Compositions and methods for diagnosis and treating mood disorders

Abstract
The present invention provides methods for diagnosing mental disorders such as mood disorders, including bipolar disorder I and II and major depression; 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

Clinical depression, including both bipolar disorders and major depression disorders, is a major public health problem, affecting an estimated 9.5% of the adult population of the United States each year. While it has been hypothesized that mental disorders, including mood disorders such as major depression and bipolar disorder as well as psychotic disorders such as schizophrenia, have complex 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, 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 mood disorders such as bipolar disorders (BP) and major depression disorders (MDD), the inventors of the present application have used DNA microarrays to study expression profiles of human post-mortem brains from patients diagnosed with BP or MDD. The work has focused on three brain regions: the anterior cingulated cortex (AnCg), the dorsolateral prefrontal cortex (DLPFC), and the cerebellum (CB).


The present invention demonstrates, for the first time, differential expression of the 72 nucleic acids listed in Table 2, the 16 nucleic acids listed in Table 3, or the 967 nucleic acids listed in Table 4, in the brains of patients suffering from mood disorders, such as bipolar disorder and major depression disorder, in comparison with normal control subjects. In addition, the present invention identifies biochemical pathways involved in mood disorders, where the proteins encoded by the nucleic acids listed in Table 2, 3, or 4 are components of the biochemical pathways (e.g., the bFGF signal transduction pathway, the GPCR and cAMP/PI/Rho pathways, the proteasome pathway, the oxidative phosphorylation pathway, Myelination, Cytochrome P450, or the GABA and glutamate pathways; see also FIGS. 1-5, 10-13, and 15).


Finally, genes that are differentially expressed in MDD or BP and by gender are useful in diagnosing mood disorders, as the prevalence of certain mood disorders shows a gender bias. Differential expression by brain region similarly is a useful diagnostic and therapeutic tool, as certain mood disorders primarily affect certain brain regions.


This invention thus provides methods for determining whether a subject has or is predisposed for a mental disorder such as bipolar disorder or major depression 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 such as bipolar disorder and major depression disorder.


In some embodiments, 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 Table 2, 3 or 4; 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 of a gene listed in Table 2, 3, or 4. In some embodiment, the polypeptide comprises an amino acid sequence of a gene listed in Table 2, 3, or 4. 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 amniotic fluid. In some embodiments, the mental disorder is a mood disorder. In some embodiments, the mood disorder is selected from the group consisting of bipolar disorder and major depression disorder.


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 Table 2, 3, or 4; and (ii) determining the functional effect of the compound upon the polypeptide, thereby identifying a compound for treatment of a mental disorder.


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 Table 2, 3, or 4. 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 mental disorder is a mood disorder or psychotic disorder. In some embodiments, the mood disorder is selected from the group consisting of bipolar disorder I and II and major depression. In some embodiments, the psychotic disorder is schizophrenia. In some embodiments, the methods further comprise administering the compound to an animal, e.g., an animal subjected to stress as a model for depression 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 Table 2, 3, or 4; 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 Table 2, 3, or 4. 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 psychotic disorder. In some embodiments, the mood disorder is selected from the group consisting of bipolar disorder I and II and major depression. In some embodiments, the psychotic disorder is schizophrenia.


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 psychotic disorder. In some embodiments, the mood disorder is selected from the group consisting of bipolar disorder I and II and major depression. In some embodiments, the psychotic disorder is schizophrenia. In some embodiments, the compound is a small organic molecule, an antibody, an antisense molecule, aptamer, or a peptide.


The invention also provides methods of treating mental disorders 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 Table 2, 3, or 4. In some embodiments, the polypeptide comprises an amino acid sequence encoded by a gene listed in Table 2, 3, or 4. In some embodiments, the mental disorder is a mood disorder or psychotic disorder. In some embodiments, the psychotic disorder is schizophrenia. In some embodiments, the mood disorder is a bipolar disorder or major depression.


The invention also provides methods of treating mental disorders 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 Table 2, 3, or 4. In some embodiments, the mental disorder is a mood disorder or 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 differentially expressed in mood disorder subjects.


Table 2: Table 2 lists 72 genes differentially expressed in mood disorder subjects.


Table 3: Table 3 lists 16 genes differentially expressed in specific brain regions and mood disorder.


Table 4: Table 4 lists 967 genes differentially expressed in mood disorder subjects as determined by microarray analysis. Flag 1 indicates that the differential expression of the gene was confirmed by Real time PCR. Flag 2 indicates that differential expression of the gene was confirmed by anti-depressant studies. Flag 3 indicates that the gene belongs to an enriched gene ontology. Up and down indicates the direction of the changes compared to controls.


Table 5: Table 5 lists Real time PCR results on sample genes that are differentially expressed in mood disorder subjects.


Table 6: Table 6 lists anti-depressant treatment results for genes that are differentially expressed in mood disorder subjects.


Table 7: Tables 7A-D lists the gene ontology of selected genes differentially expressed in mood disorder subjects.


Table 8: Table 8 lists sample of genes that are differentially expressed in mood disorder subjects and are potential druggable targets.






FIG. 1 shows selected biochemical pathways for genes differentially expressed in mood disorder subjects.



FIG. 2 summarizes functions for signal transduction transcripts differentially expressed in MDD subjects.



FIG. 3 shows bFGF pathway transcripts differentially expressed in MDD subjects.



FIG. 4 shows values for differential expression of bFGF transcripts in MDD subjects.



FIG. 5 shows selected biochemical pathways that are dysregulated in mood disorders.



FIG. 6 shows selected biochemical pathways that are dysregulated in BP subjects.



FIG. 7 shows three genes overexpressed in mood disorder subjects that are located in the same chromosomal region.



FIG. 8 shows three genes overexpressed in mood disorder subjects that are located on 15q11-13 in the Prader-Willi region.



FIG. 9 shows certain genes regulated in human postmortem tissue and by antidepressants in rats.



FIG. 10 shows selected biochemical pathways (i.e., the GPCR and cAMP/PI/Rho pathways) for genes differentially expressed in mood disorder subjects. Two G protein coupled receptors, GPR37 and GPRC5B, are increased in both AnCg and DLPFC of BP patients, and decreased in MD. As downstream signaling pathways of GPCR, genes involved in cAMP pathway signaling are increased n BP patients, and decreased in MD. Genes involved in phosphatidylinositol pathways are deregulated specifically in MD.



FIG. 11 shows a selected biochemical pathway (i.e., the proteasome pathway) for genes differentially expressed in mood disorder subjects. The proteasome is an assembly of 28 alpha and beta subunits that functions to degrade proteins. The proteasome is involved in regulation of protein turnover and in particular oxidized proteins. There is an over representation of proteasome genes found in cortical regions of BP, but not in the cerebellum, suggesting that some functional compensation in the proteasome is occurring in BP patients.



FIG. 12 shows a selected biochemical pathway (i.e., the oxidative phosphorylation pathway) for genes differentially expressed in mood disorder subjects. The oxidative phosphorylation classification is involved in bioenergetics, metabolism, and as a byproduct can produce reactive oxygen species. This pathway is overly expressed in both bipolar and major depression, with differences between cortical regions and cerebellum.



FIG. 13 shows an example of a growth factor system (e.g., FGF) that is altered in mood disorders.



FIG. 14 shows RealTime PCR results which confirm that selected FGF-related genes first identified using microarray analysis are differentially expressed in mood disorders.



FIG. 15 shows selected genes in biochemical pathways involving GABA and glutamate that are differentially expressed in mood disorder subjects.





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., 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., Diagnostic and Statistical Manual of Mental Disorders, Fourth Edition, (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.


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, 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.


“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 Table 2, 3, or 4 or a polypeptide encoded by a gene of Table 2, 3, or 4), 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, calorimetric 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, 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.


“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 cl 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 1-8 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 mood disorders. The large spectrum of symptoms associated with mental disorders is likely 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 (AnCng, DLPFC, and CB) are implicated in the clinical symptoms of mental disorders such as mood disorders. Brain imaging studies focusing on particular brain regions, cytoarchitectural changes in brain regions, expression of key neurotransmitters 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 expression) of the genes of Tables 1-8 at the mRNA level in selected brain regions of patients diagnosed with mood disorders (e.g., bipolar disorder and major depression disorder) in comparison with normal individuals. This invention thus provides methods for diagnosis of mental disorders such as mood disorders (e.g., bipolar disorder, major depression, and the like), psychotic disorders (e.g., schizophrenia, and the like), and other mental disorders by detecting the level of a transcript or translation product of the genes listed in Tables 1-8 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.


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.


The genes and the polypeptides that they encode, which are associated with mood disorders such as bipolar disease and major depression, are useful for facilitating the design and development of various molecular diagnostic tools such as GeneChip™ containing probe sets specific for all or selected mental disorders, including but not limited to 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.


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 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 mood disorder.


For example, antidepressants belong to different classes, e.g., desipramine, bupropion, and fluoxetine are in general equally effect for the treatment of clinical depression, but act by different mechanisms. The similar effectiveness of the drugs for treatment of mood disorders suggests that they act through a yet as unidentified common pathway. We disclose herein that different classes of antidepressants (specific serotonin reuptake inhibitors, like fluoxetine and tricyclic antidepressants, like desipramine) regulate a common gene, and/or a common group of genes as well as a unique set of genes when the human and animal results herein are compared.


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 1-8, 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 listed in Tables 1-8, 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 Table 1-8.


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 1-8, 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 centrifuged 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 mental disorders including mood disorders or psychotic disorders 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 the cerebellum or the hippocampus, 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 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 protein, such as one comprising an amino acid sequence encoded by a gene listed in Table 1-8 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 1-8) 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 Table 1-8) 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 1-8) 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 calorimetric 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. Such brain regions are part of brain circuits or pathways that are implicated in mood disorders. 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.


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 depression (both chronic and acute), in which the rats are subjected to stress, 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 disorders and are useful in screening for modulators of mental disorders.


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 (11993)), 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. Nos. 5,506,337; benzodiazepines, 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-IV Made 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, dysthymic or cyclothymic disorders, substance-induced mood disorders, major depression, psychotic disorders, including paranoid psychosis, catatonic psychosis, delusional psychosis, 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 useful for treating mood disorders, e.g., schizophrenia, 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. Nos. 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, can be administered or 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 and psychotic disorders.


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) sipra; 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. U.S.A. 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). 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. Biochemistiy 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 Disorders 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) 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., SNPs or haplotypes associated with this genes.


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, saliva, CSF, urine or tissue samples.


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 Mood Disorders

A total of twenty mood disorder brains (9 bipolar and 11 major depression disorder patients) with twenty control brains were used in this study. Each brain pair (case and control) was matched on the basis of gender, age, and postmortem interval. Three brain regions, dorsolateral prefrontal cortex (DLPFC), anterior cingulate cortex (AnCg) and the cerebellum (CB) were extracted for RNA and subjected to microarray analysis using Affymetrix oligonucleotide GeneChips™. Each RNA sample was subjected to two independent analyses. The results were analyzed using multiple statistical tools and algorithms with various stringencies. Real time PCR analysis was used to confirm differential gene expression for selected genes. The genes identified using this study are listed in Tables 1, 2, and 3. Furthermore, biochemical pathways associated with the differentially expressed genes were identified (see FIGS. 1-5).


The two cortical regions DLPFC and AnCg had similar gene expression profiles in controls but differed significantly in MDD and BP, demonstrating distinct gene expression profiles. BP subject showed more changes in AnCg compared to DLPFC whereas MDD show less profound changes in both cortical regions but had greater effects in the DLPFC than in the AnCg. For BP, several candidate genes were located in chromosomal region 15q11-13, which is associated with the Prader-Willi syndrome (see FIGS. 6-8).


Example 2
Identification of Additional Genes Dysregulated in Mood Disorders

The RNA from three brain regions, dorsolateral prefrontal cortex (DLPFC), anterior cingulate cortex (AnCg) and the cerebellum (CB) from deceased patients diagnosed with bipolar disease or major depression, and matched controls were extracted and subjected to microarray analysis using Affymetrix oligonucleotide GeneChips™. 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. The effect of agonal factors and pH were taken into account for quality control of the RNA. Two RNA samples were subjected to independent analyses. The results were analyzed using multiple statistical tools and algorithms with various stringencies. The 967 genes identified using this study are listed in Table 4. Real time PCR analysis was used to confirm differential gene expression for selected genes. Real time PCR confirmation of differential gene expression for selected genes is listed in Table 5.


Furthermore, biochemical pathways associated with the differentially expressed genes were identified. In particular, cortical areas in BP patients showed activation of several pathways, including the proteasome pathway, the oxidative phosphorylation pathway, the ATP synthesis pathway, and chaperones (i.e., heat shock proteins). In addition, signaling pathways dysregulated in BP include, e.g., G-coupled protein receptors, the phosphatidylinositol pathway, the cAMP pathway, the mitogen activated protein kinase pathway, cytoskeletal systems, and the cortical GABA and glutamate systems. In MD, dysregulated genes includes genes involved in transmission of nerve impulses, neurogenesis, and the fibroblast growth factor system (FGF). (see FIGS. 10-12). Gene ontology (i.e., genetic signatures) for BP and MD can conveniently be used in developing diagnostic and therapeutic regiments for mood disorders.


Example 3
Identification of Additional Genes Dysregulated in Mood Disorders Using Rat Models of Depression and Anti-Depressant Treatment

Rats were exposed to chronic unpredictable stress treatments in parallel with chronic anti-depressants treatment (e.g., the tricyclic antidepressant desipramine and the specific serotonin reuptake inhibitor fluoxetine). Saline treated stressed rats (SS) and saline treated non-stressed rats (SN) were used as controls. In particular, saline treated stressed rats (SS) were compared to desipramine treated stressed rats (DS); saline treated stressed rats (SS) were compared to fluoxetine treated stressed rats (FS); saline treated non-stressed rats (SN) were compared to desipramine treated non-stressed rats (DN); saline treated non-stressed rats (SN) were compared to fluoxetine treated non-stressed rats (FN); and saline treated stressed rats (SS) were compared to saline treated non-stressed rats (SN). Gene expression changes in rat cortex following treatment were measured. The genes identified in this study are shown in Table 6. This data suggests that different classes of antidepressants, i.e., antidepressants with apparently different mechanisms of action may act through a common biochemical pathway.


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 1









GenBank
DLPFC-MDD

Chromosome



Accession#
Gene Description
OMIM
Location







NM1964
Early growth response protein 1 (EGR1)
EGR1
5q31.1



NM599
human Insulin-like growth factor binding protein
IGFBP5
2q33-34




5 (IGFBP5)



M87771
Fibroblast growth factor receptor k-sam, Splice
k-sam-III
10q26




3 (k-sam-III)



Z24725

H sapiens Mitogen-inducible gene (mig-2)

mig-2
14q22.1



M64347
human Novel growth factor receptor (FGFR3)
FGFR3
4p16.3



M80634
human Keratinocyte growth factor receptor
FGFR2
10q26




(FGFR2) (SEQ ID NO: 1)



Z14228
Nuclear mitotic apparatus protein 1, Alt. Splice
NUMA U4
11q13




Form 2 (NuMA Clone U4)



X67951
human Proliferation-associated gene (PAGA)
PAGA
1p34.1













GenBank
DLPFC-MDD



Accession #
Gene Description





AF036268
SH3-domain GRB2-like 2
OMIM - SH3 DOMAIN, GRB2-LIKE, 2; SH3GL2


AF060877
regulator of G-protein signalling 20
OMIM - REGULATOR OF G PROTEIN SIGNALING 20; RGS20


AL049538
ras association (RalGDS/AF-6) domain containing
OMIM - RAL GUANINE NUCLEOTIDE DISSOCIATION STIMULATOR;



protein JC265
RALGDS


D14838
fibroblast growth factor 9 (glia-activating factor)
OMIM - FIBROBLAST GROWTH FACTOR 9; FGF9


D26070
inositol 1,4,5-triphosphate receptor, type 1
OMIM - INOSITOL 1,4,5-TRIPHOSPHATE RECEPTOR, TYPE 1; ITPR1


J02902
protein phosphatase 2 (formerly 2A), regulatory
OMIM - PROTEIN PHOSPHATASE 2, STRUCTURAL/REGULATORY



subunit A (PR 65), alpha isoform
SUBUNIT A, ALPHA; PPP2R1A


J04513
fibroblast growth factor 2 (basic)
OMIM - FIBROBLAST GROWTH FACTOR 2; FGF2


L05624
mitogen-activated protein kinase kinase 1
OMIM - MITOGEN-ACTIVATED PROTEIN KINASE KINASE 1; MAP2K1


M64788
RAP1, GTPase activating protein 1
OMIM - RAP1, GTPase-ACTIVATING PROTEIN 1; RAP1GA1


M87771
fibroblast growth factor receptor 2 (bacteria-
OMIM - FIBROBLAST GROWTH FACTOR RECEPTOR 2; FGFR2



expressed kinase, keratinocyte growth factor



receptor, craniofacial dysostosis 1,



Crouzon syndrome, Pfeiffer syndrome, Jackson-



Weiss syndrome)


M96995
growth factor receptor-bound protein 2
OMIM - GROWTH FACTOR RECEPTOR-BOUND PROTEIN 2; GRB2


U09759
mitogen-activated protein kinase 9
OMIM - MITOGEN-ACTIVATED PROTEIN KINASE 9; MAPK9


U24152
p21/Cdc42/Rac1-activated kinase 1 (STE
OMIM - p21/CDC42/RAC1-ACTIVATED KINASE 1; PAK1



20 homolog, yeast)


U49857
transcriptional activator of the c-fos promoter


W28432
Cluster Incl. W28432: 47f2 Homo sapiens
OMIM - NEUROTROPHIC TYROSINE KINASE, RECEPTOR, TYPE 2; NTRK2



cDNA /gb = W28432 /gi = 1308443/ug = Hs.92030



/len = 921


X07109
protein kinase C, beta 1
OMIM - PROTEIN KINASE C, BETA-1; PRKCB1


X54938
inositol 1,4,5-trisphosphate 3-kinase A
OMIM - INOSITOL 1,4,5-TRISPHOSPHATE 3-KINASE A; ITPKA


Z71929
fibroblast growth factor receptor 2 (bacteria-
OMIM - FIBROBLAST GROWTH FACTOR RECEPTOR 2; FGFR2



expressed kinase, keratinocyte growth factor



receptor, craniofacial dysostosis 1,



Crouzon syndrome, Pfeiffer syndrome, Jackson-



Weiss syndrome)














GenBank
Antcg BP




Accession #
Description
Symbol







NM_004794
RAB33A, member RAS oncogene family
RAB33A



NM_002844
protein tyrosine phosphatase, receptor type, K
PTPRK



M14752
M14752 HUMABLA Human c-abl gene |Gen
ABL1




Bank==M14752



NM_005252
v-fos FBJ murine osteosarcoma viral oncogene
FOS




homolog



NM_002229
jun B proto-oncogene
JUNB



NM_014813
KIAA0806 gene product
KIAA0806



AB007943
AB007943: Homo sapiens mRNA for KIAA0474
RAP1GA1




protein |GenBank==AB007943



NM_004067
chimerin (chimaerin) 2
CHN2



NM_003676
degenerative spermatocyte homolog, lipid
DEGS




desaturase (Drosophila)



NM_000830
glutamate receptor, ionotropic, kainate 1
GRIK1



NM_002487
necdin homolog (mouse)
NDN



NM_002921
retinal G protein coupled receptor
RGR



NM_001390
dystrobrevin, alpha
DTNA



NM_006000
tubulin, alpha 1 (testis specific)
TUBA1



NM_001634
S-adenosylmethionine decarboxylase 1
AMD1



NM_006931
solute carrier family 2 (facilitated glucose transporter),
SLC2A3




member 3



NM_003832
phosphoserine phosphatase-like
PSPHL



NM_005010
neuronal cell adhesion molecule
NRCAM



NM_002073
guanine nucleotide binding protein (G protein),
GNAZ




alpha z polypeptide



L24123
L24123: Homo sapiens NRF1 protein (NRF1)
NFE2L1




mRNA/cds = UNKNOWN /gb = L24123




/gi = 438646 /ug = Hs.83469 /len = 4992|Gen




Bank==L24123



NM_000810
gamma-aminobutyric acid (GABA) A receptor,
GABRA5




alpha 5



NM_005398
protein phosphatase 1, regulatory (inhibitor)
PPP1R3C




subunit 3C



AI526089
AI526089: DU3.2-7.H07.r Homo sapiens cDNA|
COX5B




GenBank==AI526089



NM_000840
glutamate receptor, metabotropic 3
GRM3



NM_012249
ras-like protein TC10
TC10



NM_004791
integrin, beta-like 1 (with EGF-like repeat
ITGBL1




domains)



NM_000615
neural cell adhesion molecule 1
NCAM1



NM_003916
adaptor-related protein complex 1, sigma
AP1S2




2 subunit



NM_001406
ephrin-B3
EFNB3



NM_001718
bone morphogenetic protein 6
BMP6



X66358
X66358 cds#2 HSSTHPKB H. sapiens mRNA
CDKL1




KKIALRE for serine/threonine protein




kinase|GenBank==X66358




DLPC-BP



D00654
actin, gamma 2, smooth muscle, enteric
ACTG2



U19599
U19599 HSU19599 Human (BAX delta) mRNA|
BAX




GenBank==U19599



NM_006908
ras-related C3 botulinum toxin substrate 1
RAC1




(rho family, small GTP binding protein Rac1)



NM_002374
microtubule-associated protein 2
MAP2



AJ001612
phosphoserine phosphatase-like
PSPHL



NM_000293
phosphorylase kinase, beta
PHKB



NM_020217
hypothetical protein DKFZp547I014
DKFZp547I014



NM_004379
cAMP responsive element binding protein 1
CREB1



NM_032041
neurocalcin delta
NCALD



NM_015716
Misshapen/NIK-related kinase
MINK



AF059274

Homo sapiens cDNA FLJ37320 fis, clone

CSPG5




BRAMY2018106



NM_006158
neurofilament, light polypeptide 68 kDa
NEFL



NM_002730
protein kinase, cAMP-dependent, catalytic,
PRKACA




alpha



NM_003885
cyclin-dependent kinase 5, regulatory sub
CDK5R1




unit 1 (p35)



NM_003020
Secretory granule, neuroendocrine protein




1 (SGNE1)(7B2 protein) located at chromosome




band 15q13


















TABLE 2







NM1964
Early growth response protein 1 (EGR1)


NM599
human insulin-like growth factor binding protein 5 (IGFBP5)


M87771
Fibroblast growth factor receptor k-sam, Splice 3 (k-sam-III)


Z24725

H sapiens Mitogen-inducible gene (mig-2)



M64347
human Novel growth factor receptor (FGFR3)


M80634
human Keratinocyte growth factor receptor (FGFR2) (SEQ ID NO: 1)


Z14228
Nuclear mitotic apparatus protein 1, Alt. Splice Form 2 (NuMA Clone U4)


X67951
human Proliferation-associated gene (PAGA)


AF036268
SH3-domain GRB2-like 2


AF060877
regulator of G-protein signalling 20


AL049538
ras association (RaIGDS/AF-6) domain containing protein JC265


D14838
fibroblast growth factor 9 (glia-activating factor)


D26070
inositol 1,4,5-triphosphate receptor, type 1


J02902
protein phosphatase 2 (formerly 2A), regulatory subunit A (PR 65), alpha isoform


J04513
fibroblast growth factor 2 (basic)


L05624
mitogen-activated protein kinase kinase 1


M64788
RAP1, GTPase activating protein 1


M87771
fibroblast growth factor receptor 2 (bacteria-expressed kinase, keratinocyte growth



factor receptor, craniofacial dysostosis 1, Crouzon syndrome, Pfeiffer syndrome,



Jackson-Weiss syndrome)


M96995
growth factor receptor-bound protein 2


U09759
mitogen-activated protein kinase 9


U24152
p21/Cdc42/Rac1-activated kinase 1 (STE20 homolog, yeast)


U49857
transcriptional activator of the c-fos promoter


W28432
Cluster Incl. W28432: 47f2 Homo sapiens cDNA /gb = W28432 /gi = 1308443 /ug =



Hs.92030 /len = 921


X07109
protein kinase C, beta 1


X54938
inositol 1,4,5-trisphosphate 3-kinase A


Z71929
fibroblast growth factor receptor 2 (bacteria-expressed kinase, keratinocyte growth



factor receptor, craniofacial dysostosis 1, Crouzon syndrome,



Pfeiffer syndrome, Jackson-Weiss syndrome)


NM_004794
RAB33A, member RAS oncogene family


NM_002844
protein tyrosine phosphatase, receptor type, K


M14752
M14752 HUMABLA Human c-abl gene|GenBank==M14752


NM_005252
v-fos FBJ murine osteosarcoma viral oncogene homolog


NM_002229
jun B proto-oncogene


NM_014813
KIAA0806 gene product


AB007943
AB007943: Homo sapiens mRNA for KIAA0474 protein|GenBank==AB007943


NM_004067
chimerin (chimaerin) 2


NM_003676
degenerative spermatocyte homolog, lipid desaturase (Drosophila)


NM_000830
glutamate receptor, ionotropic, kainate 1


NM_002487
necdin homolog (mouse)


NM_002921
retinal G protein coupled receptor


NM_001390
dystrobrevin, alpha


NM_006000
tubulin, alpha 1 (testis specific)


NM_001634
S-adenosylmethionine decarboxylase 1


NM_006931
solute carrier family 2 (facilitated glucose transporter), member 3


NM_003832
phosphoserine phosphatase-like


NM_005010
neuronal cell adhesion molecule


NM_002073
guanine nucleotide binding protein (G protein), alpha z polypeptide


L24123
L24123: Homo sapiens NRF1 protein (NRF1) mRNA /cds = UNKNOWN /gb =



L24123 /gi = 438646 /ug = Hs.83469 /len = 4992|GenBank==L24123


NM_000810
gamma-aminobutyric acid (GABA) A receptor, alpha 5


NM_005398
protein phosphatase 1, regulatory (inhibitor) subunit 3C


AI526089
AI526089: DU3.2-7.H07.r Homo sapiens cDNA|GenBank==AI526089


NM_000840
glutamate receptor, metabotropic 3


NM_012249
ras-like protein TC10


NM_004791
integrin, beta-like 1 (with EGF-like repeat domains)


NM_000615
neural cell adhesion molecule 1


NM_003916
adaptor-related protein complex 1, sigma 2 subunit


NM_001406
ephrin-B3


NM_001718
bone morphogenetic protein 6


X66358
X66358 cds#2 HSSTHPKB H. sapiens mRNA KKIALRE for serine/threonine



protein kinase|GenBank==X66358


D00654
actin, gamma 2, smooth muscle, enteric


U19599
U19599 HSU19599 Human (BAX delta) mRNA|GenBank==U19599


NM_006908
ras-related C3 botulinum toxin substrate 1 (rho family, small GTP binding protein Rac1)


NM_002374
microtubule-associated protein 2


AJ001612
phosphoserine phosphatase-like


NM_000293
phosphorylase kinase, beta


NM_020217
hypothetical protein DKFZp547I014


NM_004379
cAMP responsive element binding protein 1


NM_032041
neurocalcin delta


NM_015716
Misshapen/NIK-related kinase


AF059274

Homo sapiens cDNA FLJ37320 fis, clone BRAMY2018106



NM_006158
neurofilament, light polypeptide 68 kDa


NM_002730
protein kinase, cAMP-dependent, catalytic, alpha


NM_003885
cyclin-dependent kinase 5, regulatory subunit 1 (p35)


















TABLE 3







Acc.


Disorder/Region
Description
Numb.







MD DLPFC
carboxypeptidase D
U65090



prostaglandin D2 synthase (21 kD, brain)
AI207842



NEL-like 1 (chicken)
D83017



zinc finger protein 36, C3H type-like 1
X79067



phosphoribosyl pyrophosphate synthetase 1
X15331


MD AnCng
solute carrier family 1 (glial high affinity glutamate transporter), member 3
D26443



clathrin, light polypeptide (Lcb)
M20470



aldolase A, fructose-bisphosphate
X05236



ubiquitin carboxyl-terminal esterase L1 (ubiquitin thiolesterase)
X04741


BP AnCng
v-raf-1 murine leukemia viral oncogene homolog 1
X03484



cytochrome c oxidase subunit Vb
AI526089



proteasome (prosome, macropain) 26S subunit, non-ATPase, 1
D44466



tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation
X56468



protein, theta polypeptide



nuclear receptor subfamily 4, group A, member 1
L13740



chondroitin sulfate proteoglycan 3 (neurocan)
AF02654



fatty acid binding protein 7, brain
AJ00296


BP DLPFC
carboxypeptidase D
U65090






indicates data missing or illegible when filed














TABLE 4







Summary














Genbank









Accession No.
AnCg BP
AnCg MD
DLPFC BP
DLPFC MD
CB BP
CB MD
flags





D50310

up


up

1


L08485


up



1, 2, 3


U28964




up

1


AF016917



up

up
1, 3


L19182



down


1


AJ001612

up
down
up
down
up
1, 3


U66879



up


1


J04046

up




1


X63575

up

up
up

1, 3


S74445



up


1, 2, 3


X71490
up

up
up
up

1


AF112471

up

up


1, 2, 3


AB006626

up




1


U37143

down

down


1, 3


AC004131
up
down
up
down

down
1


M29273

down
up
down


1, 2


X76220
up

up



1, 2


M12267


up



3


AF060877

down

down

down
1


AB018305



down


1, 2


U58334

down

down

down
1


AB020629
up
down
up
down


1, 3


U37122

down

down


1, 3


AL080061

down

down
down
down
1, 3


M34309

down

down


1


M80634

down

down


1


(SEO ID NO: 1)


M64347

down

down


1


X57206

down

down

down
1, 3


X77196

down

down


1, 3


Z24725

down

down

down
1


AB018342

down

down


1, 3


Y10275


down


up
1, 3


AB007943
down





1


AL049538

down

down

down
1


M14758


down
down

down
3


X13839


down
down


3


X63432


up

up

3


X04098
up

up

up

3


AF006082
up




down
3


D67031

down

down

down
3


L22214


up



3


J03473


up



3


AJ236876
up
up

up


AF072902

up

up


U84011



down

down


K02215
up



up
up


AI800578

down

down

down


AL049954



down


R59606

down

down


M80899

down

down


2, 3


U00957



up
up


AA114830


up


U81607


up



3


M90360
down

down


X15414
up

up



3


U05861


up


D17793

down

down


3


K03000



down

down
2


U46689


up


down
3


U24267
down

down
down


M93405

down

down


3


U34252
up

up



3


X05236
up
up
up

up
up


M21154
up

up

up

3


W63793


up



3


M63175


up

up


AB028994

down

down


U29926



down


X81438
up
up
up
up


3


D14662

down

down

down
3


AF091077
down





3


X97074
up
up
up
up


2


D38293
up
up
up



3


J02611



down

down


M12529

down

down


3


D86981


up


down
3


U41518



down
down


U34846

down

down
up

2, 3


D87468

down




2


L04510


up



2, 3


AF049884

up




3


U02570



down

down


AB002292



down


U50523

up




3


AI525393
up





3


AF006087
up


up


2, 3


AF006088


up



3


Z11501



up


L08424

down

down


3


M27396

up
up



3


S67156

down

down


AL096842

down

down


AB018258



down


J05096

down

down

down
2


M37457




up
up
2


W28508



down
up


AF007876

down

down


J04027




up

2, 3


L20977



up
up

3


W28589
up


up


3


AJ010953
up





2


D14710
up

up
up
up

3


U09813

up
up



3


AF087135

up
up



3


AA845575

up
up



3


AF047436

up
up



3


AA917672


up



3


X83218

up
up



3


D16469
up
up


AL049929

up
up



3


D89052
up

up



3


AI318615


down



3


AI547262


up



3


AA056747


up



3


L09235

up

up
up

3


AA877795

up
up



2


X76228
up
up
up
up


3


W26326



up
up
up


X79888


up



2


X66030



down


M76125



down


S82297

down

down


AB021288

down

down


V00567

down

down

down


AF029893
up
up
up
up


3


AF082868

down

down


3


U50708



down


U00115



down

down


AL049257


up
down

down


AB004066

down

down

down


AF001383
up

up



2, 3


U68485
up





2, 3


AF002697


up

up

3


S78771
up


up
up
up
3


AC005306


down


up


AB023169


up

up


U72649

down

down

down
2


AF047472
up




down


AB023171
up
down

down


X94910
up
up
up


up
3


AF054175

up




3


AF014837


down
down


AF009425



down
down
down


X95592


up



2, 3


AB007948
up
up

up


D86062
up





3


AL080097

down

down

down


AF006621

up
up
up


3


J03037

down
up
down

down
3


U79666


down



3


M76559



up


2


S60415

up




3


AF068862
up

up
up
up

2


U12022

up

up
up


D45887
up



up


AB020640

up


U02390



up
up

2


U02390



up
up


U02390



up


X85030

down

down


U20325


up



2


AB002376

up



down


AL035079
up

up


AF070648

down

down


L10822
up


up


AF091433



down


AF026166
up

up



3


X74801
up

up



3


AF026292
up





3


X69398
up



up


M38690


up
down

down


AF023158

down


M37712



down


AL031282
up




down


M35543

up
up


down


W27541

down

down


U59325

up

up


AF006484

down

down


3


X66364



up


2


L04658
up
up

up


X77743
up

up



3


X66358



up


U22398

down

down

down


M16965



up
up
up


W27184



up
up
up


U65887
down





3


U60808
down





3


AI056696


up



3


U78516
down


AL080084
up

up



3


AA189161
up

up


AB023203
down




down
3


U03749

up




2


Y00064

up


U07223
up

up



2


X70297


up


W29042


up



3


D49738


up



3


Z30644


down



3


U89916

down

down


M59287

down

down

down


M59287

down

down

down


AF039704


up


X91788
up





3


M20469
up
up

up


3


M20470
up
up

up
up

3


AB020709


up
up


2, 3


S80562



down


2, 3


D13146
up

up


M19650
up





3


Z21488



up
up


AB020675

up


AB014533
up



down
down


M92642



down

down


M58526

down

down


U65928
up
up
up



3


AA149486
up
up




3


M22760

up




3


M19961

up




3


AI526089
up
up
up



3


T57872
up
up




3


AA152406
down





3


AA978033

up




3


AB007618

up




3


N50520

up
up



3


U65090


up


down


X51405


up


S74445



up


2, 3


M27691
down





2


S68271
up

up



3


D10656
up



up

3


U49857

down

down


U49857

down

down

down


AL038340
up

up


AL038340
up

up


AF039397
up

up


AF053641
up





3


M27826


down


U89896


down

up
up
3


D32039



down
up


X15998



down


X15998



down


AF026547

down

down

down


M33146



down


2


L22569


up

up


X16832

down

down


3


Y07593
up

up

up


L06797

down



down
3


L47738

up

up
up


M33318
down





3


Y11307



down
down
down


M98529
up
up

up
up
up


AB002379

down

down

down


D15057


up



3


AL050152


up


down


D31767


up



3


AL050084



down

down


AB002367


up

down
down
3


AF086947

up

up


U50733
up
up
up



3


W26651
up
up
up


U48705



down


L20817

down

down


U59321



down
up


AF000982


up


AF007142



down

down


U63825
up
up




3


AF021819


up



3


AL080115


up


down


AL049944
up

up


AL049934

down

down
down
down


AL050390

up
up


AL050272
up
up


AL050159

down

down


L08069
up
up
up
up


L08069
up
up
up


AI810807

up
up



2, 3


AI540958


up



3


AF000430
up


up
up

3


D50857

down

down

down
3


AF007875


up



3


U97105


up


M97388
up

up



3


D83407

up
up


S65738


up



3


U26742


down



3


U46744

down


up

3


U84551

down


up


U46746

down




3


X68277

down

down

down
3


L05147



up


AB013382

down

down


3


U31930


up



3


U46461


down



3


D86550


up



2, 3


M91670
up


up


M91670
up


up


AL050282
up
up


M31210

down


D13168



down


X70940

up
up



3


AB023159
up
up
up
up
up
up


U03877

down

down


U66406
up
up




3


AB011542


up


L18960
down


AF035280


up



3


U36764
up





3


U39067
up

up
up


3


U94855

up
up



3


U54558
up





3


AC002544


down

down


D13748
up



up


L36055


down



3


U49436

up


up

2,3


AL080199

down

down


C18655



down


AB002303

down

down


X51956

up




2


L35594

down
up
down

down
2, 3


L35594

down
up
down

down
2, 3


D45421
down





2


AF103905



down


U81984


down



3


D83492

up

up


U12535



down


M34309

down

down
down

3


X81625
up

up


down
3


J04058


up



3


AB028990


up


J02931



down


AJ002962
up

up



3


AA977580


up



2


W26480

down

down

down


AF035284



down

down


X87241

down

down


AF000561


down


M30448
up

up
up


AB014596


up



3


D14697
up

up



3


U60060


up



3


U60061


up



3


X59065



down
down
down


Z70276

up

up
up

3


U66198



up


3


D14838



up


2


Z69641

down

down

down


Z69641

down


M87770

down

down

down


Z71929

down

down

down


X55741
up

up
up


AF070557


up


W27472

down

down


W26655
up

up


AF052106

up

up


AL049949

down

down
down
down
3


X02761
down


down


V01512

down

down

down


V01512

down

down
down
down


K00650

down



down


AF032885

down

down


M84562
down


U32519


up



3


AB014560


up



3


AI565760


up



3


AJ225028


up



3


M82919



up
up

3


X15376



up
up

3


D86181
up




down


S68805



down


D00723


up


down
3


Y13286
up

up



3


S40719

down

down

down
3


D87467


up


M65188

down




3


X52947

down

down

down
3


M57609


down
down


3


U33267
up
up
up



3


X76648

up




3


AB020645

up
up



2, 3


U08997
up
down


U08997

down


X59834

down

down

down
3


U43083
up


up


3


D90150
up
up


AF017656


up
up
up


AJ238764


up



3


AL049367



down


AB020662
down
down

down

down


AF047438
down





3


M22632

up
up



3


AF016004

down
up
down

down
3


D38449



up


3


U87460
up
down
up
down

down


AJ011001



down


X71973

up




2, 3


W28944
up

up



3


M81886
up



up

3


U10301



up


3


X82068
up





3


L19058
down





3


S40369



up

up


X77748
up

up



2, 3


D87119

down

down
down
down
3


D87119

down

down
down
down
3


X04412
up
down
up
down

down
3


M16594
down



down


J05459



up


2, 3


U90313
up
up
up



3


M95809


up



3


X03473



up


D64142



up


D64142



up


L19779



up


M37583

up
up



3


H15872
up

up


AA255502
down



up
up


M25079
down

down
down


L48215
down


down


AF019214



down
down
down


AF029890
up
up
up


U31814


up



3


AL034374

down
up
down

down


AI391567


down



3


U51004

up




3


AB014555

down




3


X58536

down

down


M32578



down


U23803


up



3


X12671


up


M16342


up



3


D89678


up



3


U01923


up



3


W27191


down



3


X92814


up



2


X99209
up


AF068754


up



3


M11717

down


down
down


L26336
up
up
up
up
up


L26336
up
up
up
up


L26336
up
down
up
down

down


L26336
up


down


L12723
up





2


X87949


up



3


X13794
up
up
up


Y00371
up
up
up
up


L15189
up

up



2, 3


AL021937


up


X15183


up



3


J04988
up
up

up


W28616


up


M22382


up



3


AI912041


up



2, 3


X57830

up




2, 3


AI434146


down



3


AF012023


up


X77956



down

down


AL022726



down


U49283
up
up
up


AA522698
up
up
up



3


X17025


up



3


U66042

up

up


M24594
up





3


X16302

up

up


AB017563



down


2


L42572


up



3


U26398



up
up


X77567



up
up
up
3


U96876


up



2


X53586



down

down


X07979



down


2


AL021786



down


AA477898
up



up


X54938
up
up

up
up

3


U23850

up


up

3


AB016492
up
up


J04111
up
up


X51345

down

down

down


AF070523


up


D79994

down

down

down


L02840
down





3


U52155

down




3


U39196



up


Y15065

up




3


D26067


up



3


D31887


up
up


3


D14663


up



3


AL049250
down
down
down


D87074

up



down


D87443
up




down
3


D87445



down


AB002347


up


AB002361
up


AB007903


up



3


AB007963



down

down


AB011095



down
down


AB014526

down

down


AB014544

down

down
down
down


AB018335

down

down


AB020637


up


AB020661



down

down


AB023152
up


AB023209

down

down


AB023230

down

down


3


AB028972
up
up


up


AB028977
down





3


AB029034



down

down


AF070621


up



3


Y08319
up





3


AB002357


up



3


AF035621
up
up

up


3


U59919


up



3


AJ001685

down

down


J04182
up

up


down
3


U36336

down
up
down

down
3


Y11395
up

up


AL050126





down
2


M90424


down



3


X02152
up
up




3


X13794


up

up


AI535946
up
up
up

up


D55696
up





3


AF087693




up

2


X76488
up

up


down
3


U41060


up



3


X61118



down


U79297
up

up


AL039458

down

down


2


AB011540



down


M63959
up
up
up

up

3


M92439
up
up
up
up


AB012293
up
up

up


3


W26633
up

up


U03100
up

up


down
3


U03100
up




down
3


D55649
up

up



3


AA420624



down


3


U01828



up
up


U89330



up
up


S76756
down





3


L05624
up
up

up


U17743
up


up
up

3


U71087



up
up

3


Z11695
up


up
up

3


X14474




up

2, 3


X66867


down


AF072250
up




down


D84557
up




down


X79440

up




3


S57212



up


AW006742
down
down
down


AI674208

down

down
up


AI674208

down

down
up


AF038186


up


W26659


up


AB014579


up

up


M16279



down


2


D25217



down


AI127424

up




3


AF001359


down


X70326

down
up
down


3


AF041080
down

down

down
down


AI670788

up
up


Z48051


up
down


U64565

down
up



3


D14812


up



2, 3


AI597616

up
up



3


Y11681
up
up
up
up
up

3


AL050361


up



3


Z98946



down


AI547258

down

down


2


AF072928
up

up


down
3


M55405
down

down


AF001548
down

down
down


2


AF013570


down
down


2, 3


AF001548
down

down
down


2


D10667


down



2, 3


D10667


down



2, 3


J02854
down

down



3


AF020267



down


3


AB029029

up



down


U42349
up
up
up



3


AF052142
up
up
up



3


AA126505
up





3


X77548





down
2


AF044209

up

up


U35139
up
up
up



2, 3


D87953



down

down


AF047185

up




3


AC002400
up
up
up


AI345944

up




3


AA203354


up



2, 3


AF047181
up

up
up


3


AA527880
up





3


AA760866


up



3


AF050640
up

up



3


AI541336

up




3


AC005329
up

up


AF053070
up
up
up
up


3


Y16241

down

down


3


D63878

down

down

down


D23662

up


X05608

up
up

up


D83017

up
up
up


3


W27762
down

down



3


X64318


up



3


Z83840
up
up

up


AB023192
up
up
up
up


3


AF019415

down

down


AF019415

down

down


X17620

up




2, 3


X73066
up
up
up



2, 3


AL038662

up




2, 3


X58965

up




2, 3


M86707
up
up

up


2


AI816034

up
up


U97669
down




down


W28770
up
up

up


3


AF002020
up




down
3


AF002020
up
down

down

down
3


AJ132583
up

up



3


U61849
up
up
up
up
up
up
2, 3


AI198311
up





2, 3


L13740
down
down
down
down
down
down


L13740
down
down
down
down
down
down


AB002341


up


down
2


U55258



up
up

2


U55258



up
up

2


X99076

up


AB011150
up
up
up
up


3


U03985

up
up



2, 3


X55740



down


AI018523

down

down

down


X75958

down

down
up
down
2, 3


W28432

down

down

down


Y10148

down

down


AL050066
up
up

up


U48250

down

down

down


U48250

down


M63623


up


down


AF061034

up

up


3


AF061034

up




3


U63717


up
up


3


U62961

up




3


AB017016
up
up

up


M80482
up




down


AB023211

down

down
down
down


L13385


up



2, 3


D63391
up



up

3


U24152
up
up

up
up

2, 3


U24152

up

up
up

2, 3


AF068864
up
up
up
up


AF068864
up
up
up
up


AF005043

up
up



3


M93650



down


3


AI521453
up

up

up

3


X73424
up

up



3


AB020631



down

down


D13892

up
up



3


D25547
up
up
up
up
up

3


U52969

up


up

3


AA535884



down


U40370
up





2


AB007946

up


S41458



down


AF056490

down

down

down
3


L42451
up





3


X98248

up

up
up


AB002345



down
down
down
2


AF093670


up


down
3


U41816
down





3


AL096719


up



3


V00572

up
up

up

2, 3


M83088
up





2, 3


X84908


up


U45976


up


down
3


AF010312
up
down
up
down


AL120815
up

up
down
down
down


Z29090
down





3


U81802
up



up

3


U49070

up



up


W28299
up
up

up


AL050371



up


D30037



up


U03090



down


U60644

up

up

up
3


U84573

down




3


M54927

down
up
down

down
3


M22299


up


down
3


X57398
up
up
up
up
up


D11428
up
down
up
down

down
3


AF001601

down

down


AL050161

down

down

down


AF017786

down

down
up

2


AF016371
up





3


AF001691

up


down

3


Y18207

down

down


Y18207

down


N36638

down

down

down
3


Z50749

up




3


J02902

up


J02902

up


M64929
up

up


M64929


up


M29551

up




2


X89416
up


up


U44772


up



3


AB014512
down





3


X67951


up



2


L19185
up
up
up


U25182
up

up


M33336


up

up

3


M33336


up

up

3


M33336


up


X07109

up

up


3


X06318

up

up


3


Z15108

up

up


3


U29185


up



2


AB011124

up


up
up


X15331
up


up
up

3


D87258

down

down

down


D87258

down

down

down


J03077
up

up



3


M85169
up

up



3


L76517
up

up


down
3


D00760


up



2, 3


D00762
up

up



3


D00761
up

up



3


D26598
up





2, 3


D26600
up

up



3


D26600
up

up



3


D29011
up

up



2


D29012
up





3


D38048
up
up
up



3


AF035309

up


D44466
up

up



2, 3


AL031177
up

up


AB009398
up





2, 3


D78151
up





3


U51007

up


D50063


up



3


D38047
up
up
up
up


3


D38047
up





3


AJ001612
down
up
down
up
down
up
3


D14694
up


M98539

down

down

down


AI207842

down



down
3


AI207842

down

down


3


U33284



up


M14630



down


M57399

down

down


2, 3


M57399
down
down

down


2, 3


X54131

down


L77886
up


Z48541
up


up


2


D64053
up


M93426

down

down

down
2, 3


X63578




up

2


Z48054
down





3


AL031781

down


AL031781

down

down

down


AI540957


up



3


AF052113
up

up



3


M28209
up

up



3


AL050268


up



3


AL050268


up



3


U59877
down
down

down


3


AI189226

down

down


D14889
up

up



3


D14889
up

up
up


3


M28212
up



up

3


AJ133534
up

up



3


X98001


up


D25274


up


U41654


up



2, 3


M35416

up


M31469
up
up
up
up
up


M31469
up
up

up
up


AF054183
up
up
up
up
up

3


X63465
up

up


X63465


up


S80343
up





3


D79990

down

down

down


U28686
down
down

down
down
down


U89505
up

up


U23946
up


M11433



down


3


X00129
up
up

up


3


N92548

down

down


AW044624
up





3


U27768



up


2, 3


U78166

up
up



3


D26129
up

up
down

down


AF037204


up


down
3


X13973
up

up


M63488
up


up
up


D87735

up




2, 3


X55954
up
up




3


AI708983

up
up



3


X57958


up



3


Y00281
up

up



2, 3


AL031659
up

up


AA977163
up
up
up



3


M13932

up




3


AI526078

up




3


D14530
up





3


S79522

up




3


X55715
up

up



3


M84711

up
up


up
2, 3


Y11651


up



3


L10333
up
up
up



2, 3


AB020693


up

up


M84820



up


AL049940
up

up


down


AJ001515



down


AB020658


up


X91257
up
up
up
up


3


M55580

down

down


AF051323
up
up

up


D12676
up



up

3


M25756


up

down
down
2


AF070614


up



3


L10338
down





3


AF049498
up
up

up

up
2, 3


AB011178

down

down

down


AB007937



down
down


AB015345
up
up

up
up


X97064


up



3


AJ131245


up



3


AF055006
up





3


AF054184

up
up



2, 3


U73167

down

down


AB000220


up


AB002438

down

down

down


Z11793



down


D86957

down

down

down
3


AI743134

down

down

down


Z81326


up



3


D28423
up



up


AL031681
up




down


L41887


up


down


Y00757

up
up



2


AF036268

up
up
up


3


AB007960

down

down


U33760
up
up
up

up


W26700

up

up
up


U08989



up


3


U01824

down

down


3


W28850

down




3


D26443

down

down

down
3


H10201



down


X60036


up



3


M20681
up
up




3


AF007216

down

down

down
2, 3


AF011390

down


up

2, 3


AF015926

down

down


3


D86959


up


down


U96094
up



up
up
3


D80000


up


down
2, 3


X59960
up

up



3


AF053136

up




2


AL049650
up
up

up


AA733050
up
up


U40571



down


3


AF034546


up



3


X02317


up



3


X63753


up


down
3


AJ001183



down


2


Z46629

down

down

down


AB011088
down


down


J03040



down

down


J04765
up

up


AF052124
up

up


down


AF039843

down

down

down


Y08685


up


down
3


D78130


up



3


M32313



up


2


M32886

down

down


3


U88666


up



3


J00306
up





2


AI636761
up





2, 3


AB011107

down

down

down


L78440

up

up


U04735
up

up
up
up


M86752



up
up


X99325
up
up

up
up

2, 3


AF099989


up


down
3


M31303


up



2


X85116

down

down
down
down


AL035306


up



3


U77942





down
2


D63506



down

down


U34804

up


U40215
up
up

up
up

3


AF039945

down




2


U93305

up

up


X68194

down
up
down

down
3


D38522


up


U18062
up

up



3


M95787
down

down


AF010400
up

up


AL050265


up



3


AL050107



down

down


D50495
up





2


M80627

down

down

down


D15050

down

down

down


U19969
down




down


X52882

up




3


U49188

up
up


L24804


up


X75861


up



3


W28869
up



up

3


L06139

down

down

down


X93512



up
up


S95936
up

up
down

down
3


M55153


down



3


L12350



down


L12350

down

down


AJ133115

down

down


M24748
up


up


X97544


up



3


X97544


up



3


L27476



down

down


AB028950



down


AI688299

down
up
down

down
3


R16035
up





3


U81006


up



3


M92383

up


D38305



down

down


D13641

up




3


U09477
up





3


M12125


down



3


M12125


down
down


3


U12595
up





3


X00437
down

down


AB011089


up



3


AF084260


up



3


AJ133769



up
up


X89066
up





3


AF042181



up
down
up


AF001294


up


AF035283

down

down


X06956
up
up

up
up


X06956
up
up
up



2


AF005392
up
up
up


X01703
up

up


AF035316


up
down

down
3


X02344
up




down


U47634
up





3


X00734
up



up


S75463
up





3


D17517


up


down


U18934


up


AI310002


up



2


AF075599

up

up
up
up


U67122


up



3


X04741
up
up
up



3


U27460

up
up


U30930

down
up
down


3


T79616

up




3


J04973

up
up



2, 3


L32977


up


AA526497

up




3


U30888



up


M36200



up
up


AL050223


up



3


U56833


up



3


L06132

up




3


AJ002428
up
up

up
up


L08666


up



3


AF024710

down

down


AF022375

down

down


M63978

down


up


X51521



down


Z19554

down

down

down
3


AF060902

up

up


D26068
down





2


AB011113

up



down


W27944

down

down


W26496

down

down

down


D14661


up


Y08614


up



3


J04977
up

up
up


3


M30938
up



up


U89436
up
up




3


X56468


up



3


X56468
up

up



3


M92843

down

down

down
2, 3


U07802



down

down


X78992

down

down

down


AL050276

down

down


L11672
down


AD000092
up
up


V00599


up


down


X55989


down


S81916
up
up
up

up


J00153
down

down
down


AL049423

down

down


AF052148
down

down


AL022101
up

up

up


AL118582



down

down


AI095508




up

2


W28612

down

down

down


AL049378
down

down


AI700633

down
up
down


AF070536
down

down


AF052119

up
up
up


AL080113


up


AL049265

down


AL049390



down

down


AF070577

up
up



2


AI827895



down

down


X95677



up
up


AL080093


up


AL049969

down

down

down


AF052141

up
up
up


L27560



down


W27522


up


AL022718
up




down


AJ005694


down



3


AL120687



up
up


AL046322
up


up


AW043812


down


H12054



up


AC003007

up

up


J03071
down


down


M57417
down



down
down


M33764
up


M58028
up


X74262
up


U19796
up


U22028
down


X79568


down


M55914
up


M21154
up


M10905



down


U33429



up


AB014539



down


X63432


up

up


X56841



down


Y00067


up


AF007140
up


X13839


down
down


AF023268


up


AF053356
down


U37122

down

down


AB000450
up


AI126004


up


AF002668
up


X54304



down


U57843
down


X02344
up


X04098
up

up

up


U96074
up


D32053
up

up


U59632


down


X14346
down


Z98046
up


AL096737



down


AB014598



down


U17886


up


AI986201


up


AL080181



down


AB014514
up


R92331



down


U24183


up


D00860
up


U09510


up


AI635895


up


U66033



down


U51334



down


AF020762


up


U24105



up


M36820



down


U59912



up


X63368
up


AF047863



up


U11861
up


AL080122


up


M14648



down


Y14153



up


X81637
down


M88108


up


AF042384
up


AA704137



up


AB011156
up


AI862521
up


AF047469



up


AF025887


up


AF091085
up


AL035494


up


AI540925
down


D32129



down


AB028972
up


AF091071



up


AL040137



down


X15187
up


U48730
down


L08488
up


K03460
up


AF005361



up


M95585



up


M91670
up
















TABLE 5







RT-PCR Confirmation












Gene Bank

AnCg

DLPFC
DLPFC


Acc. No.
Gene Name
BP
AnCg MD
BP
MD





AB020629
ABCA8



down


U37122
ADD3



down


X63575
ATP2B2


down


X71490
ATP6V0D1

up


U66879
BAD
down


J04046
CALM3


up


AF112471
CAMK2B

up


D50310
CCNI
up


AL080061
CLIC4



down


S74445
CRABP1

up

up


U37143
CYP2J2

down

down


M34309
ERBB3



down


M80634
FGFR2



down


(SEQ ID NO: 1)


M64347
FGFR3



down


L08485
GABRA5
up


AF016917
GABRD
down

down


AC004131
GPRC5B

down

down


AB006626
HDAC4

up


L19182
IGFBP7
down

down


X57206
ITPKB



down


X77196
LAMP2



down


M29273
MAG

down

down


X76220
MAL

down

down


Z24725
MIG2



down


AB018342
MYO10



down


M12267
OAT

down

down


Y10275
PSPH



down


AJ001612
PSPHL
down

down


AB007943
RAP1GA1



down


AF060877
RGS20

down

down


AL049538
RIN2



down


AB018305
SPON1

down

down


U58334
TP53BP2

down

down


U28964
YWHAZ
up
















TABLE 6







Summary of Anti-Depressant Treatment Data













Genbank Accession No.
Gene Name
SSvDS
SSvFS
SNvDN
SNvFN
SNvSS





M80899
AHNAK


down

down


K03000
ALDH1A1
down


down


X97074
AP2S1
down


U34846
AQP4
down

down
up


D87468
ARC
up

down
down
down


L04510
ARFD1
down


AF006087
ARPC4




down


J05096
ATP1A2
down


M37457
ATP1A3
down

down

up


J04027
ATP2B1
up


AJ010953
ATP2C1
down


AA877795
ATP6V1D
down


X79888
AUH
down


AF001383
BIN1
down


U68485
BIN1
down


U72649
BTG2
up


X95592
C1D
down


M76559
CACNA2D1
up


AF068862
CALB1
down

down


AF112471
CAMK2B
down


U02390
CAP2
down

down
up


U20325
CART



up


X66364
CDK5
down



up


U03749
CHGA




down


U07223
CHN2



down


AB020709
CNK2
down


S80562
CNN3
down


S74445
CRABP1
up


S74445
CRABP1
up


M27691
CREB1




up


M33146
CSRP1
down


AI810807
DNCI1
down


D86550
DYRK1A
down


U49436
EIF5
down


X51956
ENO2


up

up


L35594
ENPP2




down


L35594
ENPP2




down


D45421
ENPP2




down


AA977580
FACL3
down


D14838
FGF9


up

up


L08485
GABRA5
down


AB020645
GLS
down
up


down


X71973
GPX4


up


X77748
GRM3
down

down


J05459
GSTM3
down


X92814
HRASLS3
down


L12723
HSPA4
down
down


up


L15189
HSPA9B
up


AI912041
HSPE1
down


X57830
HTR2A
down



down


AB017563
IGSF4
up


U96876
INSIG1
down


X07979
ITGB1
down


AL050126
LAP1B
down

down


AF087693
LIN7A
down

down


AL039458
LRIG1




up


M29273
MAG


up


X76220
MAL
down


down


X14474
MAPT
up


M16279
MIC2
down



down


D14812
MRGX
down


AI547258
MT2A
down


down
down


AF013570
MYH11
down

down
down
down


D10667
MYH11
down

down
down
down


D10667
MYH11
down

down
down
down


AF001548
MYH11
down

down
down
down


AF001548
MYH11
down

down
down
down


X77548
NCOA4
up

up


U35139
NDN




down


AA203354
NDUFB3
down


X73066
NME1
down


X17620
NME1
down


AL038662
NME1
down


X58965
NME2
down


M86707
NMT1
down


U61849
NPTX1
down

down


AI198311
NPY




down


U55258
NRCAM
down


AB002341
NRCAM
down


U55258
NRCAM
down


U03985
NSF


down


X75958
NTRK2


down


L13385
PAFAH1B1
down


U24152
PAK1
down


U24152
PAK1
down


U40370
PDE1A
up


AB002345
PER2

down
down
down


V00572
PGK1
down


M83088
PGM1



up


AF017786
PPAP2B
down


M29551
PPP3CB
down

down


X67951
PRDX1
down


U29185
PRNP




up


D00760
PSMA2
down


D26598
PSMB3
down


D29011
PSMB5

up


D44466
PSMD1


up


AB009398
PSMD13
down


M57399
PTN
down


M57399
PTN
down


Z48541
PTPRO

up


M93426
PTPRZ1
down


X63578
PVALB
down



up


U41654
RAGA
down


U27768
RGS4
down

down


D87735
RPL14
down

down


Y00281
RPN1
down


up


M84711
RPS3A
down



down


L10333
RTN1
down
up


M25756
SCG2
down


up


AF049498
SCN2B


down


AF054184
SEC61G


down


Y00757
SGNE1
down


AF007216
SLC4A4
down

down


AF011390
SLC4A4
down

down


D80000
SMC1L1


down


AF053136
SNCB


up


AJ001183
SOX10




up


AB018305
SPON1
down


M32313
SRD5A1
up


AI636761
SST
down
up


down


J00306
SST
down
up


down


X99325
STK25


down


M31303
STMN1
down


U77942
STX7
down
down


up


AF039945
SYNJ2
down


D50495
TCEA2
up

up


X06956
TUBA1




down


AI310002
UBE2D2
down


J04973
UQCRC2
down


D26068
WBSCR1
down


M92843
ZFP36




up


AI095508

down

down


AF070577

down
















TABLE 7a







AnCg BP Genetic Ontology









Genbank




Accession No.
Gene Name
Description







26S proteasome


D00762
PSMA3
proteasome (prosome, macropain) subunit, alpha type, 3


D44466
PSMD1
proteasome (prosome, macropain) 26S subunit, non-ATPase, 1


D00761
PSMB1
proteasome (prosome, macropain) subunit, beta type, 1


D38048
PSMB7
proteasome (prosome, macropain) subunit, beta type, 7


D78151
PSMD2
proteasome (prosome, macropain) 26S subunit, non-ATPase, 2


D29012
PSMB6
proteasome (prosome, macropain) subunit, beta type, 6


D38047
PSMD8
proteasome (prosome, macropain) 26S subunit, non-ATPase, 8


D26598
PSMB3
proteasome (prosome, macropain) subunit, beta type, 3


D26600
PSMB4
proteasome (prosome, macropain) subunit, beta type, 4




synaptic transmission


X82068
GRIA3
glutamate receptor, ionotrophic, AMPA 3


D11428
PMP22
peripheral myelin protein 22


AI636761
SST
somatostatin


AA126505
NCAM1
neural cell adhesion molecule 1


L10338
SCN1B
sodium channel, voltage-gated, type I, beta polypeptide


X81438
AMPH
amphiphysin (Stiff-Man syndrome with breast cancer 128 kDa autoantigen)


M19650
CNP
2′,3′-cyclic nucleotide 3′ phosphodiesterase


L19058
GRIK1
glutamate receptor, ionotropic, kainate 1


AI198311
NPY
neuropeptide Y


U68485
BIN1
bridging integrator 1


M81886
GRIA1
glutamate receptor, ionotropic, AMPA 1


Z11695
MAPK1
mitogen-activated protein kinase 1


X77748
GRM3
glutamate receptor, metabotropic 3


AF052113
RAB14
RAB14, member RAS oncogene family


U40215
SYN2
synapsin II


U61849
NPTX1
neuronal pentraxin I




Chaperone


J04988
HSPCB
heat shock 90 kDa protein 1, beta


U12595
TRAP1
heat shock protein 75


L12723
HSPA4
heat shock 70 kDa protein 4


L08069
DNAJA1
DnaJ (Hsp40) homolog, subfamily A, member 1


AL038340
CRYAB
crystallin, alpha B


AL038340
CRYAB
crystallin, alpha B


X02344
TUBB2
tubulin, beta, 2


AF026166
CCT2
chaperonin containing TCP1, subunit 2 (beta)


M63959
LRPAP1
low density lipoprotein receptor-related protein associated protein 1


L26336
HSPA2
heat shock 70 kDa protein 2


AF026292
CCT7
chaperonin containing TCP1, subunit 7 (eta)


L08069
DNAJA1
DnaJ (Hsp40) homolog, subfamily A, member 1


AA149486
COX17
COX17 homolog, cytochrome c oxidase assembly protein (yeast)


Y00371
HSPA8
heat shock 70 kDa protein 8


X74801
CCT3
chaperonin containing TCP1, subunit 3 (gamma)


X56468
YWHAQ
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein,




theta polypeptide


U41816
PFDN4
prefoldin 4


L15189
HSPA9B
heat shock 70 kDa protein 9B (mortalin-2)


L26336
HSPA2
heat shock 70 kDa protein 2
















TABLE 7b







AnCg MD: Genetic Ontology









Genbank




Accession
Gene
Description


No.
Name
transporter activity





T79616
UQCRB
ubiquinol-cytochrome c reductase binding protein


AI526089
COX5B
cytochrome c oxidase subunit Vb


AL049929
ATP6IP2
ATPase, H+ transporting, lysosomal interacting protein 2


AF006621
C4orf1
chromosome 4 open reading frame 1


L09235
ATP6V1A1
ATPase, H+ transporting, lysosomal 70 kDa, VI subunit A, isoform 1


M22760
COX5A
cytochrome c oxidase subunit Va


U01824
SLC1A2
solute carrier family 1 (glial high affinity glutamate transporter), member 2


N50520
COX7B
cytochrome c oxidase subunit VIIb


AF007216
SLC4A4
solute carrier family 4, sodium bicarbonate cotransporter, member 4


AF011390
SLC4A4
solute carrier family 4, sodium bicarbonate cotransporter, member 4


AA526497
UQCRH
ubiquinol-cytochrome c reductase hinge protein


AA845575
ATP5J
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit F6


X52947
GJA1
gap junction protein, alpha 1, 43 kDa (connexin 43)


D26443
SLC1A3
solute carrier family 1 (glial high affinity glutamate transporter), member 3


AF053070
NDUFV1
NADH dehydrogenase (ubiquinone) flavoprotein 1, 51 kDa


X76228
ATP6V1E1
ATPase, H+ transporting, lysosomal 31 kDa, VI subunit E isoform I


X63575
ATP2B2
ATPase, Ca++ transporting, plasma membrane 2
















TABLE 7c







DLPFC BP Genetic Ontology









Genbank




Accession No.
Gene Name
Description







hydrogen ion transporter activity


AA917672
ATP5L
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit g


AI526089
COX5B
cytochrome c oxidase subunit Vb


J04973
UQCRC2
ubiquinol-cytochrome c reductase core protein II


AF050640
NDUFS2
NADH dehydrogenase (ubiquinone) Fe—S protein 2, 49 kDa (NADH-coenzyme Q reductase)


AL049929
ATP6IP2
ATPase, H+ transporting, lysosomal interacting protein 2


AF047181
NDUFB5
NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 5, 16 kDa


AF047436
ATP5J2
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit f, isoform 2


D14710
ATP5A1
ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit, isoform 1


U09813
ATP5G3
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (subunit 9) isoform 3


N50520
COX7B
cytochrome c oxidase subunit VIIb


AF087135
ATP5H
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit d


D89052
ATP6V0B
ATPase, H+ transporting, lysosomal 21 kDa, V0 subunit c″


X76228
ATP6V1E1
ATPase, H+ transporting, lysosomal 31 kDa, V1 subunit E isoform 1




Chaperone


U56833
VBP1
von Hippel-Lindau binding protein 1


L08069
DNAJA1
DnaJ (Hsp40) homolog, subfamily A, member 1


AL038340
CRYAB
crystallin, alpha B


AL038340
CRYAB
crystallin, alpha B


X15183
HSPCA
heat shock 90 kDa protein 1, alpha


X56468
YWHAQ
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide


L24804
TEBP
unactive progesterone receptor, 23 kD


W28616
HSPCB
heat shock 90 kDa protein 1, beta


D49738
CKAP1
cytoskeleton-associated protein 1


AF026166
CCT2
chaperonin containing TCP1, subunit 2 (beta)


M63959
LRPAP1
low density lipoprotein receptor-related protein associated protein 1


X87949
HSPA5
heat shock 70 kDa protein 5 (glucose-regulated protein, 78 kDa)


L26336
HSPA2
heat shock 70 kDa protein 2


M22382
HSPD1
heat shock 60 kDa protein 1 (chaperonin)


L08069
DNAJA1
DnaJ (Hsp40) homolog, subfamily A, member 1


AF035316
TUBB
tubulin, beta polypeptide


AI912041
HSPE1
heat shock 10 kDa protein 1 (chaperonin 10)


Y00371
HSPA8
heat shock 70 kDa protein 8


X74801
CCT3
chaperonin containing TCP1, subunit 3 (gamma)


X56468
YWHAQ
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, theta polypeptide


W29042
CIA30
CGI-65 protein


L15189
HSPA9B
heat shock 70 kDa protein 9B (mortalin-2)




OXPHOS


X71490
ATP6V0D1
ATPase, H+ transporting, lysosomal 38 kDa, V0 subunit d isoform 1


D14710
ATP5A1
ATP synthase, H+ transporting, mitochondrial F1 complex, alpha subunit, isoform 1, cardiac muscle


U09813
ATP5G3
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (subunit 9) isoform 3


AF087135
ATP5H
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit d


AA845575
ATP5J
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit F6


AF047436
ATP5J2
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit f, isoform 2


AA917672
ATP5L
ATP synthase, H+ transporting, mitochondrial F0 complex, subunit g


X83218
ATP5O
ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit


D89052
ATP6V0B
ATPase, H+ transporting, lysosomal 21 kDa, V0 subunit c″


AA056747
ATP6V1A1
ATPase, H+ transporting, lysosomal 70 kDa, V1 subunit A


X76228
ATP6V1E1
ATPase, H+ transporting, lysosomal 31 kDa, V1 subunit E isoform 1


AI526089
COX5B
cytochrome c oxidase subunit Vb


N50520
COX7B
cytochrome c oxidase subunit VIIb


AC002400
NDUFAB1
NADH dehydrogenase (ubiquinone) 1, alpha/beta subcomplex, 1, 8 kDa


AA203354
NDUFB3
NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 3, 12 kDa


AF047181
NDUFB5
NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 5, 16 kDa


AF050640
NDUFS2
NADH dehydrogenase (ubiquinone) Fe—S protein 2, 49 kDa (NADH-coenzyme Q reductase)


J04973
UQCRC2
ubiquinol-cytochrome c reductase core protein II


L32977
UQCRFS1
ubiquinol-cytochrome c reductase, Rieske iron-sulfur polypeptide 1


U17886
SDHB
succinate dehydrogenase complex, subunit B, iron sulfur (Ip)
















TABLE 7d







DLPFC MD Genetic Ontology









Genbank




Accession
Gene


No.
Name
Description







transmission of nerve impulse


D11428
PMP22
peripheral myelin protein 22


AF049498
SCN2B
sodium channel, voltage-gated, type II, beta polypeptide


M82919
GABRB3
gamma-aminobutyric acid (GABA) A receptor, beta 3


X59834
GLUL
glutamate-ammonia ligase (glutamine synthase)


X81438
AMPH
amphiphysin (Stiff-Man syndrome with breast cancer 128 kDa autoantigen)


M54927
PLP1
proteolipid protein 1 (Pelizaeus-Merzbacher disease, spastic paraplegia 2,




uncomplicated)


Z11695
MAPK1
mitogen-activated protein kinase 1


U01824
SLC1A2
solute carrier family 1 (glial high affinity glutamate transporter), member 2


M32886
SRI
sorcin


U40215
SYN2
synapsin II


X15376
GABRG2
gamma-aminobutyric acid (GABA) A receptor, gamma 2


D26443
SLC1A3
solute carrier family 1 (glial high affinity glutamate transporter), member 3


X68194
SYPL
synaptophysin-like protein


U61849
NPTX1
neuronal pentraxin I




neurogenesis


D11428
PMP22
peripheral myelin protein 22


U30930
UGT8
UDP glycosyltransferase 8 (UDP-galactose ceramide galactosyltransferase)


W28770
NP25
neuronal protein


M54927
PLP1
proteolipid protein 1 (Pelizaeus-Merzbacher disease, spastic paraplegia 2,




uncomplicated)


D83017
NELL1
NEL-like 1 (chicken)


U34846
AQP4
aquaporin 4


Z70276
FGF12
fibroblast growth factor 12


M80899
AHNAK
AHNAK nucleoprotein (desmoyokin)


M57399
PTN
pleiotrophin (heparin binding growth factor 8, neurite growth-promoting factor 1)


AF016004
GPM6B
glycoprotein M6B


X70326
MLP
MARCKS-like protein


AF036268
SH3GL2
SH3-domain GRB2-like 2


M93426
PTPRZ1
protein tyrosine phosphatase, receptor-type, Z polypeptide 1


U61849
NPTX1
neuronal pentraxin I


M93650
PAX6
paired box gene 6 (aniridia, keratitis)




phosphoric ester hydralase activity


X55740
NT5E
5′-nucleotidase, ecto (CD73)


AF001601
PON2
paraoxonase 2


X68277
DUSP1
dual specificity phosphatase 1


L35594
ENPP2
ectonucleotide pyrophosphatase/phosphodiesterase 2 (autotaxin)


AB013382
DUSP6
dual specificity phosphatase 6


L05147
DUSP3
dual specificity phosphatase 3 (vaccinia virus phosphatase VH1-related)


Z48541
PTPRO
protein tyrosine phosphatase, receptor type, O


N36638
PPP1R3C
protein phosphatase 1, regulatory (inhibitor) subunit 3C


AJ001612
PSPHL
phosphoserine phosphatase-like


AF017786
PPAP2B
phosphatidic acid phosphatase type 2B


U60644
PLD3
phospholipase D3


AF056490
PDE8A
phosphodiesterase 8A


M93426
PTPRZ1
protein tyrosine phosphatase, receptor-type, Z polypeptide 1
















TABLE 8







Selected Potential Druggable Targets










Genbank





Accession
Gene


No.
Name
Target category
Description





AB020629
ABCA8
transporter
ATP-binding cassette, sub-family A (ABC1), member 8


X63575
ATP2B2
transporter
ATPase, Ca++ transporting, plasma membrane 2


X71490
ATP6V0D1
transporter
ATPase, H+ transporting, lysosomal 38 kDa, V0 subunit d isoform 1


S74445
CRABP1
transporter
cellular retinoic acid binding protein 1


M34309
ERBB3
tyrosine kinase
v-erb-b2 erythroblastic leukemia viral oncogene homolog 3 (avian)




receptor


M80634
FGFR2
tyrosine kinase
fibroblast growth factor receptor 2 (bacteria-expressed kinase,


(SEQ ID

receptor
keratinocyte growth factor receptor, craniofacial dysostosis 1, Crouzon


NO: 1)


syndrome, Pfeiffer syndrome, Jackson-Weiss syndrome)


M64347
FGFR3
tyrosine kinase
fibroblast growth factor receptor 3 (achondroplasia, thanatophoric dwarfism)




receptor


AC004131
GPRC5B
GPCR
G protein-coupled receptor, family C, group 5, member B


X77196
LAMP2
ligand for cell
lysosomal-associated membrane protein 2




adhesion




molecule


U37122
ADD3
regulator of
adducin 3 (gamma)




kinase


U66879
BAD
regulator of
BCL2-antagonist of cell death




protease


AB007943
RAP1GA1
regulator of
RAP1, GTPase activating protein 1




kinase


AF060877
RGS20
regulator of
regulator of G-protein signalling 20




GTPase


AL049538
RIN2
regulator of
Ras and Rab interactor 2




GTPase


U58334
TP53BP2
reuglator of
tumor protein p53 binding protein, 2




protein




degradation


U28964
YWHAZ
regulator of
tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein, zeta polypeptide




enzyme


J04046
CALM3
kinase
calmodulin 3 (phosphorylase kinase, delta)


AF112471
CAMK2B
kinase
calcium/calmodulin-dependent protein kinase (CaM kinase) II beta


D50310
CCNI
regulator of
cyclin I




kinase


U37143
CYP2J2
monooxygenase
cytochrome P450, family 2, subfamily J, polypeptide 2


AB006626
HDAC4
enzyme
histone deacetylase 4


X57206
ITPKB
kinase
inositol 1,4,5-trisphosphate 3-kinase B


M12267
OAT
enzyme
ornithine aminotransferase (gyrate atrophy)


Y10275
PSPH
enzyme
phosphoserine phosphatase


AJ001612
PSPHL
enzyme
phosphoserine phosphatase-like


AL080061
CLIC4
channel
chloride intracellular channel 4


L08485
GABRAS
channel
gamma-aminobutyric acid (GABA) A receptor, alpha 5


AF016917
GABRD
channel
gamma-aminobutyric acid (GABA) A receptor, delta


L19182
IGFBP7
regulator of
insulin-like growth factor binding protein 7




receptor ligand


M29273
MAG
myelination
myelin associated glycoprotein


X76220
MAL
myelination
mal, T-cell differentiation protein


Z24725
MIG2
signal
mitogen inducible 2




transduction


AB018342
MYO10
partner for
myosin X




calmodulin-like




protein


AB018305
SPON1
axon growth
spondin 1, (f-spondin) extracellular matrix protein




guidance








Claims
  • 1. A method for determining whether a subject is predisposed for major depression disorder, the method comprising the steps of: (i) isolating a subject's brain tissue, wherein the brain tissue is dorsolateral prefrontal cortex tissue;(ii) contacting the subject's isolated brain tissue with a nucleic acid reagent that selectively associates with a polynucleotide with 95% identity to SEQ ID NO. 1;(iii) detecting the level of reagent that selectively associates with the said polynucleotide; and(iv) comparing the detected level of selectively associated reagent with a control, whereby if the detected level is significantly less than the control, an increased likelihood that the subject has or is predisposed for major depression disorder is determined; and whereby, if the detected level is not significantly less than the control, an increase in said likelihood is not determined by the method.
  • 2.-29. (canceled)
  • 30. The method of claim 1, wherein the subject is deceased.
CROSS-REFERENCES TO RELATED APPLICATIONS

The present application claims the benefit of U.S. Ser. No. 60/423,247, filed Nov. 1, 2002 and U.S. Ser. No. 60/431,454, filed Dec. 6, 2002, the disclosures of which are hereby incorporated by reference in their entirety for all purposes.

Provisional Applications (2)
Number Date Country
60431454 Dec 2002 US
60423247 Nov 2002 US
Continuations (1)
Number Date Country
Parent 10701263 Nov 2003 US
Child 12154419 US