controls the robotic armature, a label detector, a data storage unit which records label detection, and an assay component such as a microtiter dish comprising a well having a reaction mixture or a substrate comprising a fixed nucleic acid or immobilization moiety.


A number of robotic fluid transfer systems are available, or can easily be made from existing components. For example, a Zymate XP (Zymark Corporation; Hopkinton, Mass.) automated robot using a Microlab 2200 (Hamilton; Reno, Nev.) pipetting station can be used to transfer parallel samples to 96 well microtiter plates to set up several parallel simultaneous STAT binding assays.


Optical images viewed (and, optionally, recorded) by a camera or other recording device (e.g., a photodiode and data storage device) are optionally further processed in any of the embodiments herein, e.g., by digitizing the image and storing and analyzing the image on a computer. A variety of commercially available peripheral equipment and software is available for digitizing, storing and analyzing a digitized video or digitized optical image, e.g., using PC, MACINTOSH®, or UNIX® based (e.g., SUN® work station) computers.


One conventional system carries light from the specimen field to a cooled charge-coupled device (CCD) camera, in common use in the art. A CCD camera includes an array of picture elements (pixels). The light from the specimen is imaged on the CCD. Particular pixels corresponding to regions of the specimen (e.g., individual hybridization sites on an array of biological polymers) are sampled to obtain light intensity readings for each position. Multiple pixels are processed in parallel to increase speed. The apparatus and methods of the invention are easily used for viewing any sample, e.g., by fluorescent or dark field microscopic techniques. Lasar based systems can also be used.


VIII. Administration and Pharmaceutical Compositions


Modulators of the polynucleotides or polypeptides of the invention (e.g., antagonists or agonists) can be administered directly to a mammalian subject for modulation of activity of those molecules in vivo. Administration is by any of the routes normally used for introducing a modulator compound into ultimate contact with the tissue to be treated and is well known to those of skill in the art. Although more than one route can be used to administer a particular composition, a particular route can often provide a more immediate and more effective reaction than another route.


Diseases that can be treated include the following, which include the corresponding reference number from Morrison, DSM-IVMade Easy, 1995: Schizophrenia, Catatonic, Subchronic, (295.21); Schizophrenia, Catatonic, Chronic (295.22); Schizophrenia, Catatonic, Subchronic with Acute Exacerbation (295.23); Schizophrenia, Catatonic, Chronic with Acute Exacerbation (295.24); Schizophrenia, Catatonic, in Remission (295.55); Schizophrenia, Catatonic, Unspecified (295.20); Schizophrenia, Disorganized, Subchronic (295.11); Schizophrenia, Disorganized, Chronic (295.12); Schizophrenia, Disorganized, Subchronic with Acute Exacerbation (295.13); Schizophrenia, Disorganized, Chronic with Acute Exacerbation (295.14); Schizophrenia, Disorganized, in Remission (295.15); Schizophrenia, Disorganized, Unspecified (295.10); Schizophrenia, Paranoid, Subchronic (295.31); Schizophrenia, Paranoid, Chronic (295.32); Schizophrenia, Paranoid, Subchronic with Acute Exacerbation (295.33); Schizophrenia, Paranoid, Chronic with Acute Exacerbation (295.34); Schizophrenia, Paranoid, in Remission (295.35); Schizophrenia, Paranoid, Unspecified (295.30); Schizophrenia, Undifferentiated, Subchronic (295.91); Schizophrenia, Undifferentiated, Chronic (295.92); Schizophrenia, Undifferentiated, Subchronic with Acute Exacerbation (295.93); Schizophrenia, Undifferentiated, Chronic with Acute Exacerbation (295.94); Schizophrenia, Undifferentiated, in Remission (295.95); Schizophrenia, Undifferentiated, Unspecified (295.90); Schizophrenia, Residual, Subchronic (295.61); Schizophrenia, Residual, Chronic (295.62); Schizophrenia, Residual, Subchronic with Acute Exacerbation (295.63); Schizophrenia, Residual, Chronic with Acute Exacerbation (295.94); Schizophrenia, Residual, in Remission (295.65); Schizophrenia, Residual, Unspecified (295.60); Delusional (Paranoid) Disorder (297.10); Brief Reactive Psychosis (298.80); Schizophreniform Disorder (295.40); Schizoaffective Disorder (295.70); Induced Psychotic Disorder (297.30); Psychotic Disorder NOS (Atypical Psychosis) (298.90); Personality Disorders, Paranoid (301.00); Personality Disorders, Schizoid (301.20); Personality Disorders, Schizotypal (301.22); Personality Disorders, Antisocial (301.70); Personality Disorders, Borderline (301.83) and bipolar disorders, maniac, hypomaniac, dysthymnic or cyclothymic disorders, substance-induced major depression, psychotic disorder, including schizophrenia (paranoid, catatonic, delusional) having schizoaffective disorder, and substance-induced psychotic disorder.


In some embodiments, modulators of polynucleotides or polypeptides of the invention can be combined with other drugs useful for treating mental disorders including psychotic disorders, e.g., schizophrenia; and mood disorders, e.g., bipolar disorders, or major depression. In some preferred embodiments, pharmaceutical compositions of the invention comprise a modulator of a polypeptide of polynucleotide of the invention combined with at least one of the compounds useful for treating schizophrenia, bipolar disorder, or major depression, e.g., such as those described in U.S. Pat. No. 6,297,262; 6,284,760; 6,284,771; 6,232,326; 6,187,752; 6,117,890; 6,239,162 or 6,166,008.


The pharmaceutical compositions of the invention may comprise a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are determined in part by the particular composition being administered, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of pharmaceutical compositions of the present invention (see, e.g., Remington's Pharmaceutical Sciences, 17th ed. 1985)).


The modulators (e.g., agonists or antagonists) of the expression or activity of the a polypeptide or polynucleotide of the invention, alone or in combination with other suitable components, can be made into aerosol formulations (i.e., they can be “nebulized”) to be administered via inhalation or in compositions useful for injection. Aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like.


Formulations suitable for administration include aqueous and non-aqueous solutions, isotonic sterile solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. In the practice of this invention, compositions can be administered, for example, orally, nasally, topically, intravenously, intraperitoneally, or intrathecally. The formulations of compounds can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials. Solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. The modulators can also be administered as part of a prepared food or drug.


The dose administered to a patient, in the context of the present invention should be sufficient to effect a beneficial response in the subject over time. The optimal dose level for any patient will depend on a variety of factors including the efficacy of the specific modulator employed, the age, body weight, physical activity, and diet of the patient, on a possible combination with other drugs, and on the severity of the mental disorder. The size of the dose also will be determined by the existence, nature, and extent of any adverse side effects that accompany the administration of a particular compound or vector in a particular subject.


In determining the effective amount of the modulator to be administered a physician may evaluate circulating plasma levels of the modulator, modulator toxicity, and the production of anti-modulator antibodies. In general, the dose equivalent of a modulator is from about 1 ng/kg to 10 mg/kg for a typical subject.


For administration, modulators of the present invention can be administered at a rate determined by the LD-50 of the modulator, and the side effects of the modulator at various concentrations, as applied to the mass and overall health of the subject. Administration can be accomplished via single or divided doses.


IX. Gene Therapy Applications


A variety of human diseases can be treated by therapeutic approaches that involve stably introducing a gene into a human cell such that the gene is transcribed and the gene product is produced in the cell. Diseases amenable to treatment by this approach include inherited diseases, including those in which the defect is in a single or multiple genes. Gene therapy is also useful for treatment of acquired diseases and other conditions. For discussions on the application of gene therapy towards the treatment of genetic as well as acquired diseases, see, Miller, Nature 357:455-460 (1992); and Mulligan, Science 260:926-932 (1993).


In the context of the present invention, gene therapy can be used for treating a variety of disorders and/or diseases in which the polynucleotides and polypeptides of the invention has been implicated. For example, compounds, including polynucleotides, can be identified by the methods of the present invention as effective in treating a mental disorder. Introduction by gene therapy of these polynucleotides can then be used to treat, e.g., mental disorders including mood disorders or psychotic disorders (e.g., schizophrenia).


A. Vectors for Gene Delivery


For delivery to a cell or organism, the polynucleotides of the invention can be incorporated into a vector. Examples of vectors used for such purposes include expression plasmids capable of directing the expression of the nucleic acids in the target cell. In other instances, the vector is a viral vector system wherein the nucleic acids are incorporated into a viral genome that is capable of transfecting the target cell. In a preferred embodiment, the polynucleotides can be operably linked to expression and control sequences that can direct expression of the gene in the desired target host cells. Thus, one can achieve expression of the nucleic acid under appropriate conditions in the target cell.


B. Gene Delivery Systems


Viral vector systems useful in the expression of the nucleic acids include, for example, naturally occurring or recombinant viral vector systems. Depending upon the particular application, suitable viral vectors include replication competent, replication deficient, and conditionally replicating viral vectors. For example, viral vectors can be derived from the genome of human or bovine adenoviruses, vaccinia virus, herpes virus, adeno-associated virus, minute virus of mice (MVM), HIV, sindbis virus, and retroviruses (including but not limited to Rous sarcoma virus), and MoMLV. Typically, the genes of interest are inserted into such vectors to allow packaging of the gene construct, typically with accompanying viral DNA, followed by infection of a sensitive host cell and expression of the gene of interest.


As used herein, “gene delivery system” refers to any means for the delivery of a nucleic acid of the invention to a target cell. In some embodiments of the invention, nucleic acids are conjugated to a cell receptor ligand for facilitated uptake (e.g., invagination of coated pits and internalization of the endosome) through an appropriate linking moiety, such as a DNA linking moiety (Wu et al., J. Biol. Chem. 263:14621-14624 (1988); WO 92/06180). For example, nucleic acids can be linked through a polylysine moiety to asialo-oromucocid, which is a ligand for the asialoglycoprotein receptor of hepatocytes.


Similarly, viral envelopes used for packaging gene constructs that include the nucleic acids of the invention can be modified by the addition of receptor ligands or antibodies specific for a receptor to permit receptor-mediated endocytosis into specific cells (see, e.g., WO 93/20221, WO 93/14188, and WO 94/06923). In some embodiments of the invention, the DNA constructs of the invention are linked to viral proteins, such as adenovirus particles, to facilitate endocytosis (Curiel et al., Proc. Natl. Acad. Sci. U.S.A. 88:8850-8854 (1991)). In other embodiments, molecular conjugates of the instant invention can include microtubule inhibitors (WO/9406922), synthetic peptides mimicking influenza virus hemagglutinin (Plank et al., J. Biol. Chem. 269:12918-12924 (1994)), and nuclear localization signals such as SV40 T antigen (WO93/19768).


Retroviral vectors are also useful for introducing the nucleic acids of the invention into target cells or organisms. Retroviral vectors are produced by genetically manipulating retroviruses. The viral genome of retroviruses is RNA. Upon infection, this genomic RNA is reverse transcribed into a DNA copy which is integrated into the chromosomal DNA of transduced cells with a high degree of stability and efficiency. The integrated DNA copy is referred to as a provirus and is inherited by daughter cells as is any other gene. The wild type retroviral genome and the proviral DNA have three genes: the gag, the pol and the env genes, which are flanked by two long terminal repeat (LTR) sequences. The gag gene encodes the internal structural (nucleocapsid) proteins; the pol gene encodes the RNA directed DNA polymerase (reverse transcriptase); and the env gene encodes viral envelope glycoproteins. The 5′ and 3′ LTRs serve to promote transcription and polyadenylation of virion RNAs. Adjacent to the 5′ LTR are sequences necessary for reverse transcription of the genome (the tRNA primer binding site) and for efficient encapsulation of viral RNA into particles (the Psi site) (see, Mulligan, In: Experimental Manipulation of Gene Expression, Inouye (ed), 155-173 (1983); Mann et al., Cell 33:153-159 (1983); Cone and Mulligan, Proceedings of the National Academy of Sciences, U.S.A., 81:6349-6353 (1984)).


The design of retroviral vectors is well known to those of ordinary skill in the art. In brief, if the sequences necessary for encapsidation (or packaging of retroviral RNA into infectious virions) are missing from the viral genome, the result is a cis-acting defect which prevents encapsidation of genomic RNA. However, the resulting mutant is still capable of directing the synthesis of all virion proteins. Retroviral genomes from which these sequences have been deleted, as well as cell lines containing the mutant genome stably integrated into the chromosome are well known in the art and are used to construct retroviral vectors. Preparation of retroviral vectors and their uses are described in many publications including, e.g., European Patent Application EPA 0 178 220; U.S. Pat. No. 4,405,712, Gilboa Biotechniques 4:504-512 (1986); Mann et al., Cell 33:153-159 (1983); Cone and Mulligan Proc. Natl. Acad. Sci. USA 81:6349-6353 (1984); Eglitis et al. Biotechniques 6:608-614 (1988); Miller et al. Biotechniques 7:981-990 (1989); Miller (1992) supra; Mulligan (1993), supra; and WO 92/07943.


The retroviral vector particles are prepared by recombinantly inserting the desired nucleotide sequence into a retrovirus vector and packaging the vector with retroviral capsid proteins by use of a packaging cell line. The resultant retroviral vector particle is incapable of replication in the host cell but is capable of integrating into the host cell genome as a proviral sequence containing the desired nucleotide sequence. As a result, the patient is capable of producing, for example, a polypeptide or polynucleotide of the invention and thus restore the cells to a normal phenotype.


Packaging cell lines that are used to prepare the retroviral vector particles are typically recombinant mammalian tissue culture cell lines that produce the necessary viral structural proteins required for packaging, but which are incapable of producing infectious virions. The defective retroviral vectors that are used, on the other hand, lack these structural genes but encode the remaining proteins necessary for packaging. To prepare a packaging cell line, one can construct an infectious clone of a desired retrovirus in which the packaging site has been deleted. Cells comprising this construct will express all structural viral proteins, but the introduced DNA will be incapable of being packaged. Alternatively, packaging cell lines can be produced by transforming a cell line with one or more expression plasmids encoding the appropriate core and envelope proteins. In these cells, the gag, pol, and env genes can be derived from the same or different retroviruses.


A number of packaging cell lines suitable for the present invention are also available in the prior art. Examples of these cell lines include Crip, GPE86, PA317 and PG13 (see Miller et al., J. Virol. 65:2220-2224 (1991)). Examples of other packaging cell lines are described in Cone and Mulligan Proceedings of the National Academy of Sciences, USA, 81:6349-6353 (1984); Danos and Mulligan Proceedings of the National Academy of Sciences, USA, 85:6460-6464 (1988); Eglitis et al. (1988), supra; and Miller (1990), supra.


Packaging cell lines capable of producing retroviral vector particles with chimeric envelope proteins may be used. Alternatively, amphotropic or xenotropic envelope proteins, such as those produced by PA317 and GPX packaging cell lines may be used to package the retroviral vectors.


In some embodiments of the invention, an antisense polynucleotide is administered which hybridizes to a gene encoding a polypeptide of the invention. The antisense polypeptide can be provided as an antisense oligonucleotide (see, e.g., Murayama et al., Antisense Nucleic Acid Drug Dev. 7:109-114 (1997)). Genes encoding an antisense nucleic acid can also be provided; such genes can be introduced into cells by methods known to those of skill in the art. For example, one can introduce an antisense nucleotide sequence in a viral vector, such as, for example, in hepatitis B virus (see, e.g., Ji et al., J Viral Hepat. 4:167-173 (1997)), in adeno-associated virus (see, e.g., Xiao et al., Brain Res. 756:76-83 (1997)), or in other systems including, but not limited, to an HVJ (Sendai virus)-liposome gene delivery system (see, e.g., Kaneda et al., Ann. NY Acad. Sci. 811:299-308 (1997)), a “peptide vector” (see, e.g., Vidal et al., CR Acad. Sci III 32:279-287 (1997)), as a gene in an episomal or plasmid vector (see, e.g., Cooper et al., Proc. Natl. Acad. Sci. USA. 94:6450-6455 (1997), Yew et al. Hum Gene Ther. 8:575-584 (1997)), as a gene in a peptide-DNA aggregate (see, e.g., Niidome et al., J. Biol. Chem. 272:15307-15312 (1997)), as “naked DNA” (see, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466), in lipidic vector systems (see, e.g., Lee et al., Crit Rev Ther Drug Carrier Syst. 14:173-206 (1997)), polymer coated liposomes (U.S. Pat. Nos. 5,213,804 and 5,013,556), cationic liposomes (Epand et al., U.S. Pat. Nos. 5,283,185; 5,578,475; 5,279,833; and 5,334,761), gas filled microspheres (U.S. Pat. No. 5,542,935), ligand-targeted encapsulated macromolecules (U.S. Pat. Nos. 5,108,921; 5,521,291; 5,554,386; and 5,166,320).


Upregulated transcripts listed in Tables 1-14 which are correlated with schizophrenia may be targeted with one or more short interfering RNA (siRNA) sequences that hybridize to specific sequences in the target, as described above. Targeting of certain brain transcripts with siRNA in vivo has been reported, for example, by Zhang et al., J. Gene. Med., 12:1039-45 (2003), who utilized monoclonal antibodies against the transferrin receptor to facilitate passage of liposome-encapsulated siRNA molecules through the blood brain barrier. Targeted siRNAs represent useful therapeutic compounds for attenuating the over-expressed transcripts that are associated with disease states, e.g., schizophrenia.


In another embodiment, conditional expression systems, such as those typified by the tet-regulated systems and the RU-486 system, can be used (see, e.g., Gossen & Bujard, PNAS 89:5547 (1992); Oligino et al., Gene Ther. 5:491-496 (1998); Wang et al., Gene Ther. 4:432-441 (1997); Neering et al., Blood 88:1147-1155 (1996); and Rendahl et al., Nat. Biotechnol. 16:757-761 (1998)). These systems impart small molecule control on the expression of the target gene(s) of interest.


C. Pharmaceutical Formulations


When used for pharmaceutical purposes, the vectors used for gene therapy are formulated in a suitable buffer, which can be any pharmaceutically acceptable buffer, such as phosphate buffered saline or sodium phosphate/sodium sulfate, Tris buffer, glycine buffer, sterile water, and other buffers known to the ordinarily skilled artisan such as those described by Good et al. Biochemistry 5:467 (1966).


The compositions can additionally include a stabilizer, enhancer, or other pharmaceutically acceptable carriers or vehicles. A pharmaceutically acceptable carrier can contain a physiologically acceptable compound that acts, for example, to stabilize the nucleic acids of the invention and any associated vector. A physiologically acceptable compound can include, for example, carbohydrates, such as glucose, sucrose or dextrans; antioxidants, such as ascorbic acid or glutathione; chelating agents; low molecular weight proteins or other stabilizers or excipients. Other physiologically acceptable compounds include wetting agents, emulsifying agents, dispersing agents, or preservatives, which are particularly useful for preventing the growth or action of microorganisms. Various preservatives are well known and include, for example, phenol and ascorbic acid. Examples of carriers, stabilizers, or adjuvants can be found in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 17th ed. (1985).


D. Administration of Formulations


The formulations of the invention can be delivered to any tissue or organ using any delivery method known to the ordinarily skilled artisan. In some embodiments of the invention, the nucleic acids of the invention are formulated in mucosal, topical, and/or buccal formulations, particularly mucoadhesive gel and topical gel formulations. Exemplary permeation enhancing compositions, polymer matrices, and mucoadhesive gel preparations for transdermal delivery are disclosed in U.S. Pat. No. 5,346,701.


E. Methods of Treatment


The gene therapy formulations of the invention are typically administered to a cell. The cell can be provided as part of a tissue, such as an epithelial membrane, or as an isolated cell, such as in tissue culture. The cell can be provided in vivo, ex vivo, or in vitro.


The formulations can be introduced into the tissue of interest in vivo or ex vivo by a variety of methods. In some embodiments of the invention, the nucleic acids of the invention are introduced into cells by such methods as microinjection, calcium phosphate precipitation, liposome fusion, or biolistics. In further embodiments, the nucleic acids are taken up directly by the tissue of interest.


In some embodiments of the invention, the nucleic acids of the invention are administered ex vivo to cells or tissues explanted from a patient, then returned to the patient. Examples of ex vivo administration of therapeutic gene constructs include Nolta et al., Proc Natl. Acad. Sci. USA 93(6):2414-9 (1996); Koc et al., Seminars in Oncology 23 (1):46-65 (1996); Raper et al., Annals of Surgery 223(2):116-26 (1996); Dalesandro et al., J. Thorac. Cardi. Surg., 11(2):416-22 (1996); and Makarov et al., Proc. Natl. Acad. Sci. USA 93(1):402-6 (1996).


X. Diagnosis of Mood and Psychotic Disorders


The present invention also provides methods of diagnosing mood disorders (such as major depression or bipolar disorder), psychotic disorders (such as schizophrenia). In one preferred embodiment, the disease state encompasses psychotic disorders. Diagnosis involves determining the level of a polypeptide or polynucleotide of the invention in a patient and then comparing the level to a baseline or range. Typically, the baseline value is representative of a polypeptide or polynucleotide of the invention in a healthy person not suffering from a mood disorder or psychotic disorder or under the effects of medication or other drugs. Variation of levels of a polypeptide or polynucleotide of the invention from the baseline range (either up or down) indicates that the patient has a mood disorder or psychotic disorder or at risk of developing at least some aspects of a mood disorder or psychotic disorder. In some embodiments, the level of a polypeptide or polynucleotide of the invention are measured by taking a blood, urine or tissue sample from a patient and measuring the amount of a polypeptide or polynucleotide of the invention in the sample using any number of detection methods, such as those discussed herein, e.g., detection of expression levels or SNPs or haplotypes associated with these genes. The genes provided herein also can be used to develop probe sets for PCR and chip assays.


Single nucleotide polymorphism (SNP) analysis is also useful for detecting differences between alleles of the polynucleotides (e.g., genes) of the invention. SNPs linked to genes encoding polypeptides of the invention are useful, for instance, for diagnosis of diseases (e.g., mood disorders such as bipolar disease, major depression, and schizophrenia disorders) whose occurrence is linked to the gene sequences of the invention. For example, if an individual carries at least one SNP linked to a disease-associated allele of the gene sequences of the invention, the individual is likely predisposed for one or more of those diseases. If the individual is homozygous for a disease-linked SNP, the individual is particularly predisposed for occurrence of that disease. In some embodiments, the SNP associated with the gene sequences of the invention is located within 300,000; 200,000; 100,000; 75,000; 50,000; or 10,000 base pairs from the gene sequence.


Various real-time PCR methods can be used to detect SNPs, including, e.g., Taqman or molecular beacon-based assays (e.g., U.S. Pat. Nos. 5,210,015; 5,487,972; Tyagi et al., Nature Biotechnology 14:303 (1996); and PCT WO 95/13399) are useful to monitor for the presence of absence of a SNP. Additional SNP detection methods include, e.g., DNA sequencing, sequencing by hybridization, dot blotting, oligonucleotide array (DNA Chip) hybridization analysis, or are described in, e.g., U.S. Pat. No. 6,177,249; Landegren et al., Genome Research, 8:769-776 (1998); Botstein et al., Am J Human Genetics 32:314-331 (1980); Meyers et al., Methods in Enzymology 155:501-527 (1987); Keen et al., Trends in Genetics 7:5 (1991); Myers et al., Science 230:1242-1246 (1985); and Kwok et al., Genomics 23:138-144 (1994).


In some embodiments, the level of the enzymatic product of a polypeptide or polynucleotide of the invention is measured and compared to a baseline value of a healthy person or persons. Modulated levels of the product compared to the baseline indicates that the patient has a mood disorder or psychotic disorder or is at risk of developing at least some aspects of a mood disorder or psychotic disorder. Patient samples, for example, can be blood, PBS, lymphocytes, saliva, CSF, urine or tissue samples.


Immunoassays using antigens and antibodies for genes differentially expressed in psychotic disorders are also useful for immunoassays such as ELISA and immunohistochemical assays. The genes described herein are also useful for making differential diagnoses for psychiatric disorders.


In some embodiments, schizophrenia in a patient may be diagnosed or otherwise evaluated by visualizing expression in situ of one or more of the gene sequences in Tables 1-14. Those skilled in the art of visualizing the presence or expression of molecules including nucleic acids, polypeptides and other biochemicals in the brains of living patients will appreciate that the gene expression information described herein may be utilized in the context of a variety of visualization methods. Such methods include, but are not limited to, single-photon emission-computed tomography (SPECT) and positron-emitting tomography (PET) methods. See, e.g., Vassaux and Groot-wassink, “In Vivo Noninvasive Imaging for Gene Therapy,” J. Biomedicine and Biotechnology, 2: 92-101 (2003); Turner, J., Smyth, P., Fallon, J. F., Kennedy, J. L., Potkin, S. G., FIRST BIRN (2006). Imaging and genetics in schizophrenia. Neuroinformatics, in press.


PET and SPECT imaging shows the chemical functioning of organs and tissues, while other imaging techniques—such as X-ray, CT and MRI—show structure. The use of PET and SPECT imaging is useful for qualifying and monitoring the development of brain diseases, including schizophrenia and related disorders. In some instances, the use of PET or SPECT imaging allows diseases to be detected years earlier than the onset of symptoms. The use of small molecules for labelling and visualizing the presence or expression of polypeptides and nucleotides has had success, for example, in visualizing proteins in the brains of Alzheimer's patients, as described by, e.g., Herholz K et al., Mol Imaging Biol., 6(4):239-69 (2004); Nordberg A, Lancet Neurol., 3(9):519-27 (2004); Neuropsychol Rev., Zakzanis K K et al., 13(1):1-18 (2003); Kung M P et al, Brain Res., 1025(1-2):98-105 (2004); and Herholz K, Ann Nucl Med., 17(2):79-89 (2003).


The dysregulated genes disclosed in Tables 1-14, or their encoded peptides (if any), or fragments thereof, can be used in the context of PET and SPECT imaging applications. After modification with appropriate tracer residues for PET or SPECT applications, molecules which interact or bind with the transcripts in Tables 1-14 or with any polypeptides encoded by those transcripts may be used to visualize the patterns of gene expression and facilitate diagnosis of schizophrenia as described herein. Similarly, if the encoded polypeptides encode enzymes, labeled molecules which interact with the products of catalysis by the enzyme may be used for the in vivo imaging and diagnostic application described herein.


Antisense technology is particularly suitable for detecting the the transcripts identified in Tables 1-14 herein. For example, the use of antisense peptide nucleic acid (PNA) labeled with an appropriate radionuclide, such as 111In, and conjugated to a brain drug-targeting system to enable transport across biologic membrane barriers, has been demonstrated to allow imaging of endogenous gene expression in brain cancer. See Suzuki et al., Journal of Nuclear Medicine, 10:1766-1775 (2004). Suzuki et al. utilize a delivery system comprising monoclonal antibodies that target transferring receptors at the blood-brain barrier and facilitate transport of the PNA across that barrier. Modified embodiments of this technique may be used to target upregulated genes associated with schizophrenia, such as the upregulated genes which appear in Tables 1-5, in methods of treating schizophrenic patients.


In other embodiments, the dysregulated genes listed in Tables 1-14 may be used in the context of prenatal and neonatal diagnostic methods. For example, fetal or neonatal lymphocytes can be isolated and the expression levels of appropriate transcripts (e.g., the transcripts in Tables 4-5) may be measured and correlated with the presence or increased likelihood of a mental disorder, e.g., schizophrenia. Similarly, the presence of one or more of the SNPs identified in Table 6 may be used to infer or corroborate dysregulated expression of ASG and the likelihood of schizophrenia in prenatal, neonatal, children and adult patients.


In other embodiments, the brain labeling and imaging techniques described herein or variants thereof may be used in conjunction with any of the dysregulated gene sequences in Tables 1-4 or 6 in a forensic analysis, i.e., to determine whether deceased individual suffered from schizophrenia. Similarly, forensic examination of lymphocyte expression of any of the genes identified in Tables 4-5 may be used alone or in conjunction with other methods to determine whether a deceased individual suffered from schizophrenia.


It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims.


EXAMPLES
Example 1
Identification of Genes Dysregulated in Schizophrenic Patients

Post mortem mental disorder brains (i.e., from schizophrenia patients) and control brains were used in this study. Each brain pair (case and control) was matched on the basis of gender, age, and postmortem interval. The patient's particular conditions in their terminal phase (agonal factors, e.g., seizure, coma, hypoxia, dehydration, and pyrexia) and the conditions of the brain tissue after death (postmortem factors, e.g., postmortem interval, and freezer interval) are two major influences on RNA preservation in postmortem brain tissue. Brain pH has been evaluated as an indicator for agonal status, and as an indicator of RNA preservation. Subjects with agonal factors and low pH samples, in which RNA quality was found to be compromised were eliminated from the study.


In this study, dysregulation of gene expression was studied in six brain regions: the anterior cingulate cortex (AnCg), dorsolateral prefrontal cortex (DLPFC), cerebellar cortex (CB), superior temporal gyrus (STG), parietal cortex (PC), and nucleus accumbens (nAcc). Gene expression patterns in schizophrenic patients versus healthy controls were analyzed by using Affymetrix GeneChips {(HG_U133 set (A+B)} to interrogate the transcriptome from human postmortem brains that met the strict quality control criteria referred to above, i.e., the agonal factor index of the donors was 0.0 (zero), and the pH of each individual brain was at least 6.4 (see Tomita et al., 2004; Li et al., 2004).


The probe set sequences used in data analysis are as defined in the CDF files created by the UMICH bioinformatics group (http://brainarray.mhri.med.umich.edu/Brainarray/) eliminating sequence redundancy that is inherent to UniGene definitions. The GeneChip hybridization data were processed, background-corrected, and normalized first with GCRMA using diagnosis (SZ, and control) and site {(University of California at Irvine (UCI), University of Michigan (UM) and University of California at Davis (UCD)} as factors in the analysis of sample replicates. The probe sets that showed (i) statistical significance of p<0.05; (ii) an at least 1.2-fold change (FC) in expression in cases relative to controls in either direction; and (iii) Present (P) call of at least 10% in any given brain region out of the six analyzed (listed above) were selected as differentially expressed, i.e., dysregulated genes.


A total of 1336 transcripts/genes were identified as differentially expressed in schizophrenic versus control brains in one or more of the six brain regions analyzed. Of those, 246 genes were upregulated in AnCg; 166 in CB; 156 in DLPFC; 124 in nAcc; 76 in PC; and 84 in STG. 138 genes were downregulated in AnCg; 187 in CB; 94 in DLPFC; 83 in nAcc; 125 in PC; and 105 in STG. This data is presented in Table 1.


Out of the six regions analyzed, only AnCg and DLPFC showed probe sets with Gene Ontology (GO) term enrichments and no significant enrichment of KEGG pathways. GO terms enriched in AnCg are listed in Table 2. Table 2 shows that 3 GO terms were enriched in AnCg, specifically: GO:0050874, organismal physiological process (with 11 probe sets); GO:0058550, eukaryotic translation factor 2 complex (with 3 probe sets); and GO:0005739, mitochondrion (with 25 probe sets).


Table 3 shows that a single GO term was enriched in DLPFC, specifically: GO:0005622 intracellular (with 90 probe sets).


Example 2
Peripheral Biomarker Expression of Dysregulated Genes Found in Brain

For this study, a separate cohort of individuals with schizophrenia (n=5) were matched for gender and age to unaffected (n=5) members of a pedigree. Freshly isolated lymphocytes from each individual were transformed using the Epstein-Barr Virus and grown until confluent in RPMI-1640 media supplemented with 15% fetal bovine serum (heat-inactivated), 2 mM L-glutamine and 25 mg of gentamicin. RNA was extracted from ˜5×107 lymphoblastic cells using the standard TRIzol isolation protocol (Invitrogen, Carlsbad, Calif.). Affymetrix Human Genome U133A Arrays were used for gene expression according to the manufacturer's protocol. The gene expression traits were derived from the U133A chips and analyzed by robust multiarray condensation algorithm (RMA). Differential gene expression (gene expression trait for the purpose of this analysis) was defined as a gene that displayed a significant two-tailed t-test (p<0.05) in schizophrenia versus unaffected family members. There were 1344 genes that passed the t-test for dysregulation in lymphocytes in schizophrenia compared to unaffecteds. This list was compared to genes in Table 1 (showing brain dysregulated genes in schizophrenia). The genes that were dysregulated in both brain and lymphocytes are shown in Table 4.


The list of 84 dysregulated genes in Table 4 may be grouped into those genes that agree in direction between brain and lymphoblasts, and those genes that disagree in direction between brain and lymphoblasts. Both dysregulated gene sets are biomarkers. The lymphoblasts do not have agonal factors, pH, or medication effects such as commonly seen in brain tissue. Thus, the gene transcripts in Table 4 may be used for monitoring lymphoblasts during treatment or for diagnostic purposes.


The subset of the 1344 genes identified by microarray analysis as significantly dysregulated in lymphoblasts only (i.e., not brains) is shown in Table 5. The microarray data was validated using Q-PCR to determine the fold change and direction of gene expression in the lymphocyte samples. Eight of these genes meet statistical significance in Q-PCR by t-test (two-tailed).


Aspartylglucosaminuria (AGA) gene expression is dysregulated in both the brain and lymphocytes of individuals with schizophrenia (Table 4). Eleven single nucleotide polymorphic markers were identified which correlate with AGA gene expression are shown in Table 6. Table 6 also shows the regression p-values of genotype with lymphocyte gene expression. Of the 11 markers, 8 are associated with a cis-regulatory site (i.e., the Cis value is less then 5 Mb) and 3 are related to a trans-regulatory site (the Cis value is greater than 5 Mb). Detecting these SNPs can facilitate the prediction of AGA gene expression in lymphoblasts. Similarly, detecting SNPs correlated with the expression of other dysregulated genes can facilitate the prediction of expression levels of those genes. The SNPs in Table 6 also represent targets for controlling expression of AGA, for diagnosing and treating schizophrenia, or for diagnosing and treating other disorders associated with altered AGA expression. For SNPs rs723820, rs723819, rs1112286, rs1375749, Table 6 shows that the minor alleles are associated with the decreased AGA expression in schizophrenia.


Example 3
Validation of PSPHL Insertion Deletion Mutation

The present invention extended the previous findings regarding the insertion-deletion polymorphism of phosphoserine phosphatase-like gene, and the association between deletion allele of PSPHL and susceptibility to bipolar disorder (BPD).


We previously determined 1) PSPHL gene consists of 4 exons. Exons 1, 2, 3 and 4 are 213 bp, 114 bp, 122 bp and 501 bp, in length, respectively, and span introns 1, 2 and 3 (3221 bp, 829 bp and 11939 bp, in length, respectively). 2) PSPHL and PSPH are highly homologous, which locate 200 kb apart from each other on chromosome 7p 11.2 region. 3) PSPHL gene has two alternative transcripts, one of which utilizes the exons 1-4 (PSPHL-A), while another utilizes the exons 1, 2 and 4 (PSPHL-B). Predicted proteins of PSPHL-A and PSPHL-B share N-terminal 57 common amino acids, transcribed from exons 1 and 2. PSPH and the predicted PSPHL-A&B have 31 amino acids in common. 4) There locates an insertion/deletion polymorphism at the PSPHL locus. The deleted genomic region spans more than 30 kb, including the promoter region and the exons 1, 2 and 3 of PSPHL gene. 5) PSPHL shows a dichotomous (present or absent) pattern of expression among human population, which may due to the insertion/deletion polymorphism at the PSPHL locus. 6) Number of individuals who expresses PSPHL was significantly smaller in BPD patient group compared to control group. 7) Since PSPH is the rate limiting enzyme for serine synthesis, PSPHL may be involved in serine amino acid metabolic pathway, but might be involved in other pathways.


In the present invention, we further determined the followings:


1) We verified involvement of the insertion-deletion polymorphism at the PSPHL locus on the expression pattern of the gene.


Among the 125 human postmortem brain tissues (19 BPD, 22 MDD, 20 SCZ patients and 64 controls) analyzed regarding the PSPHL genotype and PSPHL mRNA expression, 81 subjects (18 BPD, 8 MDD, 12 SCZ, and 43 Controls) showed homozygous pattern of the deletion allele (Del/Del) for the PSPHL locus, and all of the 81 Del/Del individuals lacked PSPHL mRNA expression. On the other hand, 40 subjects (1 BPD, 13 MDD, 8 SCZ, and 18 Controls) showed heterozygous pattern of the insertion and deletion alleles (Ins/Del) for the PSPHL locus, and 4 subjects (1 MDD and 3 Controls) showed homozygous pattern of the insertion allele (Ins/Ins). All of the 44 subjects (40 Ins/Del and 4 Ins/Ins), which have at least one insertion allele, showed PSPHL mRNA expression. This findings support that the presence/absence of PSPHL mRNA expression is due to the insertion/deletion polymorphism at the PSPHL locus. Our observations on the 64 control subjects exactly matched Hardy Weinberg expectations. Allele frequencies for the insertion and deletion alleles for the PSPHL locus were estimated 0.18 and 0.82, respectively.


2) We verified that number of individuals who expresses PSPHL was significantly smaller in BPD patient group compared to control group, and significantly larger in MDD patient group compared to the control group.


Based on hypergeometric distribution, the cumulative p value for observing 1 or less PSPHL non-expressed individuals in the 19 BPD patients is 0.0015. Also, the cumulative p value for observing 14 or more PSPHL-expressed individuals in the 22 MDD patients is 0.0002. There is no significant difference in the distribution between SCZ patients and controls. It is noteworthy that PSPHL non-expressed individuals are predominant in the BPD subjects, whereas PSPHL-expressed individuals are predominant in the MDD subjects. The probability of the observed difference in the distribution between BPD and MDD is 0.000098 based on the Fisher's exact test. These findings could be applicable for genetic testing to predict potential BPD patients among depressed patients who come to see physicians in their early stage of the chronic illnesses.


We characterized that PSPHL-B mRNA expression level was also about 10 times higher than PSPHL-A in human postmortem brain tissue and cell lines derived from human brain.


PSPHL has at least two alternative transcripts; PSPHL-A (consists of the exons 1, 2, 3 and 4) and PSPHL-B (consists of the exons 1, 2 and 4). Based on quantitative RT-PCR evaluation with primer sets and TaqMan probes specific to PSPHL-A and PSPHL-B, both PSPHL-A and PSPHL-B were expressed in human postmortem brain cortices, including anterior cingulate and cerebellar cortices, from subjects which have at least one PSPHL insertion allele. PSPHL-B mRNA expression level was about 10 times higher than PSPHL-A in the brain tissues analyzed. Also, both PSPHL-A and PSPHL-B were expressed in cell lines derived from human brain, including human neuroblastoma cell lines, SK-N-SH, human glioma cell line, Hs 683, and human oligodendrocyte-derived cell line, OL, which have at least one PSPHL insertion allele. PSPHL-B mRNA expression level was also about 10 times higher than PSPHL-A in these cell lines. The glioblastoma cell line, U87-MG, which has homozygous PSPHL deletion allele, lack the expression of PSPHL-A and PSPHL-B.


4) We verified promoter activity of 5′ region of the PSPHL gene. The 5′-region of PSPHL (1015 bp fragment) was cloned into pGL-basic vector show sufficient promoter activity at least in the Hela cells and human oligodendrocyte cell line, OL. The vector contains the same region in opposite direction (negative control) did not show promoter activity.

PSPHL locusPSPHL mRNADi-genotypeexpressionagnosisTotalDel/DelDel/InsIns/InsNo ExpressionExpressedBPD191810181MDD228131814SCZ201280128Control64431834321



embedded image


The above examples are provided to illustrate the invention but not to limit its scope. Other variants of the invention will be readily apparent to one of ordinary skill in the art and are encompassed by the appended claims. All publications, databases, Genbank sequences, GO terms, patents, and patent applications cited herein are hereby incorporated by reference.

TABLE 1AnnotationDirection of Change in SZ relative to ControlsUniGene IDAccSymbolChr.AnCgCBDLPFCNaccPCSTGNameHs.444558AA007604KHDRBS38upKH domain containing, RNA binding, signal transduction associated 3Hs.101139AA01725719downTranscribed locusHs.443049AA2517737downTranscribed locusHs.192788AA314461LOC3896778upSimilar to RIKEN cDNA 3000004N20Hs.485557AA418695GSTA46downdownGlutathione S-transferase A4Hs.535419AA44768118upupHypothetical LOC388459Hs.36958AA496799BCAR31downBreast cancer anti-estrogen resistance 3Hs.4204AA7004401downCDNA FLJ30779 fis, clone FEBRA2000815Hs.241548AA743526RASA23upRAS p21 protein activator 2Hs.524367AA779991CBARA110downCalcium binding atopy-related autoantigen 1Hs.532987AA827062C18orf2218downChromosome 18 open reading frame 22Hs.136905AB002310UREB1downUpstream regulatory element binding protein 1Hs.114169AB007876LRRTM25upLeucine rich repeat transmembrane neuronal 2Hs.435557AB011103KIF5C2upKinesin family member 5CHs.383564AB011146KIAA057415upKIAA0574 proteinHs.146007AB011154KIAA05822upKIAA0582Hs.118140AB018259DOCK47downDedicator of cytokinesis 4Hs.153610AB018294RIMS28upRegulating synaptic membrane exocytosis 2Hs.178471AB018341ZNF43219downdownZinc finger protein 432Hs.466261AB021644ZNF1419downZinc finger protein 14 (KOX 6)Hs.303454AB023136SEC15L22downSEC15-like 2 (S. cerevisiae)Hs.298658AB024523KLF34upKruppel-like factor 3 (basic)Hs.476399AB029028RAP1403downdownRetinoblastoma-associated protein 140Hs.132813AB029033IQSEC312downIQ motif and Sec7 domain 3Hs.12264AB032989KIAA1163downdownAmphoterin-induced geneHs.50823AB033060PDCD65downProgrammed cell death 6Hs.472285AB033098KIAA127220downKIAA1272 proteinHs.537101AB033338downKRMP1 mRNA for mitotic kinesin-related protein, partial cds, alternative exon sequence.Hs.140903AB037744MIB118upMindbomb homolog 1 (Drosophila)Hs.3346AB037811FLJ112801upHypothetical protein FLJ11280Hs.377588AB040883KIAA14504upKIAA1450 proteinHs.287362AB046767TLE315upTransducin-like enhancer of split 3 (E(sp1) homolog,Drosophila)Hs.13305AB046788ROBO23upRoundabout axon guidance receptor, homolog 2(Drosophila)Hs.474914AB051446RUTBC322downRUN and TBC1 domain containing 3Hs.510745AB097009HNLF7downPutative NFkB activating protein HNLFHs.523789AF001893TncRNA11downTrophoblast-derived noncoding RNAHs.352614AF007155LOC25453115downPLSC domain containing proteinHs.147770AF012023ITGB1BP12upIntegrin beta 1 binding protein 1Hs.42400AF022789USP1213upUbiquitin specific protease 12Hs.465784AF026030TIMM4419upTranslocase of inner mitochondrial membrane 44 homolog(yeast)Hs.12451AF035276EML114upEchinoderm microtubule associated protein like 1Hs.12473AF0521091upCDNA clone IMAGE: 5260262, partial cdsHs.13438AF0521414upClone 24626 mRNA sequenceHs.368046AF054993SNAP916upSynaptosomal-associated protein, 91 kDa homolog(mouse)Hs.129997AF07053413upClones 24632 and 24634 mRNA sequenceHs.48372AF0860924downFull length insert cDNA clone YZ87G11Hs.513509AF086220FLJ3213016downHypothetical protein FLJ32130Hs.131133AF087875PRKAG27upProtein kinase, AMP-activated, gamma 2 non-catalyticsubunitHs.34560AF116668LMO211downdownLIM domain only 2 (rhombotin-like 1)Hs.142245AF126163HHLA31upHERV-H LTR-associating 3Hs.446240AF144233PRKCBP120downProtein kinase C binding protein 1Hs.47261AF260704SLCO1C112downdownSolute carrier organic anion transporter family, member1C1Hs.371788AF318362DKFZP547E10101upDKFZP547E1010 proteinHs.125747AF38117217upAA02 pseudogene mRNA, partial sequence, mRNAsequenceHs.187946AI2405382downdownHypothetical gene supported by AK124342Hs.23187AI2424767downTranscribed locusHs.328801AI3088988downTranscribed locusHs.542164AI41715719upCDNA FLJ41369 fis, clone BRCAN2006117Hs.421200AI4800145downupClone 24571 mRNA sequenceHs.445066AI499801GRIN2B12downGlutamate receptor, ionotropic, N-methyl D-aspartate 2BHs.47995AI61067610downFull length cDNA clone CS0DC007YK10 ofNeuroblastoma Cot 25-normalized of Homo sapiens(human)Hs.503584AI638679PANX111upPannexin 1Hs.174746AI6709923downTranscribed locusHs.440729AI6718493downTranscribed locusHs.404218AI6746448downTranscribed locusHs.530218AI6798058downTranscribed locusHs.537738AI69288216upFull length insert cDNA clone ZD81C11Hs.513356AI69454416updownTranscribed locus, moderately similar to XP_498452.1 hypothetical gene supported by NM_173697[Homo sapiens]Hs.356084AI703476GPR273upupG protein-coupled receptor 27Hs.201106AI7589461upTranscribed locusHs.192788AI768185LOC3896778upSimilar to RIKEN cDNA 3000004N20Hs.202054AI796835downTranscribed locusHs.536522AI822096SMAD4upSMAD, mothers against DPP homolog 4 (Drosophila)Hs.97104AI8238795upTranscribed locusHs.94949AI934339MCEE2upMethylmalonyl CoA epimeraseHs.142869AI9355864upTranscribed locusHs.459255AI935701NTRK315upNeurotrophic tyrosine kinase, receptor, type 3Hs.114033AJ420492SSR16downSignal sequence receptor, alpha (translocon-associatedprotein alpha)Hs.445098AK000490DEPDC11upDEP domain containing 1Hs.517134AK000586C20orf4320upChromosome 20 open reading frame 43Hs.288995AK000820ZNF58719downdownZinc finger protein 587Hs.471221AK000969KLF72upupKruppel-like factor 7 (ubiquitous)Hs.301943AK001249KIAA04671downKIAA0467 proteinHs.144055AK001563SBNO112upupSno, strawberry notch homolog 1 (Drosophila)Hs.27021AK001697RIOK25downdownRIO kinase 2 (yeast)Hs.443260AK001776C20orf2020downdowndowndownChromosome 20 open reading frame 20Hs.516311AK0018562downdownHomo sapiens, clone IMAGE: 4826696, mRNAHs.92308AK002085LOC14443812upHypothetical protein LOC144438Hs.477693AK021677NCK13downNCK adaptor protein 1Hs.481819AK021922PDZK35downPDZ domain containing 3Hs.476982AK022155CPOX3upCoproporphyrinogen oxidaseHs.492445AK022204EDD8downE3 identified by differential displayHs.31431AK022233FN3KRP17; 20; 9upFructosamine-3-kinase-related proteinHs.285782AK022630LRRTM42upupLeucine rich repeat transmembrane neuronal 4Hs.132794AK022741PCYT1BupPhosphate cytidylyltransferase 1, choline, beta isoformHs.321653AK022832FLJ127701upHypothetical protein FLJ12770Hs.167165AK023037FLJ1297512downdownHypothetical protein FLJ12975Hs.440833AK023692PKN21downProtein kinase N2Hs.274422AK024220C20orf2720upChromosome 20 open reading frame 27Hs.473374AK025196PTPRK6downProtein tyrosine phosphatase, receptor type, KHs.431081AK025233USP534downUbiquitin specific protease 53Hs.499483AK025626LOC8369316upSteroid dehydrogenase-likeHs.465295AK025773LMAN118upLectin, mannose-binding, 1Hs.289092AK026033COTL116; 11upCoactosin-like 1 (Dictyostelium)Hs.537428AK026035downCDNA FLJ34018 fis, clone FCBBF2002801Hs.426324AK026149TUSC38upTumor suppressor candidate 3Hs.272759AK026156PITPNM2upPhosphatidylinositol transfer protein, membrane-associated 2Hs.535771AK026466CYFIP25downCytoplasmic FMR1 interacting protein 2Hs.524341AK026495SLC2A1312upSolute carrier family 2 (facilitated glucose transporter),member 13Hs.79881AK0266593downCDNA: FLJ23006 fis, clone LNG00414Hs.91521AK026748DKFZP761M15115upupHypothetical protein DKFZP761M1511Hs.301296AK02678413downCDNA: FLJ23131 fis, clone LNG08502Hs.415842AK026870RBM189upRNA binding motif protein 18Hs.292575AK026980ZNF37B10; 7; 1downZinc finger protein 37b (KOX 21)Hs.111286AK027059MRPS1115upupMitochondrial ribosomal protein S11Hs.130692AK027273MGC1094612downHypothetical protein MGC10946Hs.452398AK0553022downMRNA; cDNA DKFZp564E143 (from cloneDKFZp564E143)Hs.532786AK055378LOC33928717upHypothetical protein LOC339287Hs.148105AK055479PRICKLE23upPrickle-like 2 (Drosophila)Hs.536251AK056079JAM221downJunctional adhesion molecule 2Hs.47918AK056549CNKSR2XupConnector enhancer of kinase suppressor of Ras 2Hs.193115AK057990KIAA17648downKIAA1764 proteinHs.251399AK074730HRH320upHistamine receptor H3Hs.254414AK090803SRrp356upSerine-arginine repressor protein (35 kDa)Hs.242947AK091081DGKI7upDiacylglycerol kinase, iotaHs.443731AK091775USP815upUbiquitin specific protease 8Hs.494804AK091948LTB4DH9upLeukotriene B4 12-hydroxydehydrogenaseHs.179153AK0921459upCDNA FLJ34826 fis, clone NT2NE2008803Hs.177275AK092235ANKRD66upAnkyrin repeat domain 6Hs.433956AK0927112upHypothetical LOC400944Hs.144447AK092984WDR1110downWD repeat domain 11Hs.293077AK093067CHPT112upCholine phosphotransferase 1Hs.523467AK093229NRIP311upNuclear receptor interacting protein 3Hs.465462AK09358818upCDNA FLJ36269 fis, clone THYMU2003012Hs.12248AK093871CXXC44upupCXXC finger 4Hs.451846AK093939RGPR1downRegucalcin gene promoter region related proteinHs.331667AK094282MGC32001downHypothetical protein LOC284615Hs.22654AK094487SCN1A2upSodium channel, voltage-gated, type I, alphaHs.26479AK0945353updownCDNA FLJ37216 fis, clone BRALZ2008696Hs.438695AK094876FKBP1112downFK506 binding protein 11, 19 kDaHs.380250AK094968IFI161downInterferon, gamma-inducible protein 16Hs.466987AK095884PRKD219downProtein kinase D2Hs.128686AK097398NUCB211upNucleobindin 2Hs.169378AK098775MPDZ9downdownMultiple PDZ domain proteinHs.494312AK123824NTRK29upNeurotrophic tyrosine kinase, receptor, type 2Hs.525589AK123878MEG314upMaternally expressed 3Hs.164649AK124629IXL19downIntersex-like (Drosophila)Hs.476782AK126999EIF4E33upupEukaryotic translation initiation factor 4E member 3Hs.297044AK127019RUFY210downRUN and FYVE domain containing 2Hs.256801AK127476ZNF493upZinc finger protein 493Hs.520804AK1280347downSimilar to cell division cycle 100 homologHs.435767AK129838PCYT1A3downPhosphate cytidylyltransferase 1, choline, alpha isoformHs.126914AK130263KIAA14304upKIAA1430Hs.512181AK130506CXorf33downChromosome X open reading frame 33Hs.480825AK130520RNF1504upRing finger protein 150Hs.184216AL049980DKFZP564C15211downDKFZP564C152 proteinHs.155090AL117471GNB515upGuanine nucleotide binding protein (G protein), beta 5Hs.421907AL122063GLTSCR2downGlioma tumor suppressor candidate region gene 2Hs.158688AL133563EIF5B2downEukaryotic translation initiation factor 5BHs.435700AL137537ATP8B21upATPase, Class I, type 8B, member 2Hs.478125AL137692INADL1upInaD-like proteinHs.26815AL360202THAP1015upTHAP domain containing 10Hs.475103AL389951NUP5022upNucleoporin 50 kDaHs.536940AL512705APEG12downAortic preferentially expressed protein 1Hs.292549AL831922DLG13updownDiscs, large homolog 1 (Drosophila)Hs.268675AL831995MEF2A15upupMADS box transcription enhancer factor 2, polypeptide A (myocyte enhancer factor 2A)Hs.518099AL832398MGC267173downupHypothetical protein MGC26717Hs.30141AL832603FLJ2031315downHypothetical protein FLJ20313Hs.175343AL832699PIK3C2A11downPhosphoinositide-3-kinase, class 2, alpha polypeptideHs.490790AL833137THAP57downTHAP domain containing 5Hs.430300AL83336011downMRNA; cDNA DKFZp667E0114 (from cloneDKFZp667E0114)Hs.477921AL833852WWTR13downWW domain containing transcription regulator 1Hs.282855AL834302FLJI299415upHypothetical protein FLJ12994Hs.32468AV7185185upTranscribed locusHs.163924AW080999NR3C24downNuclear receptor subfamily 3, group C, member 2Hs.418198AW086065PAPD45downdownPAP associated domain containing 4Hs.446340AW291402upTranscribed locusHs.293379AW30242219upTranscribed locusHs.429AW438709ATP5G32downATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (subunit 9) isoform 3Hs.408427AW451792COMMD720upCOMM domain containing 7Hs.436556AW5717397downTranscribed locusHs.257786AW9642412upTranscribed locusHs.306423BC000825LOC3395241downHypothetical protein LOC339524Hs.434953BC001063HMGB24upHigh mobility group box 2Hs.535659BC002458MCM3AP21upMCM3 minichromosome maintenance deficient 3 (S. cerevisiae)associated proteinHs.483239BC002515ALDH7A15updownAldehyde dehydrogenase 7 family, member A1Hs.7001BC002867C14orf9upChromosome 14 open reading frame 9Hs.224282BC004887LANCL27upLanC lantibiotic synthetase component C-like 2 (bacterial)Hs.471582BC004921LOC933492downHypothetical protein BC004921Hs.516859BC005095PANK220downPantothenate kinase 2 (Hallervorden-Spatz syndrome)Hs.153546BC005258CDC235upCDC23 (cell division cycle 23, yeast homolog)Hs.18788BC006283DHRS1019downDehydrogenase/reductase (SDR family) member 10Hs.443673BC008143KIAA100212; 1downKIAA1002 proteinHs.529630BC0086253downHomo sapiens, clone IMAGE: 4183899, mRNAHs.534483BC008630MGC294117upHypopthetical protein MGC2941Hs.516859BC008667PANK220downPantothenate kinase 2 (Hallervorden-Spatz syndrome)Hs.11923BC009674DJ167A19.11; 2upupHypothetical protein DJ167A19.1Hs.447579BC010538LOC33929018upHypothetical protein LOC339290Hs.103555BC011054FLJ1477517downHypothetical protein FLJ14775Hs.194408BC014227KIAA12446upKIAA1244Hs.50823BC014604PDCD65downProgrammed cell death 6Hs.129837BC015067ZBTB81updownZinc finger and BTB domain containing 8Hs.445113BC015910MARCH-II19upupMembrane-associated RING-CH protein IIHs.487325BC016285PRKACB1updownProtein kinase, cAMP-dependent, catalytic, betaHs.536470BC016735LOC6392922updownHypothetical protein LOC63929Hs.279908BC017788TFB1M6downTranscription factor B1, mitochondrialHs.517792BC019303C3orf103upupChromosome 3 open reading frame 10Hs.499142BC019602YME1L110downYME1-like 1 (S. cerevisiae)Hs.350268BC020516IRF2BP2upInterferon regulatory factor 2 binding protein 2Hs.197922BC020630CaMKIINalpha1upCalcium/calmodulin-dependent protein kinase IIHs.208961BC021961NSD15downNuclear receptor binding SET domain protein 1Hs.525588BC023543MEG314upMaternally expressed 3Hs.436568BC024272CD745downdownCD74 antigen (invariant polypeptide of majorhistocompatibility complex, class II antigen-associated)Hs.180933BC029922CXXC118upCXXC finger 1 (PHD domain)Hs.535810BC0301225upCDNA clone IMAGE: 4814828, partial cdsHs.74655BC033998LOC12451217upHypothetical protein LOC124512Hs.479099BC035257SORCS24upSortilin-related VPS10 domain containing receptor 2Hs.459070BC036099ARNT215downAryl-hydrocarbon receptor nuclear translocator 2Hs.302631BC0366227downCDNA clone IMAGE: 5286843, partial cdsHs.98132BC036875LCN69downLipocalin 10Hs.127951BC037195FLJ14503downHypothetical protein FLJ14503Hs.90242BC0378001upHomo sapiens, clone IMAGE: 4796172, mRNAHs.212151BC039075CLSTN312downCalsyntenin 3Hs.461074BC040486ZEP9016upZinc finger protein 90 homolog (mouse)Hs.516853BC041916C20orf19420upupChromosome 20 open reading frame 194Hs.506309BC041930EEA112downEarly endosome antigen 1, 162 kDHs.536567BC042073upData not foundHs.205865BC042754LOC143458downHypothetical protein LOC143458Hs.434418BC042833MYT1L2upMyelin transcription factor 1-likeHs.122110BC043568FLJ337184upHypothetical protein FLJ33718Hs.413416BC047331JMJD1C10downJumonji domain containing 1CHs.136888BC047477LOC33875812upHypothetical protein LOC338758Hs.443258BC051799SREBF222upSterol regulatory element binding transcription factor 2Hs.169182BC052964KIF21B1; 19;upKinesin family member 21B16Hs.509314BC059410LOC2851482downdownHypothetical protein LOC285148Hs.180714BC06452315E1.212downHypothetical protein 15E1.2Hs.519904BC065748RBM246downRNA binding motif protein 24Hs.459590BC066358CCL279downChemokine (C—C motif) ligand 27Hs.433150BC0678841upCDNA clone IMAGE: 4841343, partial cdsHs.506458BC068451EB-112upE2a-Pbx1-associated proteinHs.86508BC0798335downCDNA clone IMAGE: 4841343, partial cdsHs.12862BC080181RFNG17upRadical fringe homolog (Drosophila)Hs.115284BC080572ZNF21316upZinc finger protein 213Hs.143587BE467201downTranscribed locusHs.102471BE538923PHACTR26downPhosphatase and actin regulator 2Hs.127486BE5516242downTranscribed locusHs.21691BE671266GPR752downG protein-coupled receptor 75Hs.188594BE8893019upTranscribed locusHs.118526BF056604downTranscribed locusHs.225598BF0592096upTranscribed locusHs.144022BF109731FDFT18upFarnesyl-diphosphate farnesyltransferase 1Hs.464848BF378154B4GALT618upUDP-GAl: betaGlcNAc beta 1,4-galactosyltransferase,polypeptide 6Hs.432792BF439526CBLL17upCas-Br-M (murine) ecotropic retroviral transformingsequence-like 1Hs.478465BG707584FLJ127483upHypothetical protein FLJ12748Hs.434375BI820698PTPRB12downdownProtein tyrosine phosphatase, receptor type, BHs.414028BM455428C9orf1169; 3upChromosome 9 open reading frame 116Hs.469967BM557121upTranscribed locusHs.445247BM6824605downTranscribed locus, weakly similar to NP_703324.1Plasmodium falciparum 3D7 MAL1P3.06 geneHs.133469BM719738GOLGA19downGolgi autoantigen, glolgin subfamily a, 1Hs.479766BP225938TPARL4downTPA regulated locusHs.470882BQ002778CALCRL2downCalcitonin receptor-likeHs.118769BQ0035012downTranscribed locusHs.229304BQ007533downTranscribed locusHs.487648BQ285965SNX137downSorting nexin 13Hs.104980BQ331336upData not foundHs.479808BQ477415IGFBP74downdownInsulin-like growth factor binding protein 7Hs.179238BU674160LRRC68upLeucine rich repeat containing 6Hs.31903BU6857613downTranscribed locusHs.28199BX0914477downdownTranscribed locusHs.445105BX093081GRIN2DupGlutamate receptor, ionotropic, N-methyl D-aspartate 2DHs.7413BX0971903upupTranscribed locusHs.452398BX0997222downMRNA; cDNA DKFZp564E143 (from cloneDKFZp564E143)Hs.4817BX537377OPCML11upOpioid binding protein/cell adhesion molecule-likeHs.482363BX537394SLC30A55downSolute carrier family 30 (zinc transporter), member 5Hs.343522BX537745ATP2B41; 14upATPase, Ca++ transporting, plasma membrane 4Hs.479853BX537946EPHA54upupupEPH receptor A5Hs.370510BX538269IGSF411upImmunoglobulin superfamily, member 4Hs.192221BX538289ELL2upElongation factor, RNA polymerase II, 2Hs.494178BX647070RORB9upRAR-related orphan receptor BHs.433381BX647220C6orf896upChromosome 6 open reading frame 89Hs.310545BX647240SYT112upSynaptotagmin IHs.436142BX647689PTPN134upProtein tyrosine phosphatase, non-receptor type 13 (APO-1/CD95 (Fas)-associated phosphatase)Hs.369978BX647773GTDC12upGlycosyltransferase-like 1Hs.146542BX648027NEGR11upNeuronal growth regulator 1Hs.213050BX648050ELAVL41upELAV (embryonic lethal, abnormal vision, Drosophila)-like 4 (Hu antigen D)Hs.175343BX648778PIK3C2A11downdownPhosphoinositide-3-kinase, class 2, alpha polypeptideHs.306423CA440056LOC3395241upHypothetical protein LOC339524Hs.327736CA442378KIF5B10upKinesin family member 5BHs.537332CB996893CNOT6L4downCCR4-NOT transcription complex, subunit 6-likeHs.105636CD6397341downTranscribed locusHs.443031CK300949GLB13upGalactosidase, beta 1Hs.437611CK820590XupTranscribed locusHs.29802CN260580SLIT24downSlit homolog 2 (Drosophila)Hs.444818CR456854CGGBP13; 12downCGG triplet repeat binding protein 1Hs.500333CR596764C10orf5810upChromosome 10 open reading frame 58Hs.301296CR61682613downCDNA: FLJ23131 fis, clone LNG08502Hs.120446CR623819ZCWCC122downZinc finger, CW type with coiled-coil domain 1Hs.250072CR627428SLC4A73upSolute carrier family 4, sodium bicarbonate cotransporter,member 7Hs.477134CR749341DKFZP434F20213downDKFZP434F2021 proteinHs.118351D13635UBE3C7upUbiquitin protein ligase E3CHs.35804D25215HERC34; 8upHect domain and RLD 3Hs.423163D87969SLC35A16upSolute carrier family 35 (CMP-sialic acid transporter),member A1Hs.536256F01952GNAZ22downupGuanine nucleotide binding protein (G protein), alpha zpolypeptideHs.538896F02333ANKRD1013downAnkyrin repeat domain 10Hs.27996F100104downdownTranscribed locusHs.492212H95037DECR18up2,4-dienoyl CoA reductase 1, mitochondrialHs.131711J03620DLD7downDihydrolipoamide dehydrogenase (E3 component ofpyruvate dehydrogenase complex, 2-oxo-glutaratecomplex, branched chain keto acid dehydrogenasecomplex)Hs.449451K02885downIsolate 971.4_G01 T cell receptor beta (TCBRV)Hs.274873L06845CARS11upCysteinyl-tRNA synthetaseHs.220629L17000CAMK45upCalcium/calmodulin-dependent protein kinase IVHs.196983M61199SSFA22downSperm specific antigen 2Hs.212838NM_000014A2M12downdownAlpha-2-macroglobulinHs.232375NM_000019ACAT111upAcetyl-Coenzyme A acetyltransferase 1 (acetoacetylCoenzyme A thiolase)Hs.207776NM_000027AGA4downdownAspartylglucosaminidaseHs.445358NM_000042APOH17downupApolipoprotein H (beta-2-glycoprotein I)Hs.160786NM_000050ASS9upArgininosuccinate synthetaseHs.169348NM_000057BLM15downBloom syndromeHs.476218NM_000094COL7A13downCollagen, type VII, alpha 1 (epidermolysis bullosa,dystrophic, dominant and recessive)Hs.304682NM_000099CST320; 11;downCystatin C (amyloid angiopathy and cerebral hemorrhage)22; 12Hs.335513NM_000129F13A16downCoagulation factor XIII, A1 polypeptideHs.255230NM_000181GUSB7downGlucuronidase, betaHs.303154NM_000202IDSX; 12upIduronate 2-sulfatase (Hunter syndrome)Hs.224012NM_000214JAG120downdownJagged 1 (Alagille syndrome)Hs.156519NM_000251MSH22downMutS homolog 2, colon cancer, nonpolyposis type 1 (E. coli)Hs.21213NM_000259MYO5A15upupMyosin VA (heavy polypeptide 12, myoxin)Hs.181272NM_000297PKD24downdownPolycystic kidney disease 2 (autosomal dominant)Hs.476595NM_000333ATXN73downdowndownAtaxin 7Hs.271771NM_000345SNCA4upSynuclein, alpha (non A4 component of amyloidprecursor)Hs.370771NM_000389CDKN1A6; 9downCyclin-dependent kinase inhibitor 1A (p21, Cip1)Hs.2785NM_000422KRT1717downKeratin 17Hs.250769NM_000478ALPL1downAlkaline phosphatase, liver/bone/kidneyHs.24422NM_000538RFXAP13upRegulatory factor X-associated proteinHs.529400NM_000629IFNAR121upInterferon (alpha, beta and omega) receptor 1Hs.150749NM_000633BCL218upB-cell CLL/lymphoma 2Hs.336046NM_000640IL13RA2XupInterleukin 13 receptor, alpha 2Hs.150749NM_000657BCL218upB-cell CLL/lymphoma 2Hs.334707NM_000666ACY13; 1downdownAminoacylase 1Hs.197029NM_000675ADORA2A22upAdenosine A2a receptorHs.388004NM_000687AHCY20; 16upupS-adenosylhomocysteine hydrolaseHs.34114NM_000702ATP1A21downATPase, Na+/K+ transporting, alpha 2 (+) polypeptideHs.433307NM_000709BCKDHA19; 1downBranched chain keto acid dehydrogenase E1, alphapolypeptide (maple syrup urine disease)Hs.282871NM_000770CYP2C810upCytochrome P450, family 2, subfamily C, polypeptide 8Hs.152096NM_000775CYP2J21upCytochrome P450, family 2, subfamily J, polypeptide 2Hs.202354NM_000793DIO214downDeiodinase, iodothyronine, type IIHs.175934NM_000806GABRA15; 17upGamma-aminobutyric acid (GABA) A receptor, alpha 1Hs.473648NM_000819GART21upPhosphoribosylglycinamide formyltransferase,phosphoribosylglycinamide synthetase,phoshoribosylaminoimidazole synthetaseHs.32945NM_000838GRM16upGlutamate receptor, metabotropic 1Hs.268573NM_000853GSTT122downGlutathione S-transferase theta 1Hs.376933NM_000858GUK11upGuanylate kinase 1Hs.46732NM_000898MAOBXupupMonoamine oxidase BHs.2820NM_000916OXTR3upOxytocin receptorHs.354056NM_000941POR7upP450 (cytochrome) oxidoreductaseHs.446429NM_000954PTGDS9; 11; 1;downdownProstaglandin D2 synthase 21 kDa (brain)7; 6; 14;2; 19; 17;3; 16;22Hs.374588NM_000985RPL1718; 1;upRibosomal protein L1715; 3Hs.529631NM_000996RPL35A3upupRibosomal protein L35aHs.134846NM_001001410MGC2438116upHypothetical protein MGC24381Hs.492031NM_001001482FLJ110118upHypothetical protein FLJ11011Hs.188569NM_001001483ZDHHC1311downZinc finger, DHHC domain containing 13Hs.429NM_001002258ATP5G32upATP synthase, H+ transporting, mitochondrial F0complex, subunit c (subunit 9) isoform 3Hs.430439NM_001002757HIRIP52upHIRA interacting protein 5Hs.92423NM_001002838PRKWNK3downProtein kinase, lysine deficient 3Hs.522418NM_001003722GLE1L9upGLE1 RNA export medicator-like (yeast)Hs.444445NM_001003802SMARCD37upSWI/SNF related, matrix associated, actin dependentregulator of chromatin, subfamily d, member 3Hs.528826NM_001004300LOC12441116downHypothetical protein LOC124411Hs.462230NM_001004313LOC38833517downSimilar to RIKEN cDNA A730055C05 geneHs.405925NM_001005290DDA31upDifferential display and activated by p53Hs.446623NM_001005335HNRPL19; 15; 2downdownHeterogeneous nuclear ribonucleoprotein LHs.438219NM_001005408KIAA1787upG protein pathway suppressor 2Hs.334873NM_001005502CPM12upupCarboxypeptidase MHs.445841NM_001036RYR315downRyanodine receptor 3Hs.162585NM_001046SLC12A25downupdownSolute carrier family 12 (sodium/potassium/chloridetransporters), member 2Hs.12409NM_001048SST3downSomatostatinHs.159306NM_001092ABR17upActive BCR-related geneHs.274361NM_001095ACCN212downAmiloride-sensitive cation channel 2, neuronalHs.474982NM_001098ACO222upupAconitase 2, mitochondrialHs.470316NM_001105ACVR12downActivin A receptor, type IHs.461253NM_001128AP1G116upAdaptor-related protein complex 1, gamma 1 subunitHs.480653NM_001154ANXA54downAnnexin A5Hs.483239NM_001182ALDH7A15updownAldehyde dehyrogenase 7 family, member A1Hs.25447NM_001268CHC1L13downChromosome condensation 1-likeHs.162233NM_001273CHD412upChromodomain helicase DNA binding protien 4Hs.150793NM_001275CHGA14upChromogranin A (parathyroid secretory protein 1)Hs.249129NM_001279CIDEA18upCell death-inducing DFFA-like effector aHs.23748NM_001290LDB24upLIm domain binding 2Hs.13313NM_001310CREBL212downCAMP responsive element binding protein-like 2Hs.166011NM_001331CTNND111; 16;downdownCatenin (cadherin-associated protein), delta 119Hs.336916NM_001350DAXX6downupDeath-associated protein 6Hs.159195NM_001380DOCK110downDedicator of cytokinesis 1Hs.117060NM_001393ECM29downExtracellular matrix protein 2, female organ and adipocytespecificHs.196176NM_001398ECH119; XupEnoyl Coenzyme A hydratase 1, peroxisomalHs.132483NM_001410EGFL419upEGF-like-domain, multiple 4Hs.299002NM_001436FBL19downFibrillarinHs.357637NM_001439EXTL21downExostoses (multiple)-like 2Hs.272011NM_001497B4GALT19upUDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase,polypeptide 1Hs.47338NM_001549IFIT310downInterferon-induced protein with tetratricopeptide repeats 3Hs.469386NM_001566INPP4A2upInositol polyphosphate-4-phosphatase, type I, 107 kDaHs.289795NM_001584C11orf811updownChromosome 11 open reading frame 8Hs.166160NM_001607ACAA13downAcetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme A thiolase)Hs.368486NM_001649APXLXupupApical protein-like (Xenopus laevis)Hs.286221NM_001658ARF11; 19downADP-ribosylation factor 1Hs.3109NM_001666ARHGAP4XupRho GTPase activating protein 4Hs.413137NM_001680FXYD211upFXYD domain containing ion transport regulator 2Hs.198365NM_001724BPGM7up2,3-bisphosphoglycerate mutaseHs.274873NM_001751CARS11upCysteinyl-tRNA synthetaseHs.445570NM_001780CD6312; XdownCD63 antigen (melanoma 1 antigen)Hs.170129NM_001821CHML1downChoroideremia-like (Rab escort protein 2)Hs.129966NM_001842CNTFR9downCiliary neurotrophic factor receptorHs.421621NM_001864COX7A119downCytochrome c oxidase subunit subunit VIIa polypeptide 1(muscle)Hs.2242NM_001891CSN24downCasein betaHs.410037NM_001901CTGF6; 16downConnective tissue growth factorHs.465413NM_001914CYB518upCytochrome b-5Hs.113227NM_001917DAO12downD-amino-acid oxidaseHs.433839NM_001958EEF1A220; 19;upEukaryotic translation elongation factor 1 alpha 217; 9Hs.326035NM_001964EGR15downdownEarly growth response 1Hs.306251NM_001982ERBB312downV-erb-b2 erythroblastic leukemia viral oncogene homolog3 (avian)Hs.370666NM_002015FOXO1A13downdownForkhead box O1A (rhabdomyosarcoma)Hs.103183NM_002024FMR1XdowndownFragile X mental retardation 1Hs.62661NM_002053GBP11downGuanylate binding protein 1, interferon-inducible, 67 kDaHs.430425NM_002074GNB11upGuanine nucleotide binding protein (G protein), betapolypeptide 1Hs.309763NM_002092GRSF14; 17updownG-rich RNA sequence binding factor 1Hs.445733NM_002093GSK3B3downGlycogen synthase kinase 3 betaHs.181244NM_002116HLA-A6; 2; 19downdowndownMajor histocompatibility complex, class I, AHs.32309NM_002194INPP12downInosital polyphosphate-1-phosphataseHs.374097NM_002199IRF24downInterferon regulatory factor 2Hs.25292NM_002229JUNB19downdownJun B proto-oncogeneHs.408960NM_002241KCNJ101downPotassium inwardly-rectifying channel, subfamily J,member 10Hs.41696NM_002277KRTHA117downKeratin, hair, acidic, 1Hs.159590NM_002347LY6H8upupLymphocyte antigen 6 complex, locus HHs.388613NM_002499NEO115upNeogenin homolog 1 (chicken)Hs.444934NM_002525NRD11upupNardilysin (N-arginine dibasic convertase)Hs.153952NM_002526NT5E6; 3down5′-nucleotidase, ecto (CD73)Hs.467701NM_002539ODC12upOrnithine decarboxylase 1Hs.435714NM_002576PAK111upP21/Cdc42/Rac1-activated kinase 1 (STE20 homolog,yeast)Hs.483564NM_002622PFDN15downPrefoldin 1Hs.464071NM_002631PGD1upPhosphogluconate dehydrogenaseHs.468415NM_002643PIGF2upupPhosphatidylinositol glycan, class FHs.444975NM_002656PLAGL16upupPleiomorphic adenoma gene-like 1Hs.2868NM_002677PMP28downPeripheral myelin protein 2Hs.321234NM_002685EXOSC101upExosome component 10Hs.331420NM_002703PPAT4downdownPhosphoribosyl pyrophosphate amidotransferaseHs.484371NM_002752MAPK95downdownMitogen-activated protein kinase 9Hs.461777NM_002768PCOLN316upupupProcollagen (type III) N-endopeptidaseHs.368121NM_002788PSMA314downProteasome (prosome, macropain) subunit, alpha type, 3Hs.446260NM_002791PSMA614upProteasome (prosome, macropain) subunit, alpha type, 6Hs.422990NM_002797PSMB514; Y; XupProteasome (prosome, macropain) subunit, beta type, 5Hs.386866NM_002841PTPRG3downProtein tyrosine phosphatase, receptor type, GHs.127657NM_002852PTX33upPentaxin-related gene, rapidly induced by IL-1 betaHs.521640NM_002874RAD23B9downdownRAD23 homolog B (S. cerevisiae)Hs.148178NM_002885RAP1GA11upRAP1, GTPase activating protein 1Hs.423935NM_002904RDBP11; 6updownRD RNA binding proteinHs.370620NM_002908REL2downV-rel reticuloendotheliosis viral oncogene homolog(avian)Hs.115474NM_002915RFC313upReplication factor C (activator 1) 3, 38 kDaHs.23978NM_002967SAFB19; YdownScaffold attachment factor BHs.135787NM_002968SALL116downSal-like 1 (Drosophila)Hs.280202NM_002972SBF122upSET binding factor 1Hs.465924NM_003000SDHB1upSuccinate dehydrogenase complex, subunit B, iron sulfur(Ip)Hs.433795NM_003029SHC11; 6upSHC (Src homolog 2 domain containing) transformingprotein 1Hs.323878NM_003038SLC1A42downSolute carrier family 1 (glutamate/neutral amino acidtransporter), member 4Hs.443874NM_003042SLC6A13downSolute carrier family 6 (neurotrasmitter tranporter,GABA), member 1Hs.334629NM_003063SLN11upSarcolipinHs.360174NM_003068SNAI28upSnail homolog 2 (Drosophila)Hs.1063NM_003093SNRPC6upSmall nuclear ribonucleoprotein polypeptide CHs.185597NM_003119SPG716downSpastic paraplegia 7, paraplegin (pure and complicatedautosomal recessive)Hs.443861NM_003137SRPK16upSFRS protein kinase 1Hs.288229NM_003165STXBP19upSyntaxin binding protein 1Hs.482390NM_003243TGFBR31downTransforming growth factor, beta receptor III (betaglycan,300 kDa)Hs.104839NM_003255TIMP217upTissue inhibitor of metalloproteinase 2Hs.332173NM_003260TLE219upTranducin-like enhancer of split 2 (E(sp1) homolog,Drosophila)Hs.432424NM_003291TPP213downTripeptidyl peptidase IIHs.12084NM_003321TUFM16upTu translation elongation factor, mitochondrialHs.439672NM_003378VGF7downdownVGF nerve growth factor inducibleHs.388927NM_003403YY114upYY1 transcription factorHs.399810NM_003422ZNF4219upZinc finger protein 42 (myeloid-specific retinoic acid-responsive)Hs.172979NM_003451ZNF17719upZinc finger protein 177Hs.144442NM_003561PLA2G1016upPhospholipase A2, group XHs.36958NM_003567BCAR31downdownBreast cancer anti-estrogen resistance 3Hs.371698NM_003610RAE120upRAE1 RNA export 1 homolog (S. pombe)Hs.250072NM_003615SLC4A73upSolute carrier family 4, sodium bicarbonate cotransporter,member 7Hs.104925NM_003633ENC15upEctodermal-neural cortex (with BTB-like domain)Hs.104576NM_003654CHST111upCarbohydrate (keratan sulfate Gal-6) sulfotransferase 1Hs.400556NM_003657BCAS120upBreast carcinoma amplified sequence 1Hs.161181NM_003675PRPF1810downdownPRP18 pre-mRNA processing factor 18 homolog (yeast)Hs.22393NM_003677DENR12upDensity-regulated proteinHs.213264NM_003680YARS1upTyrosyl-tRNA synthetaseHs.284491NM_003681PDXK21upPyridoxal (pridoxine, vitamin B6) kinaseHs.470608NM_003705SLC25A122upSolute carrier family 25 (mitochondrial carrier, Aralar),member 12Hs.484222NM_003729RTCD11downRNA terminal phosphate cyclase domain 1Hs.4742NM_003801GPAA18; 6upGPAA1P anchor attachment protein 1 homolog (yeast)Hs.178748NM_003813ADAM2114downA disintegrin and metalloproteinase domain 21Hs.169900NM_003819PABPC41; 15; XupPoly(A) binding protein, cytoplasmic 4 (inducible form)Hs.445511NM_003831RIOK318downRIO kinase 3 (yeast)Hs.430551NM_003870IQGAP115downIQ motif containing GTPase activating protein 1Hs.7165NM_003904ZNF259upZinc finger protein 259Hs.429180NM_003908EIF2S220; 2updownupEukaryotic translation initiation factor 2, subunit 2 beta,38 kDaHs.121592NM_003916AP1S2XdownAdaptor-related protein complex 1, sigma 2 subunitHs.371199NM_003919SGCE7downSarcoglycan, epsilonHs.200770NM_003930SCAP27upSrc family associated phosphoprotein 2Hs.158460NM_003936CDK5R22updownCyclin-dependent kinase 5, regulatory subunit 2 (p39)Hs.143728NM_003941WASL7upWiskott-Aldrich syndrome-likeHs.47357NM_003956CH25H10downCholesterol 25-hydroxylaseHs.463439NM_003971SPAG917downSperm associated antigen 9Hs.315369NM_004028APQ418downAquaporin 4Hs.412117NM_004033ANXA65; 3upAnnexin A6Hs.467898NM_004036ADCY32upAdenylate cyclase 3Hs.433732NM_004071CLK12downCDC-like kinase 1Hs.292549NM_004087DLG13updowndownDiscs, large homolog 1 (Drosophila)Hs.202095NM_004098EMX210downEmpty spiracles homolog 2 (Drosophila)Hs.7557NM_004117FKBP56upFK506 binding protein 5Hs.172791NM_004182UXTXupUbiquitously-expressed transcriptHs.484703NM_004233CD836downCD83 antigen (activated B lymphocytes, immunoglobulinsuperfamily)Hs.376206NM_004235KLF49downKruppel-like factor 4 (gut)Hs.465985NM_004317ASNA119upArsA arsenite transporter, ATP-binding, homolog 1(bacterial)Hs.471401NM_004328BCS1L2upupBCS1-like (yeast)Hs.131226NM_004331BNIP3L8upBCL2/adenovirus E1B 19 kDa interacting protein 3-likeHs.13291NM_004354CCNG24upCyclin G2Hs.220529NM_004363CEACAM519; 4upupCarcinoembryonic antigen-related cell adhesion molecule 5Hs.129452NM_004392DACH113upupupDachshund homolog 1 (Drosophila)Hs.408461NM_004397DDX611downDEAD (Asp-Glu-Ala-Asp) box polypeptide 6Hs.171695NM_004417DUSP15; 16;downdownDual specificity phosphatase 111Hs.2128NM_004419DUSP510downDual specificity phosphatase 5Hs.371218NM_004438EPHA42upEPH receptor A4Hs.213389NM_004487GOLGB13upGolgi autoantigen, golgin subfamily b, macrogolgin (withtransmembrane signal), 1Hs.248746NM_004499HNRPAB5downHeterogeneous nuclear ribonucleoprotein A/BHs.472185NM_004552NDUFS51upNADH dehydrogenase (ubiquinone) Fe—S protein 5,15 kDa (NADH-coenzyme Q reductase)Hs.408257NM_004553NDUFS65upNADH dehydrogenase (ubiquinone) Fe—S protein 6,13 kDa (NADH-coenzyme Q reductase)Hs.119316NM_004564PET112L4upPET112-like (yeast)Hs.283454NM_004569PIGH14upupPhosphatidylinositol glycan, class HHs.296169NM_004578RAB4A1upupRAB4A, member RAS oncogene familyHs.129783NM_004588SCN2B11upSodium channel, voltage-gated, type II, betaHs.433201NM_004642CDK2AP112; 2downCDK2-associated protein 1Hs.106674NM_004656BAP13upBRCA1 associated protein-1 (ubiquitin carboxy-terminalhydrolase)Hs.194695NM_004675ARHI1upRas homolog gene family, member IHs.106876NM_004691ATP6V0D116upATPase, H+ transporting, lysosomal 38 kDa, V0 subunit disoform 1Hs.471779NM_004735LRRFIP12upLeucine rich repeat (in FLII) interacting protein 1Hs.2210NM_004773TRIP317upThyroid hormone receptor interactor 3Hs.464848NM_004775B4GALT618upUDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase,polypeptide 6Hs.370487NM_004776B4GALT520upUDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase,polypeptide 5Hs.26703NM_004779CNOT85updownCCR4-NOT transcription complex, subunit 8Hs.29802NM_004787SLIT24upSlit homolog 2 (Drosophila)Hs.483238NM_004815PARG11downdownPTPL1-associated RhoGAP 1Hs.240395NM_004823KCNK619upPotassium channel, subfamily K, member 6Hs.480218NM_004827ABCG24downdownATP-binding cassette, sub-family G (WHITE), member 2Hs.431109NM_004853STX817updownSyntaxin 8Hs.408515NM_004883NRG25downupNeuregulin 2Hs.282901NM_004902RNPC220; XupRNA-binding region (RNP1, RRM) containing 2Hs.446091NM_004906WTAP6downWilms tumor 1 associated proteinHs.164410NM_004913C16orf716; 6upChromosome 16 open reading frame 7Hs.2171NM_004962GDF1010downdownGrowth differentiation factor 10Hs.303870NM_004976KCNC111downPotassium voltage-gated channel, Shaw-related subfamily,member 1Hs.32505NM_004981KCNJ422downPotassium inwardly-rectifying channel, subfamily J,member 4Hs.151219NM_004984KIF5A12upKinesin family member 5AHs.149387NM_004999MYO66downdowndownMyosin VIHs.45002NM_005052RAC317upRas-related C3 botulinum toxin substrate 3 (rho family,small GTP binding protein Rac3)Hs.46440NM_005075SLCO1A212downSolute carrier organic anion transporter family, member1A2Hs.287362NM_005078TLE315upTransducin-like enhancer of split 3 (E(sp1) homolog,Drosophila)Hs.183428NM_005086SSPN12downSarcospan (Kras oncogene-associated gene)Hs.356820NM_005108XYLB3upXylulokinase homolog (H. influenzae)Hs.37288NM_005126NR1D23upNuclear receptor subfamily 1, group D, member 2Hs.1540NM_005131THOC118downdownTHO complex 1Hs.464595NM_005134PPP4R118downProtein phosphatase 4, regulatory subunit 1Hs.410944NM_005138SCO222upSCO cytochrome oxidase deficient homolog 2 (yeast)Hs.502883NM_005146SART111upSquamous cell carcinoma antigen recognised by T cellsHs.369438NM_005238ETS111downV-ets erythroblastosis virus E26 oncogene homolog 1(avain)Hs.481371NM_005245FAT4downFAT tumor suppressor homolog 1 (Drosophila)Hs.25647NM_005252FOS14; 22;downdownV-fos FBJ murine osteosarcoma viral oncogene homolog11Hs.483305NM_005340HINT15upupHistidine triad nucleotide binding protein 1Hs.2780NM_005354JUND19; 5downJun D proto-oncogeneHs.380742NM_005393PLXNB3XdownPlexin B3Hs.303090NM_005398PPP1R3C10downProtein phosphatase 1, regulatory (inhibitor) subunit 3CHs.50732NM_005399PRKAB21upupProtein kinase, AMP-activated, beta 2 non-catayticsubunitHs.129727NM_005431XRCC27downX-ray repair complementing defective repair in Chinesehamster cells 2Hs.194148NM_005433YES118; 2downV-yes-1 Yamaguchi sarcoma viral oncogene homolog 1Hs.368610NM_005443PAPSS14up3′-phosphoadenosine 5′-phosphosulfate synthase 1Hs.198612NM_005458GPR519upG protein-coupled receptor 51Hs.278500NM_005471GNPDA15; 9upGlucosamine-6-phosphate deaminase 1Hs.429294NM_005502ABCA19downATP-binding cassette, sub-family A (ABC1), member 1Hs.153299NM_005510DOM3Z6upDom-3 homolog Z(C. elegans)Hs.250666NM_005524HES13downHairy and enhancer of split 1, (Drosophila)Hs.471508NM_005544IRS12upInsulin receptor substrate 1Hs.2795NM_005566LDHA11; 17;upLactate dehydrogenase A22; 10; 2Hs.34560NM_005574LMO211downdownLIM domain only 2 (rhombotin-like 1)Hs.18069NM_005606LGMN14upLegumainHs.484324NM_005649ZNF354A5downZinc finger protein 354AHs.183671NM_005651TDO24upTryptophan 2,3-dioxygenaseHs.458917NM_005697SCAMP215downSecretory carrier membrane protein 2Hs.21577NM_005701RNUT115upRNA, U transporter 1Hs.118118NM_005723TM4SF94upTransmembrane 4 superfamily member 9Hs.381072NM_005729PPIF10updownPeptidylprolyl isomerase F (cyclophilin F)Hs.124553NM_005741ZNF26316downZinc finger protein 263Hs.123464NM_005767P2RY513downdownPurinergic receptor P2Y, G-protein coupled, 5Hs.424126NM_005770SERF215; 17;upSmall EDRK-rich factor 27; 6Hs.294603NM_005776CNIH14upCornichon homolog (Drosophila)Hs.188879NM_005777RBM63; 19downRNA binding motif protein 6Hs.436922NM_005798RFP213downdowndowndownRet finger protein 2Hs.440168NM_005822DSCR1L16upDown syndrome critical region gene 1-like 1Hs.436944NM_005841SPRY14downdownSprouty homolog 1, antagonist of FGF signaling(Drosophila)Hs.18676NM_005842SPRY213; 3downSprouty homolog 2 (Drosophila)Hs.432862NM_005885MARCH-VI5upMembrane-associated RING-CH protein VIHs.186486NM_005923MAP3K56downMitogen-activated protein kinase kinase kinase 5Hs.433391NM_005950MT1G16; 12downdownMetallothionein 1GHs.435974NM_005956MTHFD114; 2downMethylenetetrahydrofolate dehydrogenase (NADP+dependent), methenyltetrahydrofolate cyclohydrolase,formyltetrahydrofolate synthetaseHs.107474NM_005966NAB12; 4; 3upNGFI-A binding protein 1 (EGR1 binding protein 1)Hs.2430NM_005997TCFL11upTranscription factor-like 1Hs.170107NM_006003UQCRFS119upUbiquinol-cytochrome c reductase, Rieske iron-sulfurpolypeptide 1Hs.406096NM_006007ZA20D29; 19;upZinc finger, A20 domain containing 26; 17Hs.436446NM_006010ARMET3upupArginine-rich, mutated in early stage tumorsHs.412842NM_006023C10orf710downChromosome 10 open reading frame 7Hs.203620NM_006040HS3ST4upHeparan sulfate (glucosamine) 3-O-sulfotransferase 4Hs.115830NM_006043HS3ST216upHeparan sulfate (glucosamine) 3-O-sulfotransferase 2Hs.503692NM_006106YAP111downYes-associated protein 1, 65 kDAHs.367854NM_006109SKB114upSKB1 homolog (S. pombe)Hs.15250NM_006117PECI6downPeroxisomal D3,D2-enoyl-CoA isomeraseHs.369068NM_006141DNCLI216; 2upDynein, cytoplasmic, light intermediate polypeptide 2Hs.1565NM_006154NEDD415downupNeural precursor cell expressed, developmentally down-regulated 4Hs.339831NM_006211PENK8upProenkephalinHs.38449NM_006216SERPINE22downSerine (or cysteine) proteinase inhibitor, clade E (nexin,plasminogen activator inhibitor type 1), member 2Hs.334868NM_006246PPP2R5E14upProtein phosphatase 2, regulatory subunit B (B56), epsilonisoformHs.43322NM_006251PRKAA15downdownProtein kinase, AMP-activated, alpha 1 catalytic subunitHs.375001NM_006289TLN19downTalin 1Hs.25313NM_006337MCRS112downMicrospherule protein 1Hs.465784NM_006351TIMM4419upTranslocase of inner mitochondrial membrane 44 homolog(yeast)Hs.132902NM_006366CAP26upCAP, adenylate cyclase-associated protein, 2 (yeast)Hs.376064NM_006392NOL5A20; 6downNucleolar protein 5A (56 kDa with KKE/D repeat)Hs.11417NM_006423RABAC119upRab acceptor 1 (prenylated)Hs.435342NM_006425SLU75upStep II splicing factor SLU7Hs.421509NM_006430CCT42upupChaperonin containing TCP1, subunit 4 (delta)Hs.433222NM_006432NPC214; 11; 3downNiemann-Pick disease, type C2Hs.109752NM_006443C6orf1086; 20upChromosome 6 open reading frame 108Hs.14894NM_006464TGOLN22upTrans-golgi network protein 2Hs.439153NM_006502POLH6upPolymerase (DNA directed), etaHs.530045NM_006524ZNF1387downZinc finger protein 138 (clone pHZ-32)Hs.444558NM_006558KHDRBS38upKH domain containing, RNA binding, signal transductionassociated 3Hs.309288NM_006561CUGBP210upCUG triplet repeat, RNA binding protein 2Hs.368367NM_006565CTCF16downCCCTC-binding factor (zinc finger protein)Hs.159525NM_006569CGREF12upCell growth regulator with EF hand domain 1Hs.45127NM_006574CSPG53downChondroitin sulfate proteglycan 5 (neuroglycan C)Hs.155090NM_006578GNB515upGuanine nucleotide binding protein (G protein), beta 5Hs.30696NM_006602TCFL520downTranscription factor-like 5 (basic helix-loop-helix)Hs.412870NM_006627POP419; 9upProcessing of precursor 4, ribonucleass P/MRP subunit (S. cerevisiae)Hs.415846NM_006657FTCD21downupFormiminotransferase cyclodeaminaseHs.227011NM_006658C7orf167upChromosome 7 open reading frame 16Hs.199743NM_006680ME311upMalic enzyme 3, NADP(+)-dependent, mitochondrialHs.2207NM_006685PROL34downProline rich 5 (salivary)Hs.6396NM_006694JTB1upJumping translocation breakpointHs.225949NM_006707BTNL35downdownButyrophilin-like 3Hs.146804NM_006717SPIN9upSpindlinHs.505662NM_006741PPP1R1A12upProtein phosphatase 1, regulatory (inhibitor) subunit 1AHs.301404NM_006743RBM3XdownRNA binding motif (RNP1, RRM) protein 3Hs.109655NM_006746SCML1XdownSex comb on midleg-like 1 (Drosophila)Hs.192686NM_006749SLC20A28downdownSolute carrier family 20 (phosphate transporter), member 2Hs.337295NM_006819STIP111upStress-induced-phosphoprotein 1 (Hsp70/Hsp90-organizing protein)Hs.260903NM_006874ELF24upE74-like factor 2 (ets domain transcription factor)Hs.436405NM_006899IDH3B20; 6; 1downIsocitrate dehydrogenase 3 (NAD+) betaHs.466848NM_006905PSG119upPregnancy specific beta-1-glycoprotein 1Hs.435274NM_006922SCN3A2downSodium channel, voltage-gated, type III, alphaHs.376984NM_006941SOX1022downSRY (sex determining region Y)-box 10Hs.237825NM_006947SRP724; 16upSignal recognition particle 72 kDaHs.534115NM_006988ADAMTS121downdownA disintegrin-like and metalloprotease (reprolysin type) with thrombospondin type 1motif, 1Hs.30246NM_006996SLC19A21downSolute carrier family 19 (thiamine transporter), member 2Hs.29353NM_007030TPPP5; 20downBrain-specific protein p25 alphaHs.125750NM_007035KERA12upKeratocanHs.43670NM_007054KIF3A5upKinesin family member 3AHs.436896NM_007055POLR3A10upPolymerase (RNA) III (DNA directed) polypeptide A,155 kDaHs.474797NM_007061CDC42EP122upCDC42 effector protein (Rho GTPase binding) 1Hs.160958NM_007065CDC3719upCDC37 cell division cycle 37 homolog (S. cerevisiae)Hs.142245NM_007071HHLA31upHERV-H LTR-associating 3Hs.269512NM_007085FSTL13downFollistatin-like 1Hs.106857NM_007088CALB216; 19;upCalbindin 2, 29 kDa (calretinin)15Hs.484241NM_007097CLTB5upClathrin, light polypeptide (Lcb)Hs.119014NM_007147ZNF17519downdownZinc finger protein 175Hs.158174NM_007149ZNF1846downZinc finger protein 184 (Kruppel-like)Hs.434283NM_007167ZNF2581upZinc finger protein 258Hs.205163NM_007208MRPL33downMitochondrial ribosomal protein L3Hs.388668NM_007246KLHL24upKelch-like 2, Mayven (Drosophila)Hs.377070NM_007325GRIA3XupGlutamate receptor, ionotrophic, AMPA 3Hs.189826NM_007345ZNF23618upZinc finger protein 236Hs.240770NM_007362NCBP23; 1downNuclear cap binding protein subunit 2, 20 kDaHs.500104NM_012095AP3M110upAdaptor-related protein complex 3, mu 1 subunitHs.204041NM_012111AHSA114; 9upupupAHA1, activator of heat shock 90 kDa protein ATPasehomolog 1 (yeast)Hs.212395NM_012127CIZ19upCDKN1A interacting zinc finger protein 1Hs.24178NM_012155EML219upEchinoderm microtubule associated protein like 2Hs.438454NM_012173FBXO258upupF-box protein 25Hs.22867NM_012199EIF2C11downEukaryotic translation initiation factor 2C, 1Hs.179915NM_012202GNG311upGuanine nucleotide binding protein (G protein), gamma 3Hs.155742NM_012203GRHPR9; 17downGlyoxylate reductase/hydroxypyruvate reductaseHs.494804NM_012212LTB4DH9upLeukotriene B4 12-hydroxydehydrogenaseHs.481181NM_012224NEK14downNIMA (never in mitosis gene a)-related kinase 1Hs.369779NM_012238SIRT110downSirtuin (silent mating type information regulation 2homolog) 1 (S. cerevisiae)Hs.309583NM_012254SLC27A519upupSolute carrier family 27 (fatty acid transporter), member 5Hs.144287NM_012259HEY26downHairy/enhancer-of-split related with YRPW motif 2Hs.22920NM_012261C20orf10320downChromosome 20 open reading frame 103Hs.21703NM_012281KCND27upPotassium voltage-gated channel, Shal-related subfamily,member 2Hs.433057NM_012304FBXL75; 6upF-box and leucine-rich repeat protein 7Hs.397918NM_012311KIN10downKIN, antigenic determinant of recA protein homolog(mouse)Hs.370040NM_012333MYCBP1downupC-myc binding proteinHs.470417NM_012392PEF1upPEF protein with a long N-terminal hydrophobic domain(peflin)Hs.430742NM_012395PFTK17upPFTAIRE protein kinase 1Hs.167496NM_012416RANBP69upRAN binding protein 6Hs.166313NM_012419RGS176upRegulator of G-protein signaling 17Hs.471011NM_012433SF3B12upSplicing factor 3b, subunit 1, 155 kDaHs.102735NM_012446SSBP25downSingle-stranded DNA binding protein 2Hs.235750NM_012456TIMM1011; 17downTranslocase of inner mitochondrial membrane 10 homolog(yeast)Hs.59757NM_012482ZNF2811downZinc finger protein 281Hs.115721NM_013247PRSS252upProtease, serine, 25Hs.292156NM_013253DKK311upDickkopf homolog 3 (Xenopus laevis)Hs.476291NM_013286RBM15B3downRNA binding motif protein 15BHs.517436NM_013313YPEL122upupYippee-like 1 (Drosophila)Hs.433151NM_013343LOH3CR2A3; 1upLoss of heterozygosity, 3, chromosomal region 2, gene AHs.40098NM_013372GREM115upGremlin 1 homolog, cysteine knot superfamily (Xenopuslaevis)Hs.279877NM_013393FTSJ27downFtsJ homolog 2 (E. coli)Hs.7765NM_013399C16orf516upChromosome 16 open reading frame 5Hs.416049NM_013433TNPO219downTransportin 2 (importin 3, karyopherin beta 2b)Hs.408515NM_013985NRG25downupNeuregulin 2Hs.15400NM_014015DEXI16upupDexamethasone-induced transcriptHs.13370NM_014044UNC502downUnc-50 homolog (C. elegans)Hs.436500NM_014063DBNL7upDrebrin-likeHs.368971NM_014071NCOA620upNuclear receptor coactivator 6Hs.333823NM_014078MRPL138; 18downupupMitochondrial ribosomal protein L13Hs.272215NM_014079KLF153downKruppel-like factor 15Hs.11614NM_014157HSPC065downdownHSPC065 proteinHs.127496NM_014170HSPC1353upHSPC135 proteinHs.18349NM_014175MRPL158; 12downMitochondrial ribosomal protein L15Hs.445890NM_014184HSPC1631upHSPC163 proteinHs.233458NM_014223NFYC1upNuclear transcription factor Y, gammaHs.467192NM_014225PPP2R1A19upProtein phosphatase 2 (formerly 2A), regulatory subunit A(PR 65), alpha isoformHs.114062NM_014241PTPLA10upProtein tyrosine phosphatase-like (proline instead ofcatalytic arginine), member aHs.437277NM_014275MGAT4B5; 19;updownMannosyl (alpha-1,3)-glycoprotein beta-1,4-N-10acetylglucosaminyltransferase, isoenzyme BHs.301760NM_014286FREQ9downFrequenin homolog (Drosophila)Hs.193842NM_014290TDRD79downTudor domain containing 7Hs.282998NM_014309RBM922upRNA binding motif protein 9Hs.523054NM_014313SMP111upNPD014 proteinHs.330384NM_014325CORO1C12upCoronin, actin binding protein, 1CHs.370510NM_014333IGSF411downupImmunoglobulin superfamily, member 4Hs.189810NM_014351SULT4A122upSulfotransferase family 4A, member 1Hs.310431NM_014386PKD2L25downPolycystic kidney disease 2-like 2Hs.316890NM_014426SNX520downSorting nexin 5Hs.443577NM_014452TNFRSF216upTumor necrosis factor receptor superfamily, member 21Hs.232543NM_014456PDCD410downProgrammed cell death 4 (neoplastic transformationinhibitor)Hs.111632NM_014463LSM33upLSM3 homolog, U6 small nuclear RNA associated (S. cerevisiae)Hs.438994NM_014480ZNF54419upZinc finger protein 544Hs.221436NM_014483RBMS33downRNA binding motif, single stranded interacting proteinHs.143600NM_014498GOLPH43downGolgi phosphoprotein 4Hs.252682NM_014506TOR1B9upTorsin family 1, member B (torsin B)Hs.13014NM_014570ARFGAP322downADP-ribosylation factor GTPase activating protein 3Hs.525339NM_014584ERO1L14upERO1-like (S. cerevisiae)Hs.180933NM_014593CXXC118upCXXC finger 1 (PHD domain)Hs.368808NM_014600EHD32; 6upEH-domain containing 3Hs.26704NM_014608CYFIP115; 9downCytoplasmic FMR1 interacting protein 1Hs.413801NM_014614PSME42upProteasome (prosome, macropain) activator subunit 4Hs.330073NM_014686KIAA035519upKIAA0355Hs.196054NM_014707HDAC97downHistone deacetylase 9Hs.410092NM_014741KIAA065211downKIAA0652 gene productHs.484349NM_014757MAML15downMastermind-like 1 (Drosophila)Hs.44024NM_014763MRPL192upMitochondrial ribosomal protein L19Hs.370530NM_014788TRIM149downTripartite motif-containing 14Hs.533245NM_014829DDX465downDEAD (Asp-Glu-Ala-Asp) box polypeptide 46Hs.3094NM_014876KIAA006322upKIAA0063 gene productHs.130014NM_014880DCL-12downType I transmembrane C-type lectin receptor DCL-1Hs.227602NM_014892RBM166downRNA binding motif protein 16Hs.131683NM_014912CPEB310upCytoplasmic polyadenylation element binding protein 3Hs.22109NM_014952BAHD115upBromo adjacent homology domain containing 1Hs.124490NM_014977ACIN114downApoptotic chromatin condensation inducer 1Hs.270499NM_015001SHARP1upSMART/HDAC1 associated repressor proteinHs.159669NM_015002FBXO2112upF-box protein 21Hs.189409NM_015033FNBP19downFormin binding protein 1Hs.293593NM_015071ARHGAP265upRho GTPase activating protein 26Hs.440414NM_015087SPG2013downSpastic paraplegia 20, spartin (Troyer syndrome)Hs.269775NM_015093MAP3K7IP26upMitogen-activated protein kinase kinase kinase 7interacting protein 2Hs.331431NM_015200SCC-1124downSCC-112 proteinHs.410497NM_015379BRI3upBrain protein I3Hs.105547NM_015392NPDC19upNeural proliferation, differentiation and control, 1Hs.25956NM_015464SOSTDC17upSclerostin domain containing 1Hs.472630NM_105474SAMHD120downSAM domain and HD domain 1Hs.146100NM_015553PIP3-E6upPhosphoinositide-binding protein PIP3-EHs.127401NM_015662SLBupSelective LIM binding factor, rat homologHs.369144NM_015665AAAS12upAchalasis, adrenocortical insufficiency, alacrimia(Allgrove, triple-A)Hs.391481NM_015693PDZK64upPDZ domain containing 6Hs.474914NM_015705RUTBC322downRUN and TBC1 domain containing 3Hs.512592NM_015713RRM2B8upRibonucleotide reductase M2 B (TP53 inducible)Hs.235368NM_015719COL5A319downdowndowndownCollagen, type V, alpha 3Hs.250693NM_015852ZNF1177; 19downKrueppel-related zinc finger proteinHs.128959NM_015885PCF1111downPre-mRNA cleavage complex II protein Pcf11Hs.348326NM_015894STMN320downStathmin-like 3Hs.414952NM_015910LOC510572upHypothetical protein LOC51057Hs.16606NM_015960CUTC10downCutC copper transporter homolog (E. Coli)Hs.435759NM_015963THAP42upTHAP domain containing 4Hs.44298NM_015969MRPS177upMitochondrial ribosomal protein S17Hs.370703NM_015974CRYL113downCrystallin, lambda 1Hs.370168NM_015986CRLF317downCytokine receptor-like factor 3Hs.525752NM_015995KLF1315upKruppel-like factor 13Hs.271876NM_016010CGI-628downCGI-62 proteinHs.183646NM_016011CGI-631downNuclear receptor binding factor 1Hs.106529NM_016013NDUFAF115upNADH dehydrogenase (ubiquinone) 1 alpha subcomplex,assembly factor 1Hs.514216NM_016016CGI-6917updownCGI-69 proteinHs.145386NM_016033CGI-908downdownCGI-90 proteinHs.3945NM_016045C20orf4520downChromosome 20 open reading frame 45Hs.483296NM_016048CGI-1115upCGI-111 proteinHs.271614NM_016049C14orf12214upChromosome 14 open reading frame 122Hs.157401NM_016058CGI-1212upupCGI-121 proteinHs.436161NM_016067MRPS18C4upMitochondrial ribosomal protein S18CHs.435952NM_016082CDK5RAP120downCDK5 regulatory subunit associated protein 1Hs.25829NM_016084RASD117upRAS, dexamethasone-induced 1Hs.108969NM_016145PTD00819upPTD008 proteinHs.159581NM_016155MMP1712upMatrix metalloproteinase 17 (membrane-inserted)Hs.446179NM_016200LSM87upLSM8 homolog, U6 small nuclear RNA associated (S. cerevisiae)Hs.131133NM_016203PRKAG27upupProtein kinase, AMP-activated, gamma 2 non-catalyticsubunitHs.135756NM_016218POLK5upPolymerase (DNA directed) kappaHs.328865NM_016221DCTN45downDynactin 4 (p62)Hs.191213NM_016224SNX96upSorting nexin 9Hs.208759NM_016231NLK17upNemo like kinaseHs.148685NM_016235GPRC5B16downG protein-coupled receptor, family C, group 5, member BHs.334832NM_016243NQO3A21downNAD(P)H: quinone oxidoreductase type 3, polypeptide A2Hs.18788NM_016246DHRS1019downdownDehydrogenase/reductase (SDR family) member 10Hs.125132NM_016269LEF14updownLymphoid enhancer-binding factor 1Hs.278627NM_016297PCYOX12upPrenylcysteine oxidase 1Hs.470887NM_016315GULP12downGULP, engulfment adaptor PTB domain containing 1Hs.158530NM_016339RAPGEFL117upRap guanine nucleotide exchange factor (GEF)-like 1Hs.483329NM_016340RAPGEF65upKIAA1961 proteinHs.224137NM_016390C9orf1149downChromosome 9 open reading frame 114Hs.497967NM_016396HSPC12915upHypothetical protein HSPC129Hs.283322NM_016401HSPC13811upupHypothetical protein HSPC138Hs.436502NM_016486LOC512491downHypothetical protein LOC51249Hs.408233NM_016492RANGNRF17upRAN guanine nucleotide release factorHs.524094NM_016505PS1D1upPutative S1 RNA binding domain proteinHs.29549NM_016511CLEC112; XdownC-type lectin-like receptor-1Hs.478393NM_016559PEX5L3upPeroxisomal biogenesis factor 5-likeHs.475387NM_016565E2IG211downE2IG2 proteinHs.200063NM_016596HDAC7A12downHistone deacetylase 7AHs.433439NM_016622MRPL35upMitochondrial ribosomal protein L35Hs.478064NM_016625MGC121973upBM-011 proteinHs.465144NM_016626RKHD218upRing finger and KH domain containing 2Hs.278635NM_016648HDCMA18P4downHDCMA18P proteinHs.369284NM_016649C20orf620downChromosome 20 open reading frame 6Hs.171342NM_016652CRNKL120upupCrn, crooked neck-like 1 (Drosophila)Hs.127792NM_016941DLL319upDelta-like 3 (Drosophila)Hs.40735NM_017412FZD38downFrizzled homolog 3 (Drosophila)Hs.286233NM_017425SPA1711; 19upSperm autoantigenic protein 17Hs.108112NM_017443POLE39downPolymerase (DNA directed), epsilon 3 (p17 subunit)Hs.437084NM_017544NKRFXupNF-kappa B repressing factorHs.458304NM_017578ROPN13downRopporin, rhophilin associated protein 1Hs.165636NM_017594DIRAS29upupDIRAS family, GTP-binding RAS-like 2Hs.258798NM_017615C10orf8610updowndownChromosome 10 open reading frame 86Hs.29700NM_017665ZCCHC105downZinc finger, CCHC domain containing 10Hs.369932NM_017714C20orf1320upChromosome 20 open reading frame 13Hs.483993NM_017734PALMD1downPalmdelphinHs.440401NM_017750FLJ202962downdownAll-trans-13,14-dihydroretinol saturaseHs.147836NM_017768FLJ203311downHypothetical protein FLJ20331Hs.444269NM_017776FLJ20344XupupHypothetical protein FLJ20344Hs.150122NM_017784OSBPL103upupOxysterol binding protein-like 10Hs.368710NM_017785FLJ203645downHypothetical protein FLJ20364Hs.408652NM_017794KIAA17979upupKIAA1797Hs.371210NM_017847C1orf271updownChromosome 1 open reading frame 27Hs.134406NM_017853TXNL4B16upupThioredoxin-like 4BHs.105606NM_017854FLJ2051219upupdownHypothetical protein FLJ20512Hs.377705NM_017865FLJ205311upHypothetical protein FLJ20531Hs.525238NM_017924C14orf11914upupupChromosome 14 open reading frame 119Hs.249591NM_017925C9orf559downChromosome 9 open reading frame 55Hs.30783NM_017967FLJ2085019upHypothetical protein FLJ20850Hs.297044NM_017987RUFY210downRun and FYVE domain containing 2Hs.516341NM_017991FLJ100812downHypothetical protein FLJ10081Hs.128258NM_017993FLJ1009413upupupHypothetical protein FLJ10094Hs.532296NM_017998C9orf409upChromosome 9 open reading frame 40Hs.241523NM_018008ZNF3123upupZinc finger protein 312Hs.445244NM_018013FLJ101596upHypothetical protein FLJ10159Hs.353454NM_018045FLJ102761upHypothetical protein FLJ10276Hs.213393NM_018046VG5Q5upAngiogenic factor VG5QHs.262823NM_018060FLJ103261downMitochondrial isoleucine tRNA synthetaseHs.356096NM_018067FLJ103501upHypothetical protein FLJ10350Hs.270107NM_018115SDAD14upSDA1 domain containing 1Hs.447458NM_018121C10orf610upHypothetical protein LOC143286Hs.31082NM_018126TMEM334upTransmembrane protein 33Hs.26156NM_018216PANK41; 2upPantothenate kinase 4Hs.174021NM_018225SMU19upSmu-1 suppressor of mec-8 and unc-52 homolog (C. elegans)Hs.260238NM_018238FLJ108427; 12; 2downHypothetical protein FLJ10842Hs.502773NM_018269MTCBP-12upMembrane-type 1 matrix metalloproteinase cytoplasmictail binding protein-1Hs.368960NM_018297NGLY13upN-glycanase 1Hs.503022NM_018312C11orf2311downChromosome 11 open reading frame 23Hs.58382NM_018322C6orf646downChromosome 6 open reading frame 64Hs.458312NM_018328MBD52downMethyl-CpG binding domain protein 5Hs.37558NM_018339RFK9upRiboflavin kinaseHs.176227NM_018342FLJ111554downHypothetical protein FLJ11155Hs.416755NM_018357FLJ1119615downAcheronHs.271643NM_018368C6orf2096downChromosome 6 open reading frame 209Hs.211828NM_018428HCA6617upHepatocellular carcinoma-associated antigen 66Hs.32148NM_018445SELS15downSelenoprotein SHs.477325NM_018456EAF23upELL associated factor 2Hs.370102NM_018464C10orf7010upChromosome 10 open reading frame 70Hs.479766NM_018475TPARL4downTPA regulated locusHs.12126NM_018487HCA1127; 20;downdownHepatocellular carcinoma-associated antigen 11212Hs.207433NM_018557LRP1B2downLow density lipoprotein-related protein 1B (deleted intumors)Hs.435991NM_018569C4orf164upChromosome 4 open reading frame 16Hs.519346NM_018695ERBB2IP5downErbb2 interacting proteinHs.47572NM_018696ELAC118upElaC homolog 1 (E. coli)Hs.104980NM_018706DHTKD110upDehydrogenase E1 and transketolase domain containing 1Hs.184062NM_018840C20orf2420; 10upChromosome 20 open reading frame 24Hs.145256NM_018930PCDHB105downProtocadherin beta 10Hs.190518NM_018944C21orf4521upChromosome 21 open reading frame 45Hs.484686NM_018988GFOD16upupGlucose-fructose oxidoreductase domain containing 1Hs.440534NM_019022FLJ2079318downFLJ20793 proteinHs.288224NM_019051MRPL509upMitochondrial ribosomal protein L50Hs.481836NM_019061PIP3AP5upPhosphatidylinositol-3-phosphate associated proteinHs.140950NM_019065EFCBP216downEF hand calcium binding protein 2Hs.173524NM_019081LKAP16upLimkain b1Hs.466714NM_019088PD219downdownHypothetical protein F23149_1Hs.323396NM_019557LOC561811upHypothetical protein RP1-317E23Hs.178011NM_019606FLJ202577upHypothetical protein FLJ20257Hs.443529NM_019607FLJ112678downHypothetical protein FLJ11267Hs.100890NM_019845RPRM2upReprimo, TP53 dependent G2 arrest mediator candidateHs.413083NM_019863F8XupCoagulation factor VIII, procoagulant component(hemophilia A)Hs.133512NM_020119ZC3HAV1upZinc finger CCCH type, antiviral 1Hs.193226NM_020121UGCGL213downUDP-glucose ceramide glucosyltransferase-like 2Hs.109929NM_020137GRIPAP1downGRIP1 associated protein 1Hs.262858NM_020143LOC569022upPutatative 28 kDa proteinHs.460242NM_020145SH3GLB29downSH3-domain GRB2-like endophilin B2Hs.160565NM_020154C15orf2415downChromosome 15 open reading frame 24Hs.250456NM_020162DHX3317upDEAH (Asp-Glu-Ala-His) box polypeptide 33Hs.47649NM_020166MCCC13downMethylcrotonoyl-Coenzyme A carboxylase 1 (alpha)Hs.42785NM_020186ACN97upACN9 homolog (S. cerevisiae)Hs.202011NM_020198GK00117downGK001 proteinHs.6434NM_020215C14orf13214upChromosome 14 open reading frame 132Hs.127432NM_020234MDS00915upupX 009 proteinHs.22065NM_020239CDC42SE11upCDC42 small effector 1Hs.118241NM_020247CABC11upChaperone, ABC1 activity of bc1 complex like (S. pombe)Hs.300404NM_020314MGC1682416downEsophageal cancer associated proteinHs.194408NM_020340KIAA12446upKIAA1244Hs.477869NM_020353PLSCR43downPhospholipid scramblase 4Hs.322901NM_020368SAS104upDisrupter of silencing 10Hs.390623NM_020383XPNPEP110upX-prolyl aminopeptidase (aminopeptidase P) 1, solubleHs.387755NM_020408C6orf1496downChromosome 6 open reading frame 149Hs.483841NM_020466DJ122O8.26updownHypothetical protein dJ122O8.2Hs.481545NM_020546ADCY25upupupAdenylate cyclase 2 (brain)Hs.104613NM_020640RP423upRP42 homologHs.529551NM_020654SENP73downSUMO1/sentrin specific protease 7Hs.123450NM_020655JPH316downKelch domain containing 4Hs.287374NM_020657ZNF30419downZinc finger protein 304Hs.283816NM_020660CX3615downConnexin-36Hs.47166NM_020685C3orf143; 11upChromosome 3 open reading frame 14Hs.211252NM_020689SLC24A320upSolute carrier family 24 (sodium/potassium/calciumexchanger), member 3Hs.526401NM_020695TCEB3BP119downTranscription elongation factor B polypeptide 3 bindingprotein 1Hs.17255NM_020706SFRS1521upSplicing factor, arginine/serine-rich 15Hs.434947NM_020727ZNF29521downZinc finger protein 295Hs.211751NM_020836KIAA144614downBrain-enriched guanylate kinase-associated proteinHs.515351NM_020895KIAA153319upKIAA1533Hs.443891NM_020925KIAA15731downKIAA1573 proteinHs.8453NM_020932MAGEE1downMelanoma antigen, family E, 1Hs.368525NM_020992PDLIM110upPDZ and LIM domain 1 (elfin)Hs.243994NM_021008DEAF111upDeformed epidermal autoregulatory factor 1 (Drosophila)Hs.198760NM_021076NEFH22; 20upNeurofilament, heavy polypeptide 200 kDaHs.192215NM_021116ADCY17upupAdenylate cyclase 1 (brain)Hs.437403NM_021129PP10upPyrophosphatase (inorganic)Hs.147119NM_021135RPS6KA26upRibosomal protein S6 kinase, 90 kDa, polypeptide 2Hs.154029NM_021170HES41downHairy and enhancer of split 4 (Drosophila)Hs.119889NM_021183RAP2CXdownRAP2C, member of RAS oncogene familyHs.445489NM_021200PLEKHB111; 8;downPleckstrin homology domain containting, family B12;(evectins) member 115; 20Hs.435106NM_021205RHOU1downRas homolog gene family, member UHs.494854NM_021218C9orf809upChromosome 9 open reading frame 80Hs.234282NM_021729VPS1111downdownVacuolar protein sorting 11 (yeast)Hs.301062NM_021808GALNT912upupUDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyltransferase 9 (GalNAc-T9)Hs.269764NM_021813BACH26upBTB and CNC homology 1, basic leucine zippertranscription factor 2Hs.257341NM_021818SAV114downSalvador homolog 1 (Drosophila)Hs.461954NM_021947SRR17downdowndownSerine racemaseHs.213050NM_021952ELAVL41upELAV (embryonic lethal, abnormal vision, Drosophila)-like 4 (Hu antigen D)Hs.329978NM_022087GALNT117upUDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyltransferase 11 (GalNAc-T11)Hs.128866NM_022089HSA99471upPutative ATPaseHs.247324NM_022100MRPS141upMitochondrial ribosomal protein S14Hs.198158NM_022129MAWBP10downMAWD binding proteinHs.488591NM_022479WBSCR177upWilliams-Beuren syndrome chromosome region 17Hs.480356NM_022553VPS526downupVacuolar protein sorting 52 (yeast)Hs.510265NM_022731NUCKS1downNuclear ubiquitous casein kinase and cyclin-dependentkinase substrateHs.440880NM_022753FLJ129031downHypothetical protein FLJ12903Hs.369440NM_022754SFXN15updownSideroflexin 1Hs.284630NM_022771TBC1D1512downTBC1 domain family, member 15Hs.27836NM_022823FNDC42downdownFibronectin type III domain containing 4Hs.370549NM_022893BCL11A2downB-cell CLL/lymphoma 11A (zinc finger protein)Hs.203559NM_022915MRPL442upMitochondrial ribosomal protein L44Hs.239154NM_023039ANKRA25; 3downAnkyrin repeat, family A (RFXANK-like), 2Hs.146679NM_023071SPATS212upSpermatogenesis associated, serine-rich 2Hs.169615NM_023080FLJ209898upHypothetical protein FLJ20989Hs.225641NM_023923PHACTR41downPhosphatase and actin regulator 4Hs.169054NM_023928AACS12upupAcetoacetyl-CoA synthetaseHs.157160NM_023936MRPS3416upMitochondrial ribosomal protein S34Hs.458367NM_024026MRP6313; 8upupupMitochondrial ribosomal protein 63Hs.15580NM_024075LENG519upLeukocyte receptor cluster (LRC) member 5Hs.59804NM_024077SECISBP29; 17upSECIS binding protein 2Hs.412939NM_024090ELOVL64upELOVL family member 6, elongation of long chain fattyacids (FEN1/Elo2, SUR4/Elo3-like, yeast)Hs.441734NM_024091MGC52975downHypothetical protein MGC5297Hs.177688NM_024303ZSCAN519downZinc finger and SCAN domain containing 5Hs.28144NM_024333FSD119upFibronectin type 3 and SPRY domain containing 1Hs.499205NM_024336IRX316downIroquois homeobox protein 3Hs.118394NM_024345MGC107659downHypothetical protein MGC10765Hs.211914NM_024407NDUFS719upNADH dehydrogenase (ubiquinone) Fe—S protein 7,20 kDa (NADH-coenzyme Q reductase)Hs.485004NM_024493ZNF3066downdownZinc finger protein 306Hs.250693NM_024498ZNF1177; 19downkrueppel-related zinc finger proteinHs.460568NM_024516MGC460616downHypothetical protein MGC4606Hs.211441NM_024517PHF29downPHD finger protein 2Hs.443789NM_024581C6orf606downChromosome 6 open reading frame 60Hs.470679NM_024583SCRN32downSecernin 3Hs.39311NM_024592SRD5A2L4upSteroid 5 alpha-reductase 2-likeHs.293563NM_024595FLJ126661downHypothetical protein FLJ12666Hs.200943NM_024611NARG215upNMDA receptor regulated 2Hs.317340NM_024683FLJ2272917upHypothetical protein FLJ22729Hs.445826NM_024686FLJ230331upHypothetical protein FLJ23033Hs.121915NM_024742ARMC516downArmadillo repeat containing 5Hs.468349NM_024766FLJ234512downHypothetical protein FLJ23451Hs.443061NM_024810CXorf45XdownChromosome X open reading frame 45Hs.533446NM_024812BAALC8upBrain and acute leukemia, cytoplasmicHs.187377NM_024847TMC716upTransmembrane channel-like 7Hs.193170NM_024859FLJ21687XupPDZ domain containing, X chromosomeHs.478465NM_024871FLJ127483downdownHypothetical protein FLJ12748Hs.456507NM_024874PKD1-like1upPolycystic kidney disease 1-likeHs.130712NM_024876ADCK419upAarF domain containing kinase 4Hs.374147NM_024943FLJ232354downHypothetical protein FLJ23235Hs.371096NM_025021MECT119upMucoepidermoid carcinoma translocated 1Hs.260555NM_025057C14orf4514upChromosome 14 open reading frame 45Hs.24808NM_025073FLJ211681upHypothetical protein FLJ21168Hs.302051NM_025109MYOHD1downMyosin head domain containing 1Hs.288945NM_025147FLJ134482downHypothetical protein FLJ13448Hs.288981NM_025152C14orf12714upChromosome 14 open reading frame 127Hs.301526NM_025188TRIM451upTripartite motif-containing 45Hs.443723NM_025196GRPEL14downGrpE-like 1, mitochondrial (E. coli)Hs.434075NM_025205MED284; 1upMediator of RNA polymerase II transcription, subunit 28homolog (yeast)Hs.329327NM_025235TNKS210downTankyrase, TRF1-interacting ankyrin-related ADP-ribosepolymerase 2Hs.118354NM_025263PRR36; 4upProline rich 3Hs.189445NM_030583MATN28downMatrilin 2Hs.127126NM_030627CPEB45downCytoplasmic polyadenylation element binding protein 4Hs.11067NM_030630C17orf2817downChromosome 17 open reading frame 28Hs.209561NM_030650KIAA17152downKIAA1715Hs.449628NM_030759NRBF210; 8; 1upNuclear receptor binding factor 2Hs.177841NM_030762BHLHB312downBasic helix-loop-helix domain containing, class B, 3Hs.480519NM_030821PLA2G12A4upPhospholipase A2, group XIIAHs.538547NM_030920ANP32E1downAcidic (leucine-rich) nuclear phosphoprotein 32 family,member EHs.436996NM_030948PHACTR16downPhosphatase and actin regulator 1Hs.267120NM_030963RNF1466downRing finger protein 146Hs.300816NM_030981RAB1B11; 9; 2upupRAB1B, member RAS oncogene familyHs.301048NM_031216SEH1L18upSEH1-like (S. cerevisiae)Hs.247280NM_031227C20orf1820; 18upChromosome 20 open reading frame 18Hs.109051NM_031286SH3BGRL31upSH3 domain binding glutamic acid-rich protein like 3Hs.110695NM_031287SF3B56; 20;upupSplicing factor 3b, subunit 5, 10 kDa12Hs.270437NM_031361COL4A3BP5upCollagen, type IV, alpha 3 (Goodpasture antigen) bindingproteinHs.378808NM_032025eIF2A3downdowndowndownEukaryotic translation initiation factor (eIF) 2AHs.465642NM_032108SEMA6B19upSema domain, transmembrane domain (TM), andcytoplasmic domain, (semaphorin) 6BHs.501793NM_032127DKFZP566M104611downHypothetical protein DKFZp566M1046Hs.100914NM_032142Cep19218downCentrosomal protein 192 kDaHs.519326NM_032151DCOHM5upDimerization cofactor of hepatocyte nuclear factor 1(HNF1) from muscleHs.399984NM_032168FLJ12519downdownHypothetical protein FLJ12519Hs.441378NM_032169FLJ125923upPutative acyl-CoA dehydrogenaseHs.339612NM_032177PHAX5upupRNA U, small nuclear RNA export adaptor(phosphorylation regulated)Hs.381214NM_032261C21orf5621upChromosome 21 open reading frame 56Hs.19673NM_032272MAF1upHomolog of yeast MAF1Hs.124015NM_032304HAGHL16downHydroxyacylglutathione hydrolase-likeHs.9088NM_032305MGC32001upHypothetical protein LOC284615Hs.513315NM_032349SDOS16upHypothetical protein MGC11275Hs.239500NM_032366MGC1311416upHypothetical protein MGC13114Hs.277154NM_032380GFM25upG elogation factor, mitochondrial 2Hs.438709NM_032446MEGF105updownMEGF10 proteinHs.437126NM_032496ARHGAP912upRho GTPase activating protein 9Hs.144527NM_032507PGBD16downPiggyBac transposable element derived 1Hs.486010NM_032511C6orf1686downChromosome 6 open reading frame 168Hs.293753NM_032515BOK2upBCL2-related ovarian killerHs.163642NM_032536NTNG29downNetrin G2Hs.501106NM_032550KIAA191410downKIAA1914Hs.387255NM_032569N-PAC16upCytokine-like nuclear factor n-pacHs.145010NM_032576CYorf15BYupChromosome Y open reading frame 15BHs.132868NM_032582USP3217upUbiquitin specific protease 32Hs.277101NM_032609COX4I220upCytochrome c oxidase subunit IV isoform 2 (lung)Hs.154140NM_032623OSAP4upOvary-specific acidic proteinHs.129634NM_032630CINP14; 12upCyclin-dependent kinase 2-interacting proteinHs.459379NM_032687CYHR18upCystein and histidine rich 1Hs.436035NM_032704TUBA612downTubulin alpha 6Hs.476972NM_032778MINA3upupMYC induced nuclear antigenHs.401537NM_032802SPPL2A15downPutative intramembrane cleaving proteaseHs.435948NM_032810ATAD110downATPase family, AAA domain containing 1Hs.190983NM_032813FLJ1462413downHypothetical protein FLJ14624Hs.461113NM_032830CIRH1A16downCirrhosis, autosomal recessive 1A (cirhin)Hs.520287NM_032870C6orf1116downChromosome 6 open reading frame 111Hs.388645NM_032901MGC1428812upHypothetical protein MGC14288Hs.406788NM_032932RAB11FIP44upRAB11 family interacting protein 4 (class II)Hs.505676NM_033082CIP2912upCytokine induced protein 29 kDaHs.292986NM_033115MGC161694upHypothetical protein MGC16169Hs.224843NM_033210ZNF5023upZinc finger protein 502Hs.370530NM_033220TRIM149downTripartite motif-containing 14Hs.297452NM_033260FOXQ16downForkhead box Q1Hs.27695NM_033291MID1XupMidline 1 (Opitz/BBB syndrome)Hs.335033NM_033427CTTNBP27upCortactin binding protein 2Hs.348390NM_033439C9orf269downdownChromosome 9 open reading frame 26 (NF-HEV)Hs.348262NM_033495KLHL13upKelch-like 13 (Drosophila)Hs.347270NM_033554HLA-DPA16downdownMajor histocompatibility complex, class II, DP alpha 1Hs.200600NM_052837SCAMP31upSecretory carrier membrane protein 3Hs.440092NM_052839PANX2downPannexin 2Hs.320823NM_052865C20orf7220downChromosome 20 open reading frame 72Hs.12082NM_053000TIGA1upTIGA1Hs.408427NM_053041COMMD720upCOMM domain containing 7Hs.231029NM_053045MGC143279downHypothetical protein MGC14327Hs.203717NM_054034FN12downFibronectin 1Hs.410830NM_058190C21orf7021upChromosome 21 open reading frame 70Hs.287518NM_080415PNUTL217downPeanut-like 2 (Drosophila)Hs.55940NM_080430SELMupSelenoprotein MHs.179080NM_080552SLC32A120upSolute carrier family 32 (GABA vesicular transporter),member 1Hs.156506NM_080656MGC130175upSimilar to RIKEN cDNA A4301010B06 geneHs.264208NM_080667MGC154072downSimilar to RIKEN cDNA 4931428D14 geneHs.269577NM_080841VPS1620downupProtein tyrosine phosphatase, receptor type, AHs.135805NM_080875LOC1426781upSkeletrophinHs.464422NM_130386COLEC1218downCollectin sub-family member 12Hs.304578NM_130442ELMO17downEngulfment and cell motility 1 (ced-12 homolog, C. elegans)Hs.196482NM_130469JDP214downJun dimerization protein 2Hs.16258NM_130781RAB245upRAB24, member RAS oncogene familyHs.483259NM_130809LOC1336195downHypothetical protein MGC12103Hs.103315NM_133476ZNF38412downZinc finger protein 384Hs.156316NM_133503DCN12downDecorinHs.156316NM_133504DCN12downDecorinHs.411488NM_138290RPIB97upRap2-binding protein 9Hs.264345NM_138330TIZ19upTRAF6-inhibitory zinc finger proteinHs.29645NM_138364LOC908264upHypothetical protein BC004337Hs.370055NM_138409C6orf1176upChromosome 6 open reading frame 117Hs.460487NM_138414LOC11286916upHypothetical protein BC011981Hs.348411NM_138467LOC1272531upHypothetical protein BC009514Hs.444338NM_138698LOC914314downPrematurely terminated mRNA decay factor-likeHs.129159NM_138701C7orf117upChromosome 7 open reading frame 11Hs.484371NM_139068MAPK95downdownMitogen-activated protein kinase 9Hs.356523NM_139124MAPK8IP222;downMitogen-activated protein kinase 8 interacting protein 220; 12Hs.21187NM_139169TRUB110upTruB pseudouridine (psi) synthase homolog 1 (E. coli)Hs.33470NM_139278LGI38upLeucine-rich repeat LGI family, member 3Hs.65256NM_139284LGI419downLeucine-rich repeat LGI family, member 4Hs.173034NM_139316AMPH7upAmphiphysin (Stiff-Man syndrome with breast cancer128 kDa autoantigen)Hs.193163NM_139351BIN12; 11upBridging integrator 1Hs.371240NM_144497AKAP126upA kinase (PRKA) anchor protein (gravin) 12Hs.469264NM_144563RPIA2downRibose 5-phosphate isomerase A (ribose 5-phosphateepimeraseHs.7962NM_144584FLJ305251upHypothetical protein FLJ30525Hs.432780NM_144638MGC299563upHypothetical protein MGC29956Hs.55150NM_144647MGC266105downHypothetical protein MGC26610Hs.12381NM_144669FLJ3197812upHypothetical protein FLJ31978Hs.283828NM_144770RBM1121downRNA binding motif protein 11Hs.502223NM_144981FLJ2505911upupHypothetical protein FLJ25059Hs.533086NM_144996ARL2L13upADP-ribosylation factor-like 2-like 1Hs.413359NM_145030MGC227937downdownHypothetical protein MGC22793Hs.293818NM_145043NEIL28upNei like 2 (E. coli)Hs.202207NM_145047NOR11upOxidored-nitro domain-containing proteinHs.515490NM_145056MGC1547619downThymus expressed gene 3-likeHs.294145NM_145265LOC133957upSimilar to RIKEN cDNA 0610011N22Hs.418495NM_145267C6orf576downChromosome 6 open reading frame 57Hs.522992NM_145306C10orf3510upChromosome 10 open reading frame 35Hs.21938NM_148907OSBPL91downOxysterol binding protein-like 9Hs.15783NM_152302C20orf15820downChromosome 20 open reading frame 158Hs.434914NM_152330C14orf3114downChromosome 14 open reading frame 31Hs.374556NM_152339MGC2688516downHypothetical protein MGC26885Hs.135181NM_152361FLJ3894419upHypothetical protein FLJ38944Hs.462033NM_152371MGC268181downdownHypothetical protein MGC26818Hs.523413NM_152372MYOM31upMyomesin family, member 3Hs.434945NM_152379DKFZp547B17131downHypothetical protein DKFZp547B1713Hs.524828NM_152437DKFZp761B12812upHypothetical protein DKFZp761B128Hs.520192NM_152608FLJ353821downHypothetical protein FLJ35382Hs.400698NM_152618FLJ356304upHypothetical protein FLJ35630Hs.507584NM_152705MGC985013upPolymerase (RNA) I polypeptide D, 16 kDaHs.487564NM_152745NXPH17downNeurexophilin 1Hs.534591NM_152766MGC4010717upupHypothetical protein MGC40107Hs.135997NM_152773LOC1162113upHypothetical protein BC013113Hs.200100NM_152793Ells17upupHypothetical protein Ells1Hs.12102NM_152827SNX36; 7upSorting nexin 3Hs.27788NM_153034ZNF48810upZinc finger protein 488Hs.390567NM_153048FYN6; 20upFYN oncogene related to SRC, FGR, YESHs.504943NM_153207AEBP212downAE binding protein 2Hs.460217NM_153208MGC3504816downHypothetical protein MGC35048Hs.180257NM_153231ZNF55019downZinc finger protein 550Hs.436743NM_153233FLJ3644519upHypothetical protein FLJ36445Hs.128188NM_153234C5orf115downChromosome 5 open reading frame 11Hs.511991NM_153240NPHP33downNephronophthisis 3 (adolescent)Hs.399779NM_153266MGC33486upHypothetical protein MGC33486Hs.171001NM_153456HS6ST313downHeparan sulfate 6-O-sulfotransferase 3Hs.156723NM_153498CAMK1D10downCalcium/calmodulin-dependent protein kinase IDHs.484195NM_153607LOC1532225upAdult retina proteinHs.40910NM_153634CPNE812upCopine VIIIHs.435080NM_153690FAM34A3upFamily with sequence similarity 43, member AHs.110477NM_153741DPM31upDolichyl-phosphate mannosyltransferase polypeptide 3Hs.511774NM_153750C12orf8118downdownChromosome 21 open reading frame 81Hs.463985NM_170741KCNJ1617downPotassium inwardly-rectifying channel, subfamily J,member 16Hs.297343NM_172226CAMKK212upCalcium/calmodulin-dependent protein kinase kinase 2,betaHs.131686NM_172386ABCA917downdownATP-binding cassette, sub-family A (ABC1), member 9Hs.132439NM_173054RELN7downdowndownReelinHs.165258NM_173173NR4A22downNuclear receptor subfamily 4, group A, member 2Hs.31422NM_173468MOBKL1A4upMOB1, Mps One Binder kinase activator-like 1A (yeast)Hs.513424NM_173501LOC14617416upHypothetical protein LOC146174Hs.420244NM_173642MGC478161upHypothetical protein MGC47816Hs.323482NM_173680MGC335847upHypothetical protein MGC33584Hs.418198NM_173797PAPD45downdownPAP associated domain containing 4Hs.356697NM_173827FLJ389914upHypothetical protein FLJ38991Hs.121663NM_173848LOC1380468upHypothetical protein LOC138046Hs.436405NM_174855IDH3B20;downIsocitrate dehydrogenase 3 (NAD+) beta6; 1Hs.4295NM_174871PSMD1217downProteasome (prosome, macropain) 26S subunit, non-ATPase, 12Hs.355606NM_174909MGC23909upHypothetical protein MGC23909Hs.534579NM_174923MGC319679downHypothetical protein MGC31967Hs.321709NM_175567P2RX412; 9upPurinergic receptor P2X, ligand-gated ion channel, 4Hs.406062NM_175614NDUFA1119upupNADH dehydrogenase (ubiquinone) 1 alpha subcomplex,11, 14.7 kDaHs.251673NM_175849DNMT3B20upDNA (cytosine-5-)-methyltransferase 3 betaHs.448218NM_175885MGC3384611downHypothetical protein MGC33846Hs.203NM_176875CCKBR11downCholecystokinin B receptorHs.287636NM_177963SYT1211downSynaptotagmin XIIHs.425383NM_178124CXorf40X; 12upChromosome X open reading frame 40Hs.471917NM_178579PSMF120upProteasome (prosome, macropain) inhibitor subunit 1(PI31)Hs.63236NM_181465MRPL551upMitochondrial ribosomal protein L55Hs.464697NM_181483C18orf118upChromosome 18 open reading frame 1Hs.11900NM_181485ZGPAT20downZinc finger, CCCH-type with G patch domainHs.433422NM_181716PRR6upupupProline rich 6Hs.126137NM_181866BACH1upBrain acyl-CoA hydrolaseHs.479853NM_182472EPHA54upupEPH receptor A5Hs.116567NM_182491LOC906377upHypothetical protein LOC90637Hs.437336NM_182523MGC615713upHypothetical protein MGC61571Hs.200668NM_182661CERK22upCeremide kinaseHs.529735NM_182662AADAT4downAminoadipate aminotransferaseHs.497579NM_182665RASSF51upRas association (RalGDS/AF-6) domain family 5Hs.207157NM_182703LOC34809415downHypothetical protein LOC348094Hs.310537NM_182734PLCB120upPhospholipase C, beta 1 (phosphoinositide-specific)Hs.472101NM_182797PLCB420upPhospholipase C, beta 4Hs.380021NM_182931MLL5upMyeloid/lymphoid or mixed-lineage leukemia 5 (trithoraxhomolog, Drosophila)Hs.414099NM_183010TNRC56; 20downupTrinucleotide repeat containing 5Hs.446240NM_183048PRKCBP120upupProtein kinase C binding protein 1Hs.109087NM_183075CYP2U14downCytochrome P450, family 2, subfamily U, polypeptide 1Hs.298651NM_183236RAB27A15downRAB27a, member RAS oncogene familyHs.203634NM_183239GSTO210upGlutathione S-transferase omega 2Hs.317095NM_183243IMPDH17; 10upupIMP (inosine monophosphate) dehydrogenase 1Hs.191540NM_184042COH18downCohen syndrome 1Hs.291079NM_194279HBLD114upHESB like domain containing 1Hs.268668NM_194285FLJ3944111downHypothetical protein FLJ39441Hs.465337NM_194449PLEKHE118downPleckstrin homology domain containing, family E (withleucine rich repeats) member 1Hs.274479NM_197972NME71upupupupNon-metastatic cells 7, protein expressed in (nucleoside-diphosphate kinase)Hs.149500NM_198038NUDT94upNudix (nucleoside diphosphate linked moiety X)-typemotif 9Hs.76662NM_198045ZDHHC1610upupZinc finger, DHHC domain containing 16Hs.465838NM_198058ZNF26619downdownZinc finger protein 266Hs.20521NM_198318HRMT1L219upHMT1 hmRNP methyltransferase-like 2 (S. cerevisiae)Hs.494155NM_198394C9orf859upChromosome 9 open reading frame 85Hs.446414NM_198793CD473upCD47 antigen (Rh-related antigen, integrin-associatedsignal transducer)Hs.71941NM_198867MGC15677downHypothetical protein MGC15677Hs.515032NM_199054MKNK219downMAP kinase interacting serine/threonine kinase 2Hs.449009NM_199121WARP1downVon Willebrand factor A domain-related proteinHs.528335NM_199138FLJ2547713downHypothetical protein FLJ25477Hs.274959NM_199167CLUL118upClusterin-like 1 (retinal)Hs.517155NM_199170TMEPAI20upTransmembrane, prostate androgen induced RNAHs.13645NM_199297THY28upThymocyte protein thy28Hs.13645NM_199298THY2811; 1updownThymocyte protein thy28Hs.113919NM_199342LOC3749691upHypothetical protein LOC374969Hs.188746NM_199355ADAMTS1816upA disintegrin-like and metalloprotease (reprolysin type)with thrombospondin type 1 motif, 18Hs.459362NM_199414PRC115downProtein regulator of cytokinesis 1Hs.351851NM_199461NANOS110upNanos homolog 1 (Drosophila)Hs.18128NM_199513C20orf4420downChromosome 20 open reading frame 44Hs.16803NM_201412LUC7L16; 8; 5upLUC7-like (S. cerevisiae)Hs.220950NM_201559FOXO3A6upupupForkhead box O3AHs.259750NM_201998SF111upSplicing factor 1Hs.475848NM_202758HSRTSBETA18downRTS beta proteinHs.434406NM_203282ZNF53919downZinc finger protein 539Hs.64004NM_203406LOC1533645downSimilar to metallo-beta-lactamase superfamily proteinHs.417029NM_203415DERP617; 11upS-phase 2 proteinHs.473838NM_203433DSCR221downDown syndrome critical region gene 2Hs.508266NM_203495COMMD613upupCOMM domain containing 6Hs.20013NM_207170P291downGCIP-interacting protein p29Hs.369763NM_207311LOC9255812downHypothetical protein LOC92558Hs.306423NM_207357LOC3395241downHypothetical protein LOC339524Hs.203717NM_212474FN12downFibronectin 1Hs.153752NM_212530CDC25B20upCell division cycle 25BHs.76122R058101downTranscribed locus, moderately similar to NP_055301.1neuronal thread protein AD7c-NTP [Homo sapiens]Hs.283401U22030CYP2A719downCytochrome P450, family 2, subfamily A, polypeptide 7Hs.292156U32331DKK311upDickkopf homolog 3 (Xenopus laevis)Hs.174312U69193TLR49downToll-like receptor 4Hs.209226U85992BMPER7upBMP-binding endothelial regulator precursor proteinHs.465529XM_028067MIDN19downdownMidnolinHs.534513XM_0282174upupHypothetical LOC90024Hs.311363XM_034594downData not foundHs.18564XM_035299ZSWIM65downZinc finger, SWIM domain containing 6Hs.292575XM_035863downData not foundHs.476164XM_038288ZCCHC111downZinc finger, CCHC domain containing 11Hs.314436XM_038999updownData not foundHs.471504XM_041126KIAA14862upKIAA1486 proteinHs.225974XM_043493SV2C5downSynaptic vesicle glycoprotein 2CHs.184736XM_043653BEXL1xdownBrain expressed X-linked-like 1Hs.387336XM_045423upData not foundHs.472285XM_046600KIAA127220downKIAA1272 proteinHs.298382XM_051197KIAA100516upKIAA1005 proteinHs.534526XM_057296LOC1160643downHypothetical protein LOC116064Hs.187636XM_058513LRRK212downLeucine-rich repeat kinase 2Hs.220594XM_0594923downHypothetical LOC131076Hs.380923XM_085929MEIS319upMeis1, myeloid ecotropic viral integration site 1 homolog3 (mouse)Hs.335413XM_08687922upHypothetical LOC150371Hs.169863XM_087089WDR432upWD repeat domain 43HS.180663XM_166451upData not foundHS.423725XM_168302downData not foundHS.196647XM_171054KIAA05273downKIAA0527 proteinHs.162902XM_173173AOF16upAmine oxidase (flavin containing) domain 1Hs.441783XM_290629C14orf7814; 12downChromosome 14 open reading frame 78Hs.132497XM_290941PRNPIP1upPrion protein interacting proteinHS.165762XM_291142FCHO25downFCH domain only 2Hs.29068XM_291277DKFZp761P04238downHypothetical protein DKFZp761P0423Hs.124128XM_291326KIAA2022upKIAA2022 proteinHs.380081XM_370575FBXL1510; 12upF-box and leucine-rich repeat protein 15Hs.9587XM_370878KIAA200215downKIAA2002 proteinHs.410889XM_371074DKFZP564D16617upPutative ankyrin-repeat containing proteinHs.445218XM_371116MYO5B18upMyosin VBHs.288164XM_371614FLJ107073upHypothetical protein FLJ10707Hs.136235XM_371760LOC1160685downHypothetical protein LOC116068Hs.458358XM_371844TSPYL16downTSPY-like 1Hs.136398XM_372124ZCCHC69upZinc finger, CCHC domain containing 6Hs.149940XM_3721289upSimilar to Osteotesticular phosphatase; protein tyrosinephosphatase receptor type V; protein tyrosine phosphatasereceptor type W; protein tyrosine phosphatase, receptortype, VHs.532698XM_373630LOC14584215downHypothetical protein LOC145842Hs.97540XM_3740022upHypothetical gene supported by BC032913; BC048425Hs.389638XM_3743177downHypothetical gene supported by AL713796Hs.447579XM_375527LOC33929018upHypothetical protein LOC339290Hs.408708XM_375553downdownData not foundHs.35524XM_375604downupData not foundHs.172884XM_375633SLC8A219upSolute carrier family 8 (sodium-calcium exchanger),member 2Hs.416553XM_3757141upSimilar to RIKEN cDNA 1700025K23Hs.474836XM_376010LOC38759322downTPTE/TPIP pseudogeneHs.502948XM_376043upSimilar to RIKEN cDNA 2310016E02Hs.301283XM_376193upData not foundHs.113912XM_376350RAPGEF24downdownRap guanine nucleotide exchange factor (GEF) 2Hs.148956XM_376436LOC1344665downHypothetical protein LOC134466Hs.520638XM_376567KIAA18567downKIAA1856 proteinHs.308710XM_376680KIAA17187upKIAA1718 proteinHs.23133XM_378178MGC991319upupupHypothetical protein MGC9913Hs.503862XM_37830911upHypothetical LOC399951Hs.434271XM_37870617upHypothetical LOC400621Hs.477007XM_379203LOC3488013upHypothetical protein LOC348801Hs.446474XM_379250upHypothetical gene supported by BC038466; BC062790Hs.432656XM_379438LOC2857406upHypothetical protein LOC285740Hs.489988XM_379923downData not foundHs.494204XM_495929DKFZp434N203012upHypothetical protein DKFZp434N2030Hs.17250XM_495935MGC476712upupupupHypothetical protein MGC4767Hs.435761XM_496395FLJ344331downHypothetical protein FLJ34433Hs.143840XM_496525downData not foundHs.99488XM_496588LOC1303552upHypothetical protein LOC130355Hs.428360XM_496681KIAA19824upKIAA1982 proteinHs.148988XM_496999KIAA16888downKIAA1688 proteinHs.112622XM_4976791upSimilar to Laminin receptor 1Hs.528187XM_4989172downHypothetical gene supported by AK096649Hs.329512XM_4989553upupHypothetical gene supported by BC034933; BC068085Hs.171132XM_4990085downHypothetical gene supported by AK124699Hs.31917XM_499048C6orf1766downChromosome 6 open reading frame 176Hs.308222XM_499514LOC4013217downHypothetical LOC401321Hs.21925Z399958downdownTranscribed locusHs.22697Z4002510downTranscribed locus











TABLE 2








LocusLink ID
UniGene ID
Symbol















GO: 0050874 organismal physiological


process









10175
Hs.294603
CNIH


10560
Hs.30246
SLC19A2


11255
Hs.251399
HRH3


2009
Hs.12451
EML1


28639
Hs.449451
TRBC1


3437
Hs.47338
IFIT3


4690
Hs.477693
NCK1


4929
Hs.165258
NR4A2


501
Hs.483239
ALDH7A1


641
Hs.169348
BLM


9308
Hs.484703
CD83







GO: 0058550 eukaryotic translation initiation


factor 2 complex









26523
Hs.22867
EIF2C1


83939
Hs.378808
eIF2A


8894
Hs.429180
EIF2S2







GO: 0005739 mitochondrion









10105
Hs.381072
PPIF


10730
Hs.499145
YME1L1


126328
Hs.406062
NDUFA11


1891
Hs.196176
ECH1


26292
Hs.370040
MYCBP


27429
Hs.115721
PRSS25


285521
Hs.356697
FLJ38991


374291
Hs.211914
NDUFS7


38
Hs.232375
ACAT1


4129
Hs.46732
MAOB


4726
Hs.408257
NDUFS6


51103
Hs.106529
NDUFAF1


51373
Hs.44298
MRPS17


51629
Hs.514216
CGI-69


518
Hs.429
ATP5G3


5188
Hs.119316
PET112L


53343
Hs.149500
NUDT9


56997
Hs.118241
CABC1


617
Hs.471401
BCS1L


6390
Hs.465924
SDHB


65993
Hs.157160
MRPS34


7284
Hs.12084
TUFM


84134
Hs.321653
FLJ12770


84693
Hs.94949
MCEE


84701
Hs.277101
COX4I2
















TABLE 3










GO: 0005622 intracellular










LocusLink ID
UniGene ID
Symbol
Description













50
Hs.474982
ACO2
Aconitase 2, mitochondrial


518
Hs.429
ATP5G3
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c





(subunit 9) isoform 3


9331
Hs.464848
B4GALT6
UDP-Gal: betaGlcNAc beta 1,4-galactosyltransferase, polypeptide 6


617
Hs.471401
BCS1L
BCS1-like (yeast)


665
Hs.131226
BNIP3L
BCL2/adenovirus E1B 19 kDA interacting protein 3-like


57128
Hs.387755
C6orf149
Chromosome 6 open reading frame 149


8697
Hs.153546
CDC23
CDC23 (cell division cycle 23, yeast, homolog)


8941
Hs.158460
CDK5R2
Cyclin-dependent kinase 5, regulatory subunit 2 (p39)


1108
Hs.162233
CHD4
Chromodomain helicase DNA binding protein 4


51340
Hs.171342
CRNKL1
Crn, crooked neck-like 1 (Drosophila)


1500
Hs.166011
CTNND1
Catenin (cadherin-associated protein), delta 1


1528
Hs.465413
CYB5
Cytochrome b-5


1602
Hs.129452
DACH1
Dachshund homolog 1 (Drosophila)


1656
Hs.408461
DDX6
DEAD (Asp-Glu-Ala-Asp) box polypeptide 6


9162
Hs.242947
DGKI
Diacylglycerol kinase, iota


1783
Hs.369068
DNCLI2
Dynein, cytoplasmic, light intermediate polypeptide 2


54550
Hs.140950
EFCBP2
EF hand calcium binding protein 2


83939
Hs.378808
eIF2A
Eukaryotic translation initiation factor (eIF) 2A


317649
Hs.476782
EIF4E3
Eukaryotic translation initiation factor 4E member 3


9844
Hs.304578
ELMO1
Eugulfment and cell motility 1 (ced-12 homolog, C. elegans)


30001
Hs.525339
ERO1L
ERO1-like (S. cerevisiae)


2135
Hs.357637
EXTL2
Exostoses (multiple)-like 2


23014
Hs.159699
FBXO21
F-box protein 21


55634
Hs.444269
FLJ20344
Hypothetical protein FLJ20344


2309
Hs.220950
FOXO3A
Forkhead box O3A


29997
Hs.421907
GLTSCR2
Glioma tumor suppressor candiate region gene 2


56850
Hs.109929
GRIPAP1
GRIP1 associated protein 1


119391
Hs.203634
GSTO2
Glutathione S-transferase omega 2


57801
Hs.154029
HES4
Hairy and enhancer of split 4 (Drosophila)


3148
Hs.434953
HMGB2
High-mobility group box 2


3423
Hs.303154
IDS
Iduronate 2-sulfatase (Hunter syndrome)


122953
Hs.196482
JDP2
Jun dimerization protein 2


10656
Hs.444558
KHDRBS3
KH domain containing, RNA binding, signal transduction associated 3


57677
Hs.35524
KIAA1559
Mouse zinc finger protein 14-like


3799
Hs.327736
KIF5B
Kinesin family member 5B


8609
Hs.471221
KLF7
Kruppel-like factor 7 (ubiquitous)


55915
Hs.224282
LANCL2
LanC lantibiotic synthetase component C-like 2 (bacterial)


51176
Hs.125132
LEF1
Lymphoid enhancer-binding factor 1


5641
Hs.18069
LGMN
Legumain


3998
Hs.465295
LMAN1
Lectin, mannose-binding, 1


4005
Hs.34560
LMO2
LIM domain only 2 (rhombotin-like 1)


6218
Hs.294145
LOC133957
Similar to RIKEN cDNA 0610011N22


93349
Hs.471582
LOC93349
Hypothetical protein BC004921


27258
Hs.111632
LSM3
LSM3 homolog, U6 small nuclear RNA associated (S. cerevisiae)


4129
Hs.46732
MAOB
Monoamine oxidase B


51257
Hs.445113
MARCH-II
Membrane-associated RING-CH protein II


10445
Hs.25313
MCRS1
Microspherule protein 1


1072
Hs.25313
MCRS1
Microspherule protein 1


4205
Hs.268675
MEF2A
MADS box transcription enhancer factor 2, polypeptide A (myocyte





enhancer factor 2A)


57534
Hs.140903
MIB1
Mindbomb homolog 1 (Drosophila)


28998
Hs.333823
MRPL13
Mitochondrial ribosomal protein L13


9801
Hs.44024
MRPL19
Mitochondrial ribosomal protein L19


63931
Hs.247324
MRPS14
Mitochondrial ribosomal protein S14


4664
Hs.107474
NAB1
NGFI-A binding protein 1 (EGR1 binding protein 1)


126328
Hs.406062
NDUFA11
NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 11, 14.7 kDa


57520
Hs.314436
NEDL2
NEDD4-related E3 ubiquitin ligase NEDL2


84656
Hs.387255
N-PAC
Cytokine-like nuclear factor n-pac


4925
Hs.128686
NUCB2
Nucleobindin 2


51449
Hs.278627
PCYOX1
Prenylcysteine oxidase 1


9468
Hs.132794
PCYT1B
Phosphate cytidylyltransferase 1, choline, beta isoform


5281
Hs.468415
PIGF
Phosphatidylinositol glycan, class F


5311
Hs.181272
PKD2
Polycystic kidney disease 2 (autosomal dominant)


11128
Hs.436896
POLR3A
Polymerase (RNA) III (DNA directed) polypeptide A, 155 kDa


5464
Hs.437403
PP
Pyrophosphatase (inorganic)


25865
Hs.466987
PRKD2
Protein kinase D2


8559
Hs.161181
PRPF18
PRP18 pre-mRNA processing factor 18 homology (yeast)


201161
Hs.433422
PRR6
Proline rich 6


5718
Hs.4295
PSMD12
Proteasome (prosome, macropain) 26S subunit, non-ATPase, 12


9491
Hs.471917
PSMF1
Proteasome (prosome, macropain) inhibitor subunit 1 (PI31)


10567
Hs.11417
RABAC1
Rab acceptor 1 (prenylated)


5994
Hs.24422
RFXAP
Regulatory factor X-associated protein


51320
Hs.465144
RKHD2
Ring finger and KH domain containing 2


57484
Hs.480825
RNF150
Ring finger protein 150


6096
Hs.494178
RORB
RAR-related orphan receptor B


6165
Hs.529631
RPL35A
Ribosomal protein L35a


25939
Hs.472630
SAMHD1
SAM domain and HD domain 1


79048
Hs.59804
SECISBP2
SECIS binding protein 2


23451
Hs.471011
SF3B1
Splicing factor 3b, subunit 1, 155 kDa


23013
Hs.270499
SHARP
SMART/HDAC1 associated repressor protein


6732
Hs.443861
SRPK1
SFRS protein kinase 1


10618
Hs.14894
TGOLN2
Trans-golgi network protein 2


30000
Hs.416049
TNPO2
Transportin 2 (importin 3, karyopherin beta 2b)


80263
Hs.301526
TRIM45
Tripartite motif-containing 45


55109
Hs.213393
VG5Q
Angiogenic factor VG5Q


51564
Hs.269577
VPS16
Protein tyrosine phosphatase, receptor type, A


64601
Hs.269577
VPS16
Protein tyrosine phosphatase, receptor type, A


8976
Hs.143728
WASL
Wiskott-Aldrich syndrome-like


29799
Hs.517436
YPEL1
Yippee-like 1 (Drosophila)


7528
Hs.388927
YY1
YY1 transcription factor


146198
Hs.461074
ZFP90
Zinc finger protein 90 homolog (mouse)


























TABLE 4












Lymphocyte


Fold Change









Direction


Agree
Fold Change
Brain Direction





Chromosomal
(Schizophrenia/


Brain/
Agree Brain/
(Schizophrenia/


UniGene ID
Acc
Gene Symbol
Location
Control)
Fold change
p-value
Lymphocyte
Lymphocyte
Control)
Gene Name

























Hs.232375
NM_000019
ACAT1
11q22.3-q23.1
UP
1.15
0.024
yes
yes
UP
Acetyl-Coenzyme A












acetyltransferase 1












(acetoacetyl Coenzyme A thiolase)


Hs.130712
NM_024876
ADCK4
19q13.2
DOWN
−1.11
0.042
No
No
UP
AarF domain












containing kinase 4


Hs.207776
NM_000027
AGA
4q32-q33
DOWN
−1.11
0.038
yes
yes
DOWN
Aspartylglucosaminidase


Hs.368486
NM_001649
APXL
Xp22.3
DOWN
−1.14
0.024
No
No
UP
Apical protein-like












(Xenopus laevis)


Hs.126137
NM_181866
BACH
1p36.31-p36.11
DOWN
−1.11
0.049
No
No
UP
Brain acyl-CoA












hydrolase


Hs.293753
NM_032515
BOK
2q37.3
DOWN
−1.12
0.020
No
No
UP
BCL2-related












ovarian killer


Hs.288981
NM_025152
C14orf127
14q12
DOWN
−1.16
0.019
No
No
UP
Chromosome 14












open reading frame 127


Hs.443789
NM_024581
C6orf60
6q22.31
UP
1.46
0.018
No
No
DOWN
Chromosome 6 open












reading frame 60


Hs.532296
NM_017998
C9orf40
9q21.31
UP
1.13
0.036
yes
yes
UP
Chromosome 9 open












reading frame 40


Hs.297343
NM_172226
CAMKK2
12q24.2
DOWN
−1.15
0.030
No
No
UP
Calcium/calmodulin-












dependent protein












kinase kinase 2, beta


Hs.297343
NM_172226
CAMKK2
12q24.2
DOWN
−1.13
0.018
No
No
UP
Calcium/calmodulin-












dependent protein












kinase kinase 2, beta


Hs.474797
NM_007061
CDC42EP1
22q13.1
DOWN
−1.10
0.008
No
No
UP
CDC42 effector












protein (Rho GTPase binding) 1


Hs.249129
NM_001279
CIDEA
18p11.21
DOWN
−1.16
0.016
No
No
UP
Cell death-inducing












DFFA-like effector a


Hs.29549
NM_016511
CLEC1
12p13.31
DOWN
−1.14
0.025
yes
yes
DOWN
C-type lectin-like












receptor-1


Hs.270437
NM_031361
COL4A3BP
5q13.3
UP
1.27
0.047
yes
yes
UP
Collagen, type IV,












alpha 3 (Goodpasture












antigen) binding protein


Hs.464422
NM_130386
COLEC12
18pter-p11.3
DOWN
−1.15
0.044
yes
yes
DOWN
Collectin sub-family












member 12


Hs.330384
NM_014325
CORO1C
12q24.1
DOWN
−1.17
0.040
No
No
UP
Coronin, actin












binding protein, 1C


Hs.45127
NM_006574
CSPG5
3p21.3
UP
1.16
0.023
No
No
DOWN
Chondroitin sulfate












proteoglycan 5












(neuroglycan C)


Hs.304682
NM_000099
CST3
20p11.21
UP
1.74
0.026
No
No
DOWN
Cystatin C (amyloid












angiopathy and












cerebral hemorrhage)


Hs.26704
NM_014608
CYFIP1
15q11
DOWN
−1.42
0.010
yes
yes
DOWN
Cytoplasmic FMR1












interacting protein 1


Hs.283401
U22030
CYP2A7
19q13.2
DOWN
−1.13
0.006
yes
yes
DOWN
Cytochrome P450,












family 2, subfamily












A, polypeptide 7


Hs.12451
AF035276
EML1
14q32
DOWN
−1.11
0.047
No
No
UP
Echinoderm












microtubule












associated protein like 1


Hs.104925
NM_003633
ENC1
5q12-q13.3
UP
1.45
0.046
yes
yes
UP
Ectodermal-neural












cortex (with BTB-like domain)


Hs.104925
NM_003633
ENC1
5q12-q13.3
UP
1.42
0.033
yes
yes
UP
Ectodermal-neural












cortex (with BTB-like domain)


Hs.306251
NM_001982
ERBB3
12q13
DOWN
−1.45
0.015
yes
yes
DOWN
V-erb-b2












erythroblastic leukemia viral












oncogene homolog 3 (avian)


Hs.438695
AK094876
FKBP11
12q13.12
UP
1.34
0.041
No
No
DOWN
FK506 binding












protein 11, 19 kDa


Hs.443529
NM_019607
FLJ11267
8q13.1
DOWN
−1.13
0.040
yes
yes
DOWN
Hypothetical protein












FLJ11267


Hs.147836
NM_017768
FLJ20331
1p31.2
UP
1.38
0.034
No
No
DOWN
Hypothetical protein












FLJ20331


Hs.413137
NM_001680
FXYD2
11q23
DOWN
−1.19
0.023
No
No
UP
FXYD domain












containing ion












transport regulator 2


Hs.473648
NM_000819
GART
21q22.1
UP
1.32
0.029
yes
yes
UP
Phosphoribosylglycinamide












formyltransferase,












phosphoribosylglycinamide synthetase,












phosphoribosylaminoimidazole synthetase


Hs.522418
NM_001003722
GLE1L
9q34.13
UP
1.28
0.045
yes
yes
UP
GLE1 RNA export












mediator-like (yeast)


Hs.522418
NM_001003722
GLE1L
9q34.13
DOWN
−1.12
0.015
No
No
UP
GLE1 RNA export












mediator-like (yeast)


Hs.430425
NM_002074
GNB1
1p36.33
DOWN
−1.20
0.034
No
No
UP
Guanine nucleotide












binding protein (G












protein), beta












polypeptide 1


Hs.155090
AL117471
GNB5
15q21.1
DOWN
−1.13
0.030
No
No
UP
Guanine nucleotide












binding protein (G












protein), beta 5


Hs.155090
NM_006578
GNB5
15q21.1
DOWN
−1.10
0.005
No
No
UP
Guanine nucleotide












binding protein (G












protein), beta 5


Hs.198612
NM_005458
GPR51
9q22.1-q22.3
UP
1.17
0.044
yes
yes
UP
G protein-coupled












receptor 51


Hs.445066
AI499801
GRIN2B
12p12
DOWN
−1.16
0.034
yes
yes
DOWN
Glutamate receptor,












ionotropic, N-methyl D-aspartate 2B


Hs.12126
NM_018487
HCA112
7q36.1
DOWN
−1.34
0.007
yes
yes
DOWN
Hepatocellular












carcinoma-associated antigen 112


Hs.278635
NM_016648
HDCMA18P
4q26
DOWN
−1.40
0.046
yes
yes
DOWN
HDCMA18P protein


Hs.35804
D25215
HERC3
4q21
DOWN
−1.13
0.012
No
No
UP
Hect domain and












RLD 3


Hs.380250
AK094968
IF116
1q22
UP
1.19
0.038
No
No
DOWN
Interferon, gamma-












inducible protein 16


Hs.430551
NM_003870
IQGAP1
15q26.1
UP
1.24
0.021
No
No
DOWN
IQ motif containing












GTPase activating protein 1


Hs.6396
NM_006694
JTB
1q21
UP
1.53
0.048
yes
yes
UP
Jumping












translocation breakpoint


Hs.21703
NM_012281
KCND2
7q31
UP
1.97
0.038
yes
yes
UP
Potassium voltage-












gated channel, Shal-












related subfamily, member 2


Hs.408960
NM_002241
KCNJ10
1q22-q23
DOWN
−1.14
0.005
yes
yes
DOWN
Potassium inwardly-












rectifying channel,












subfamily J,












member 10


Hs.32505
NM_004981
KCNJ4
22q13.1
DOWN
−1.10
0.028
yes
yes
DOWN
Potassium inwardly-












rectifying channel,












subfamily J, member 4


Hs.2785
NM_000422
KRT17
17q12-q21
DOWN
−1.15
0.035
yes
yes
DOWN
Keratin 17


Hs.23748
NM_001290
LDB2
4p16
DOWN
−1.20
0.011
No
No
UP
LIM domain












binding 2


Hs.352614
AF007155
LOC254531
15q13.2
UP
1.95
0.036
No
No
DOWN
PLSC domain












containing protein


Hs.47649
NM_020166
MCCC1
3q27
DOWN
−1.16
0.022
yes
yes
DOWN
Methylcrotonoyl-












Coenzyme A












carboxylase 1 (alpha)


Hs.535659
BC002458
MCM3AP
21q22.3
DOWN
−1.16
0.046
No
No
UP
MCM3












minichromosome maintenance












deficient 3 (S. cerevisiae)












associated protein


Hs.460217
NM_153208
MGC35048
16p13.11
DOWN
−1.18
0.013
yes
yes
DOWN
Hypothetical protein












MGC35048


Hs.21213
NM_000259
MYO5A
15q21
DOWN
−1.14
0.027
No
No
UP
Myosin VA (heavy












polypeptide 12, myoxin)


Hs.472185
NM_004552
NDUFS5
1p34.2-p33
UP
1.21
0.035
yes
yes
UP
NADH












dehydrogenase












(ubiquinone) Fe—S












protein 5, 15 kDa












(NADH-coenzyme Q reductase)


Hs.211914
NM_024407
NDUFS7
19p13.3
DOWN
−1.11
0.039
No
No
UP
NADH












dehydrogenase












(ubiquinone) Fe—S












protein 7, 20 kDa












(NADH-coenzyme Q reductase)


Hs.459255
AI935701
NTRK3
15q25
DOWN
−1.14
0.040
No
No
UP
Neurotrophic












tyrosine kinase,












receptor, type 3


Hs.459255
AI935701
NTRK3
15q25
DOWN
−1.12
0.021
No
No
UP
Neurotrophic












tyrosine kinase, receptor, type 3


Hs.435714
NM_002576
PAK1
11q13-q14
UP
1.23
0.030
yes
yes
UP
P21/Cdc42/Rac1-












activated kinase 1












(STE20 homolog, yeast)


Hs.503584
AI638679
PANX1
11q21
UP
1.27
0.036
yes
yes
UP
Pannexin 1


Hs.278627
NM_016297
PCYOX1
2p13.3
UP
1.14
0.033
yes
yes
UP
Prenylcysteine












oxidase 1


Hs.481819
AK021922
PDZK3
5p13.3
UP
1.24
0.009
No
No
DOWN
PDZ domain












containing 3


Hs.468415
NM_002643
PIGF
2p21-p16
DOWN
−1.12
0.020
No
No
UP
Phosphatidylinositol












glycan, class F


Hs.466848
NM_006905
PSG1
19q13.2
DOWN
−1.11
0.036
No
No
UP
Pregnancy specific












beta-1-glycoprotein 1


HS.413801
NM_014614
PSME4
2p16.3
DOWN
−3.66
0.003
No
No
UP
Proteasome












(prosome,












macropain) activator subunit 4


Hs.446429
NM_000954
PTGDS
9q34.2-q34.3
DOWN
−2.93
0.016
yes
yes
DOWN
Prostaglandin D2












synthase 21 kDa (brain)


Hs.114062
NM_014241
PTPLA
10p14-p13
DOWN
−1.12
0.034
No
No
UP
Protein tyrosine












phosphatase-like (proline instead of catalytic












arginine), member a


Hs.434375
BI820698
PTPRB
12q15-q21
UP
1.32
0.019
No
No
DOWN
Protein tyrosine












phosphatase, receptor type, B


Hs.296169
NM_004578
RAB4A
1q42-q43
UP
1.13
0.031
yes
yes
UP
RAB4A, member












RAS oncogene family


Hs.411488
NM_138290
RPIB9
7q21.13
DOWN
−1.16
0.001
No
No
UP
Rap2-binding












protein 9


Hs.374588
NM_000985
RPL17
18q21
DOWN
−1.17
0.012
No
No
UP
Ribosomal protein












L17


Hs.374588
NM_000985
RPL17
18q21
DOWN
−1.13
0.026
No
No
UP
Ribosomal protein












L17


Hs.465924
NM_003000
SDHB
1p36.1-p35
DOWN
−1.12
0.038
No
No
UP
Succinate












dehydrogenase complex, subunit B, iron sulfur (Ip)


Hs.146804
NM_006717
SPIN
9q22.1-q22.3
DOWN
−1.32
0.039
No
No
UP
Spindlin


Hs.237825
NM_006947
SRP72
4q11
DOWN
−1.13
0.044
No
No
UP
Signal recognition












particle 72 kDa


Hs.102735
NM_012446
SSBP2
5q14.1
UP
1.22
0.050
No
No
DOWN
Single-stranded












DNA binding protein 2


Hs.12409
NM_001048
SST
3q28
DOWN
−1.15
0.045
yes
yes
DOWN
Somatostatin


Hs.482390
NM_003243
TGFBR3
1p33-p32
UP
1.18
0.017
No
No
DOWN
Transforming












growth factor, beta












receptor III (betaglycan, 300 kDa)


Hs.14894
NM_006464
TGOLN2
2p11.2
UP
1.15
0.016
yes
yes
UP
Trans-golgi network












protein 2


Hs.465784
AF026030
TIMM44
19p13.3-p13.2
DOWN
−1.15
0.033
No
No
UP
Translocase of inner












mitochondrial membrane 44 homolog (yeast)


Hs.465784
NM_006351
TIMM44
19p13.3-p13.2
DOWN
−1.15
0.033
No
No
UP
Translocase of inner












mitochondrial membrane 44 homolog (yeast)


Hs.287362
AB046767
TLE3
15q22
DOWN
−1.19
0.009
No
No
UP
Transducin-like












enhancer of split 3












(E(sp1) homolog,













Drosophila)



Hs.287362
NM_005078
TLE3
15q22
DOWN
−1.19
0.009
No
No
UP
Transducin-like enhancer of split 3 (E(sp1)












homolog, Drosophila)


Hs.499145
BC019602
YME1L1
10p14
UP
1.13
0.043
No
No
DOWN
YME1-like 1 (S. cerevisiae)


Hs.172979
NM_003451
ZNF177
19p13.2
DOWN
−1.17
0.021
No
No
UP
Zinc finger protein 177























TABLE 5










Microarray
Q-PCR
Microarray
Q-PCR




Accession
Gene
Fold
Fold
(p-value t-
(p-value
Cytogenetic


Number
Symbol
Change
Change
test)
t-test)
Band
Gene Name






















NM_002110
HCK
2.58
4.71
0.030
0.02
20q11-q12
hemopoietic cell kinase


NM_021005
NR2F2
1.74
4.15
0.047
0.02
15q26.2
nuclear receptor subfamily 2, group F, member 2


NM_017423
GALNT7
1.34
2.81
0.041
0.46
4q34.1
UDP-N-acetyl-alpha-D-









galactosamine:polypeptide N-









acetylgalactosaminyltransferase 7 (GalNAc-T7)


NM_139045
SMARCA2
1.75
2.27
0.028
0.03
9p22.3
SWI/SNF related, matrix associated, actin









dependent regulator of chromatin, subfamily a, member 2


NM_018639
WSB2
1.49
2.18
0.003
0.02
12q24.23
WD repeat and SOCS box containing protein 2


NM_203504
G3BP2
1.28
2.04
0.045
0.07
4q21.21
Ras-GTPase activating protein SH3 domain-









binding protein 2


NM_023927
NS3TP2
1.27
1.75
0.030
0.08
5q23.3
HCV NS3-transactivated protein 2


NM_003816
ADAM9
1.45
1.70
0.024
0.04
8p11.22
a disintegrin and metalloproteinase domain 9









(meltrin gamma)


BQ000126
TMOD3
1.21
1.39
0.031
0.12
15q21.1-q21.2
tropomodulin 3 (ubiquitous)


NM_003100
SNX2
1.43
0.95
0.042
0.45
5q23
sorting nexin 2


AL041379
HERC3
0.89
0.90
0.023
0.34
4q21
hect domain and RLD 3


AA487462
FLJ20637
0.81
0.68
0.014
0.08
4q22.1
hect domain and RLD 6


BM855607
IGJ
0.45
0.63
0.038
0.21
4q21
immunoglobulin J polypeptide, linker protein for









immunoglobulin alpha and mu polypeptides


NM_005214
CTLA4
0.82
0.14
0.036
0.04
2q33
cytotoxic T-lymphocyte-associated protein 4


AV700777
ADH1B
0.89
0.10
0.023
0.02
4q21-q23
alcohol dehydrogenase IB (class I), beta









polypeptide



















TABLE 6








db SNP RS ID
NCBI (Mb)
Cis < 5
Regression (p-value)







rs1318822
170460379
8.6
0.0015


rs1403225
171867785
7.2
0.0158


rs1812424
173851484
5.2
0.0158


rs2119788
175143279
3.9
0.0020


rs723820
177930796
1.1
0.0020


rs723819
177930836
1.1
0.0020


rs1112286
178134007
0.9
0.0037


rs1375749
178157255
0.9
0.0037


rs1902018
179382986
0.3
0.0016


rs722387
179808887
0.8
0.0400


rs1112857
181655677
2.6
0.0400
















TABLE 7










Genes dysregulated in MD, BP, and schizophrenia














UniGene







1
ID
probe set
Acc
Name
Symbol
Direction of Change
















2

text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed



3

text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed


text missing or illegible when filed



4
Hs.530712
Hs.530712_at
NM_017917
Chromosome 14 open
C14orf10
DLPFC-BPD&MDD&SCZ-U






reading frame 10


5
Hs.56294
Hs.56294_at
NM_004794
RAB33A, member
RAB33A
DLPFC-BPD&MDD&SCZ-U






RAS oncogene family


6
Hs.21577
Hs.21577_at
NM_005701
RNA, U transporter 1
RNUT1
DLPFC-BPD&SCZ-U


7
Hs.274479
Hs.274479−_at
NM_197972
Non-metastatic cells 7,
NME7
DLPFC-BPD&SCZ-U






protein expressed in






(nucleoside-






diphosphate kinase)


8
Hs.368486
Hs.368486_at
NM_001649
Apical protein-like
APXL
DLPFC-BPD&SCZ-U






(Xenopus laevis)


9
Hs.468415
Hs.468415_at
NM_002643
Phosphatidylinositol
PIGF
DLPFC-BPD&SCZ-U






glycan, class F


10
Hs.471401
Hs.471401_at
NM_004328
BCS1-like (yeast)
BCS1L
DLPFC-BPD&SCZ-U


11
Hs.502145
Hs.502145_at
NM_006157
NEL-like 1 (chicken)
NELL1
DLPFC-BPD&SCZ-U


12
Hs.514036
Hs.514036_at
NM_006923
Stromal cell-derived
SDF2
DLPFC-BPD&SCZ-U






factor 2


13
Hs.532853
Hs.532853+_at
NM_004146
NADH dehydrogenase
NDUFB7
DLPFC-BPD&SCZ-U






(ubiquinone) 1 beta






subcomplex, 7, 18 kDa


14
Hs.111779
Hs.111779_at
NM_003118
Secreted protein,
SPARC
DLPFC-BPD&MDD&SCZ-D






acidic, cysteine-rich






(osteonectin)


15
Hs.171695
Hs.171695_at
NM_004417
Dual specificity
DUSP1
DLPFC-BPD&MDD&SCZ-D






phosphatase 1


16
Hs.212838
Hs.212838−_at
NM_000014
Alpha-2-macroglobulin
A2M
DLPFC-BPD&MDD&SCZ-D


17
Hs.34560
Hs.34560−_at
NM_005574
LIM domain only 2
LMO2
DLPFC-BPD&MDD&SCZ-D






(rhombotin-like 1)


18
Hs.347270
Hs.347270−_at
NM_033554
Major
HLA-
DLPFC-BPD&MDD&SCZ-D






histocompatibility
DPA1






complex, class II, DP






alpha 1


19
Hs.436568
Hs.436568_at
BC024272
CD74 antigen
CD74
DLPFC-BPD&MDD&SCZ-D






(invariant polypeptide






of major






histocompatibility






complex, class II






antigen-associated)


20
Hs.491582
Hs.491582_at
NM_000931
Plasminogen activator,
PLAT
DLPFC-BPD&MDD&SCZ-D






tissue


21
Hs.534115
Hs.534115_at
NM_006988
A disintegrin-like and
ADAMTS1
DLPFC-BPD&MDD&SCZ-D






metalloprotease






(reprolysin type) with






thrombospondin type 1






motif, 1


22
Hs.17109
Hs.17109_at
NM_004867
Integral membrane
ITM2A
DLPFC-BPD&SCZ-D






protein 2A


23
Hs.485130
Hs.485130_at
K016
Major
HLA
DLPFC-BPD&SCZ-D






histocompatibility
DPB1






complex, class II, DP






beta 1


24
Hs.504877
Hs.504877_at
X69549
Rho GDP dissociation
ARHGDIB
DLPFC-BPD&SCZ-D






inhibitor (GDI) beta


25
Hs.520048
Hs.520048_at
NM_019111
Major
HLA-DRA
DLPFC-BPD&SCZ-D






histocompatibility






complex, class II, DP






alpha1
















TABLE 8










Genes dysregulated in MD, BP, and schizophrenia













Symbol
Name
UniGene ID
AnCg
DLPFC
CB
nAcc





PTGDS
Prostaglandin
Hs.446429


CB-D
nAcc-D


PLAT
Plasminogen activator, tissue
Hs.491582
AnCg-D
DLPFC-D-


ADAMTS1
Disintegrin-like and metalloprotease
Hs.534115

DLPFC-D

nAcc-D
















TABLE 9








Differentially Expressed Genes in Amygdala


˜Passed 4 Analysis Platforms˜


∘ BPD ∘ MDD ∘ SZ












embedded image


















TABLE 10








Differentially Expressed Genes in Amygdala


˜Passed 4 Analysis Platforms˜


∘ BPD ∘ MDD ∘ SZ












embedded image


















TABLE 11








Differentially Expressed Genes in Amygdala


˜Passed 4 Analysis Platforms˜


∘ BPD ∘ MDD ∘ SZ












embedded image






embedded image


















TABLE 12








Differentially Expressed Genes in Amygdala


˜Passed 4 Analysis Platforms˜


∘ BPD ∘ MDD ∘ SZ












embedded image


















TABLE 13










Genes Dysregulated in both SZ and BPD


DLPFC










NAME
SYMBOL
NAME
SYMBOL





Kelch-like 12 (Drosophila)
KLHL12
LIM domain only 2 (rhombotin-like 1)
LMO2


Trinucleotide repeat containing 17
CENTG2
Major histocompatibility complex, class II, DP
HLA-DPA1


Chromosome 14 open reading frame 10
C14orf10
alpha 1


RAB33A, member RAS oncogene family
RAB33A
CD74 antigen (invariant polypeptide of major
CD74


RNA, U transporter 1
RNUT1
histocompatibility complex, class II antigen-


Non-metastatic cells 7, protein expressed in
NME7
associated)


(nucleoside-diphosphate kinase)

Plasminogen activator, tissue
PLAT


Apical protein-like (Xenopus laevis)
APXL
A disintegrin-like and metalloprotease
ADAMTS1


Phosphatidylinositol glycan, class F
PIGF
(reprolysin type) with thrombospondin


BCS1-like (yeast)
BCS1L
type 1 motif, 1


NEL-like 1 (chicken)
NELL1
Integral membrane protein 2A
ITM2A


Stromal cell-derived factor 2
SDF2
Major histocompatibility complex, class II, DP
HLA-DPB1


NADH dehydrogenase (ubiquinone) 1 beta
NDUFB7
beta 1


subcomplex, 7, 18 kDa

Rho GDP dissociation inhibitor (GDI) beta
ARHGDIB


Secreted protein, acidic, cysteine-rich
SPARC
Major histocompatibility complex, class II, DR
HLA-DRA


(osteonectin)

alpha


Dual specificity phosphatase 1
DUSP1
Major histocompatibility complex, class II, DR
HLA-DRA


Alpha-2-macroglobulin
A2M
alpha
















TABLE 14










Li_UP_DOWN_candidates_AND_BPD_26 JAN 06



text missing or illegible when filed



AMY










NM_002781
19q13.2
PSG5
Pregnancy specific beta-1-glycoprotein 5


NM_003998
4q24

text missing or illegible when filed

Nuclear factor of kappa light polypeptide gene enhance


NM_004898
4q12
CLOCK
Clock homolog (mouse)


NM_013263
16q12
BRD7
Bromodomain containing 7







AnCg










NM_001048
3q28
SST
Somatostatin


NM_002093
3q13.3

text missing or illegible when filed

Glycogen synthase kinase 3 beta


NM_003489
21q11.2
NRIP1
Nuclear receptor interacting protein 1


NM_003936
2q35
CDK5R2
Cyclin-dependent kinase 5, regulatory subunit 2 (p39)


NM_013444
Xp11.23-p11.1
UBQLN2
Ubiquilin 2


NM_017993
13q14.11
FLJ10094
Hypothetical protein FLJ10094


NM_020178
17q21
CA10
Carbonic anhydrase X


NM_021952
1p34
ELAVL4
ELAV (embryonic lethal, abnormal vision, Drosophila)-





like 4 (Hu antigen D)







DLPFC










NM_025098
11q13.5
MOGAT2
Monoacylglycerol O-acyltransferase 2







HC







None







UP


AMY










NM_002065
1q31
GLUL
Glutamate-ammonia ligase (glutamine synthetase)





(not in BPD)


NM_000014
12p13.3-p12.3
A2M
Alpha-2-macroglobulin


NM_001004
11p15.5-p15.4
RPLP2
Ribosomal protein, large, P2


NM_001283
7q22.1
AP1S1
Adaptor-related protein complex 1, sigma 1 subunit


NM_004355
5q32
CD74
CD74 antigen (invariant polypeptide of major





histocompatibility complex, class II antigen-





associated)


NM_004368
21q11.1
CNN2
Calponin 2


NM_005617
5q31-q33
RPS14
Ribosomal protein S14


NM_006119
10q24

text missing or illegible when filed

Fibroblast growth factor 8 (androgen-induced)


NM_006870
20p12.1
DSTN
Destrin (actin depolymerizing factor)


NM_080391
1p35
PTP4A2
Protein tyrosine phosphatase type IVA, member 2


NM_153477
Xp11.23-p11.22
UXT
Ubiquitously-expressed transcript







AnCg







None







DLPFC2










NM_000014
12p13.3-p12.3
A2M
Alpha-2-macroglobulin


NM_001004
11p15.5-15.4
RPLP2
Ribosomal protein, large, P2


NM_001013
19q13.4
RPS9
Ribosomal protein S9


NM_001283
7q22.1
AP1S1
Adaptor-related protein complex 1, sigma 1 subunit


NM_002178
12q13
IGFBP6
Insulin-like growth factor binding protein 6


NM_019597
Xq22
HNRPH2
Heterogeneous nuclear ribonucleoprotein H2 (H′)


NM_018584
1p36.12
CaMKIINalpha
Calcium/calmodulin-dependent protein kinase II





inhibitor 1 (not in BPD)







HC










NM_053025
3q21
MYLK
Myosin, light polypeptide kinase


NM_006593
2q24

text missing or illegible when filed

T-box, brain, 1 (not inBPD)








Claims
  • 1. A method for determining whether a subject has or is predisposed for a mental disorder, the method comprising the steps of: (i) obtaining a biological sample from a subject; (ii) contacting the sample with a reagent that selectively associates with a polynucleotide or polypeptide encoded by a nucleic acid that hybridizes under stringent conditions to a nucleotide sequence of Tables 1-14; and (iii) detecting the level of reagent that selectively associates with the sample, thereby determining whether the subject has or is predisposed for a mental disorder.
  • 2. The method of claim 1, wherein the reagent is an antibody.
  • 3. The method of claim 1, wherein the reagent is a nucleic acid.
  • 4. The method of claim 1, wherein the reagent associates with a polynucleotide.
  • 5. The method of claim 4, wherein the reagent selectively associates with a polynucleotide comprising at least one of the SNPs listed in Table 6.
  • 6. The method of claim 1, wherein the regent associates with a polypeptide.
  • 7. The method of claim 1, wherein the level of reagent that associates with the sample is different from a level associated with humans without a mental disorder.
  • 8. The method of claim 1, wherein the biological sample is obtained from amniotic fluid.
  • 9. The method of claim 1, wherein the mental disorder is a psychotic disorder or a mood disorder.
  • 10. The method of claim 1, wherein the sample is derived from brain tissue or lymphocytes.
  • 11. The method of claim 10, wherein the sample is derived from brain tissue.
  • 12. The method of claim 11, wherein the sample is derived from a brain region selected from the group consisting of the anterior cingulate cortex (AnCg), dorsolateral prefrontal cortex (DLPFC), cerebellar cortex (CB), superior temporal gyrus (STG), parietal cortex (PC), nucleus accumbens (nAcc), and amygdala (amy).
  • 13. The method of claim 7, wherein the level of reagent that associates with the sample is higher than a level associated with humans without a mental disorder.
  • 14. The method of claim 9, wherein the psychotic disorder is schizophrenia.
  • 15. The method of claim 9, wherein the mood disorder is major depression disorder or bipolar disorder.
  • 16. A method of identifying a compound for treatment of a mental disorder, the method comprising the steps of: (i) contacting the compound with a polypeptide, the polypeptide encoded by a polynucleotide that hybridizes under stringent conditions to a nucleic acid sequence comprising a nucleotide sequence of Tables 1-14; and (ii) determining the functional effect of the compound upon the polypeptide, thereby identifying a compound for treatment of a mental disorder.
  • 17. The method of claim 16, wherein the contacting step is performed in vitro.
  • 18. The method of claim 16, wherein the polypeptide is expressed in a cell and the cell is contacted with the compound.
  • 19. The method of claim 16, wherein the mental disorder is a psychotic disorder.
  • 20. The method of claim 19, wherein the psychotic disorder is schizophrenia.
  • 21. The method of claim 16, wherein the mental disorder is a mood disorder.
  • 22. The method of claim 19, wherein the psychotic disorder is major depression disorder or bipolar disorder.
  • 23. The method of claim 19, further comprising administering the compound to an animal and determining the effect on the animal.
  • 24. The method of claim 23, wherein the determining step comprises testing the animal's mental function.
  • 25. A method of identifying a compound for treatment of a mental disorder in a subject, the method comprising the steps of: (i) contacting the compound to a cell, the cell comprising a polynucleotide that hybridizes under stringent conditions to a nucleotide sequence of Tables 1-14; and (ii) selecting a compound that modulates expression of the polynucleotide, thereby identifying a compound for treatment of a mental disorder.
  • 26. The method of claim 25, wherein the expression of the polynucleotide is enhanced.
  • 27. The method of claim 25, wherein the expression of the polynucleotide is decreased.
  • 28. The method of claim 25, further comprising administering the compound to an animal and determining the effect on the animal.
  • 29. The method of claim 28, wherein the determining step comprises testing the animal's mental function.
  • 30. The method of claim 25, wherein the mental disorder is a psychotic disorder.
  • 31. The method of claim 30, wherein the psychotic disorder is schizophrenia.
  • 32. The method of claim 25, wherein the mental disorder is a mood disorder.
  • 33. The method of claim 30, wherein the psychotic disorder is major depression disorder or bipolar disorder.
  • 34. A method of treating a mental disorder in a subject, the method comprising the step of administering to the subject a therapeutically effective amount of a compound identified using the method of claim 16 or claim 25.
  • 35. The method of claim 34, wherein the mental disorder is a psychotic disorder or a mood disorder.
  • 36. The method of claim 34, wherein the compound is a small organic molecule.
  • 37. The method of claim 35, wherein the psychotic disorder is schizophrenia.
  • 38. The method of claim 35, wherein the mood disorder is major depression disorder or bipolar disorder.
  • 39. A method of treating mental disorder in a subject, the method comprising the step of administering to the subject a therapeutically effective amount of a polypeptide, the polypeptide encoded by a polynucleotide that hybridizes under stringent conditions to a nucleotide sequence of Tables 1-14.
  • 40. The method of claim 39, wherein the mental disorder is schizophrenia, major depression disorder, or bipolar disorder.
  • 41. A method of treating mental disorder in a subject, the method comprising the step of administering to the subject a therapeutically effective amount of a nucleic acid, wherein the nucleic acid hybridizes under stringent conditions to a nucleotide sequence of Tables 1-14.
  • 42. The method of claim 41, wherein the mental disorder is schizophrenia, major depression disorder, or bipolar disorder.
  • 43. A method for determining whether a subject has or is predisposed for a mental disorder, the method comprising the steps of: (i) contacting the tissue of one or regions of the subject's brain with a detectably labeled molecule that selectively binds to a gene listed in any of Tables 1-14; (ii) visualizing the distribution of the detectably labeled molecule in the brain tissue; and (iii) correlating the distribution of the detectably labeled molecule with the presence of or predisposition for schizophrenia in the subject.
  • 44. The method of claim 43 wherein said one or more regions are selected from the group consisting of the anterior cingulate cortex (AnCg), dorsolateral prefrontal cortex (DLPFC), cerebellar cortex (CB), superior temporal gyrus (STG), parietal cortex (PC), nucleus accumbens (nAcc), and amygdala (amy).
  • 45. The method of claim 43 wherein said labeled molecule is an antisense RNA molecule.
  • 46. The method of claim 43 wherein said contacting occurs in vivo.
CROSS-REFERENCES TO RELATED APPLICATIONS

The present application claims priority to U.S. Ser. No. 60/667,299, filed Mar. 31, 2005, and U.S. Ser. No. 60/776,103, filed Feb. 22, 2006, herein incorporated by reference in their entirety.

Provisional Applications (2)
Number Date Country
60667299 Mar 2005 US
60776103 Feb 2006 US