Cardiac pressure overload associated genes

Abstract
The present invention identifies genes whose gene products are differentially expressed pressure overload of the heart. The invention provides methods for diagnosing or assessing an individual's susceptibility to heart failure from many etiologies, as well as the presence and severity of hypertrophy, chamber enlargement, or systolic heat failure. Also provided are therapeutic methods for treating a heart patient or methods for prophylactically treating an individual susceptible to heart failure. Additionally, the invention describes screening methods for identifying agents that can be administered to treat individuals that have suffered a heart attack or are at risk of heart failure.
Description
INTRODUCTION

Heart failure is the leading cause of morbidity in western cultures. Congestive heart failure (CHF) develops when plasma volume increases and fluid accumulates in the lungs, abdominal organs (especially the liver), and peripheral tissues. In many forms of heart disease, the clinical manifestations of HF may reflect impairment of the left or right ventricle. Left ventricular (LV) failure characteristically develops in coronary artery disease, hypertension, cardiac valvular disease, many forms of cardiomyopathy, and with congenital defects. Right ventricular (RV) failure is most commonly caused by prior LV failure, which increases pulmonary venous pressure and leads to pulmonary arterial hypertension and tricuspid regurgitation. Heart failure is manifest by systolic or diastolic dysfunction, or both. Combined systolic and diastolic abnormalities are common.


In systolic dysfunction, primarily a problem of ventricular contractile dysfunction, the heart fails to provide tissues with adequate circulatory output. A wide variety of defects in energy utilization, energy supply, electrophysiologic functions, and contractile element interaction occur, which appear to reflect abnormalities in intracellular Ca++ modulation and adenosine triphosphate (ATP) production. Systolic dysfunction has numerous causes; the most common are coronary artery disease, hypertension, valvular disease, and dilated cardiomyopathy. Additionally, there are many known and probably many unidentified causes for dilated myocardiopathy, e.g. virus infection, toxic substances such as alcohol, a variety of organic solvents, certain chemotherapeutic drugs (e.g., doxorubicin), β-blockers, Ca blockers, and antiarrhythmic drugs.


Diastolic dysfunction accounts for 20 to 40% of cases of heart failure. It is generally associated with prolonged ventricular relaxation time, as measured during isovolumic relaxation. Resistance to filling directly relates to ventricular diastolic pressure; this resistance increases with age, probably reflecting myocyte loss and increased interstitial collagen deposition. Diastolic dysfunction is presumed to be dominant in hypertrophic cardiomyopathy, circumstances with marked ventricular hypertrophy, e.g. hypertension, advanced aortic stenosis, and amyloid infiltration of the myocardium. Without intervention, hypertrophic cardiomyopathy and diastolic dysfunction often progress to systolic dysfunction and overt, symptomatic heart failure in the natural course of the disease.


The mammalian heart responds to pressure overload by undergoing left ventricular hypertrophy (LVH) and left atrial enlargement (LAE). These adaptive responses to increases in hemodynamic overload involve many alterations in myocardial structure and function. Although these responses are necessary in the short term to maintain cardiac output in the face of increased afterload, LVH and LAE are associated with increased risk for sudden death and progression to heart failure, the leading cause of morbidity in western cultures. A detailed understanding of the molecular events accompanying these changes is an important step toward the ability to interrupt or reverse their progression.


While the LV takes the brunt of the pressure insult, during pressure overload the left atrium faces physiological challenges due to mitral regurgitation and increased wall stress, which result in enlargement and remodeling. Many of the most important clinical complications of hypertrophic cardiomyopathy, valvulvar heart disease, and congestive heart failure are due to atrial enlargement, and include atrial fibrillation and other electrophysiological disturbances, as well as hemodynamic compromise caused by decreased ventricular filling. In humans, the hemodynamic and electrophysiological sequelae of left atrial enlargement are nearly as important as those stemming from LVH.


In view of the importance of cardiomyopathy for human mortality and morbidity, the identification of genes involved in the disease, and development of methods of treatment is of great interest.


SUMMARY OF THE INVENTION

The present invention provides methods and compositions for the diagnosis and treatment of heart diseases relating to pressure overload, including but not limited to those which lead to heart failure. Among other pathologies, pressure overload induces the development of left ventricular hypertrophy (LVH) and left atrial enlargement (LAE) in the mammalian heart.


Specifically, genes are identified and described herein that are differentially expressed following induced pressure overload of the heart. The detection of the coding sequence and/or polypeptide products of these genes provides useful methods for early detection, diagnosis, staging, and monitoring of conditions leading to hypertrophy and enlargement of the heart, e.g. by the analysis of blood samples, biopsy material, in vivo imaging, metabolic assays for enzymatic activities, and the like. Expression signatures of a set of genes in heart tissue may also be evaluated for conditions indicative of pressure overload of the heart.


The invention also provides methods for the identification of compounds that modulate the expression of genes or the activity of gene products in heart diseases involving pressure overload, as well as methods for the treatment of disease by administering such compounds to individuals exhibiting heart failure symptoms or tendencies.




BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. Summary of data analysis. After background subtraction and dye bias normalization, poor quality features with low signal intensity were excluded from further analysis. Features with valid values in at least 66% of the experiments for each pairwise comparison (e.g., LA>66% AND TAC LA>66%) were retained for further analysis using SAM and t-test. Lists of genes identified as up-or downregulated by SAM were then mapped to GO terms and Fisher's exact test used to identify biological process groups with significant groupwide regulation.



FIG. 2. Hierarchical clustering. Left atria from TAC animals cluster more closely with ventricles than atria.



FIGS. 3A-3B. SAM analysis. Heatmaps of the top most significantly up- and downregulated genes in TAC LA(a) and LV(b). The order of the genes reflects decreasing SAM score, or d-statistic.



FIG. 4. Heatmap of the 891 upregulated and 1001 downregulated genes identified by SAM in the TAC LA. Blocks of genes with ventricle-like, atrial-like, and novel TAC expression patterns are highlighted. Red color denotes high expression, green denotes low expression level.



FIG. 5A-5C. Top statistically significantly regulated gene ontology biological process groups for TAC LA(a and b) and LV(c). The figure lists the biological process group, the total number of annotated genes in that group on the array, the number of genes identified by SAM as up- or downregulated in the group, and the one sided Fisher's exact p-value for differential regulation of each group.



FIG. 6. Energy pathway genes downregulated in TAC LA. This figure shows the breadth of downregulation of the TCA cycle, fatty acid metabolism, and oxidative phosphorylation genes that occur in response to pressure overload in the LA. Downregulated genes from each oxidative phosphorylation complex are listed in the graphic. A similar number of genes is downregulated in the TAC LV.



FIG. 7. Comparison of microarray and qRT-PCR results. Expression is plotted as log(10) fold expression change versus sham operated control for LA and LV tissues. This figure illustrates that fold changes in expression are usually greater in the LA than LV. Results are shown for the 9 regulated genes (frizzled-related protein (Frzb), cyclin D1, TGFβ2, HIF1a, endothelin receptor b (Ednrb), four-and-a-half LIM domains 2 (FHL2), regulator of G-protein signaling 2 (RGS2), diacylglycerol O-acyltransferase 2 (DGAT2), and homeodomain-only protein (Hop)) for which qRT-PCR validation was performed.




Table I pg. 1-pg. 26 provides a list of genetic sequences differentially expressed following transverse aortic constriction. The Stanford Gene ID refers to the internet address of genome-www5.stanford.edu, which provides a database including Genbank accession numbers. Pages 1-12 provide for significantly upregulated genes, and pages 13-26 provide for significantly down-regulated genes. Table IA pg. 1-pg. 3 provides a subset of upregulated genes of interest, and includes under the heading “UGRepAcc [A]” the accession numbers for representative genetic sequences available at Genbank. Under the heading “LLRepProtAcc [A]” are provided accession numbers for representative protein sequences at Genbank. Table IB provides a further subset of sequences of interest, similarly annotated. The sequences of Table IA or Table IB pg. 1-pg. 2 may be further sub-divided according to their representation in Tables II, III or IV.


Table II pg. 1-pg. 4 provides a list of genetic sequences set forth in Table I, which are differentially expressed following transverse aortic constriction, which are of interest for serologic assays. Table II further provides Genbank accession numbers, Genbank accession numbers of human homologs, and whether the gene is upregulated in transverse aortic constriction in the left atrium (designated UP TAC LA) and/or the left ventricle (designated UP TAC LV).


Table III pg. 1-pg. 4 provides a list of genetic sequences set forth in Table I, differentially expressed following transverse aortic constriction, which are of interest for imaging assays. Table III further provides Genbank accession numbers, Genbank accession numbers of human homologs, and whether the gene is upregulated in transverse aortic constriction in the left atrium (designated UP TAC LA) and/or the left ventricle (designated UP TAC LV).


Table IV pg 1-pg. 3 provides a list of genetic sequences set forth in Table I, differentially expressed following transverse aortic constriction, which are of interest for metabolic assays. Table IV further provides Genbank accession numbers, Genbank accession numbers of human homologs, and whether the gene is upregulated in transverse aortic constriction in the left atrium (designated UP TAC LA) and/or the left ventricle (designated UP TAC LV).


DETAILED DESCRIPTION OF THE EMBODIMENTS

Methods and compositions for the diagnosis and treatment of heart diseases involving pressure overload, including but not limited to cardiomyopathies; heart failure; and the like, are provided. The invention is based, in part, on the evaluation of the expression and role of genes that are differentially expressed in response to pressure overload, e.g. during left atrial enlargement and left ventricular hypertrophy. The right chambers may have similar changes in gene expression in association with pathologies such as pulmonary hypertension, etc. Such sequences are useful in the diagnosis and monitoring of cardiac disease. The gene products are also useful as therapeutic targets for drug screening and action.


To systematically investigate the transcriptional changes that mediate these processes, a genome-wide transcriptional profiling of each of the four heart chambers was performed following transverse aortic constriction. It is shown herein that during enlargement, the left atrium undergoes radical changes in gene transcription. Structural changes in the LA and LV are correlated with significant changes in the transcriptional profile of these chambers. Statistical analysis of the results identified biological process groups with significant group-wide changes, including angiogenesis, fatty acid oxidation, oxidative phosphorylation, cytoskeletal and matrix reorganization, and G-protein coupled receptor signaling. The genes thus identified, and their classification into biological process groups, are provided in Table I. Subsets of the upregulated genes are provided in Tables IA and IB. Table IA is a subset of Table I, and Table IB is a subset of Table IA.


For some methods of the invention, a panel of sequences will be selected, comprising, for example, at least one, at least two, at least three, at least four, at least five, at least ten, at least 15, at least 20, and may include substantially all the sequences of a specific Table (I, IA, IB; and/or II, III, IV), or may be limited to not more than about 100 distinct sequences, not more than about 50 distinct sequences, not more than about 25 distinct sequences, and the like. The selection of sequences for inclusion in arrays, use in diagnostic panels, and the like may be based on representation of a sequence in one or more of the sub-tables, e.g. selecting sequences present in Table IA or Table IB; representation of a sequence in both Table IB and Table II; Table IB and Table III; Table IB and Table IV, and the like. The use of human homologs of the sequences is of particular interest. Selection of sequences may alternatively be based on a cut-off for significance or for fold-change in expression, e.g. those sequences have a fold-change of at least about 3, at least about 6, at least 10, or more. Selection of sequences may also be based on biological activity grouping, e.g. using the grouping as set forth in FIG. 5, genes can be divided into energy pathways, cell adhesion, cell communication, signal transduction, etc., where


The identification of pressure overload associated genes provides diagnostic and prognostic methods, which detect the occurrence of a disorder, e.g. cardiomyopathy; atrial enlargement; myocardial hypertrophy; etc., particularly where such a disorder is indicative of a propensity for heart failure; or assess an individual's susceptibility to such disease, by detecting altered expression of pressure overload associated genes. Early detection of genes or their products can be used to determine the occurrence of developing disease, thereby allowing for intervention with appropriate preventive or protective measures.


Various techniques and reagents find use in the diagnostic methods of the present invention. In one embodiment of the invention, blood samples, or samples derived from blood, e.g. plasma, serum, etc. are assayed for the presence of polypeptides encoded by pressure overload associated genes, e.g. cell surface and, of particular interest, secreted polypeptides. Such polypeptides may be detected through specific binding members. The use of antibodies for this purpose is of particular interest. Various formats find use for such assays, including antibody arrays; ELISA and RIA formats; binding of labeled antibodies in suspension/solution and detection by flow cytometry, mass spectroscopy, and the like. Detection may utilize one or a panel of antibodies. A subset of genes and gene products of interest for serologic assays are provided in Table II. These sequences may be further defined by reference to the sequences set forth in Table IA and/or Table IB, i.e. sequences that are present in both Table II, and Table IA or Table IB, may be of particular interest for serologic assays.


In another embodiment, in vivo imaging is utilized to detect the presence of pressure overload associated gene on heart tissue. Such methods may utilize, for example, labeled antibodies or ligands specific for cell surface pressure overload associated gene products. Included for such methods are gene products differentially expressed on chambers of the heart, which can be localized by in situ binding of a labeled reagent. In these embodiments, a detectably-labeled moiety, e.g., an antibody, ligand, etc., which is specific for the polypeptide is administered to an individual (e.g., by injection), and labeled cells are located using standard imaging techniques, including, but not limited to, magnetic resonance imaging, computed tomography scanning, and the like. Detection may utilize one or a cocktail of imaging reagents. A subset of genes and gene products of interest for imaging assays are provided in Table III. These sequences may be further defined by reference to the sequences set forth in Table IA and/or Table IB, i.e. sequences that are present in both Table III, and Table IA or Table IB, may be of particular interest for imaging assays.


In another embodiment, metabolic tests are performed, e.g. with a labeled substrate, to determine the level of enzymatic activity of a pressure overload associated gene product. Gene products of interest for such assays include enzymes whose reaction product is readily detected, e.g. in blood samples. It is shown herein, for example, that oxidative phosphorylation is markedly downregulated during left ventricular hypertrophy and atrial enlargement, and provides a marker for risk of heart failure. A subset of genes and gene products of interest for metabolic assays are provided in Table IV. These sequences may be further defined by reference to the sequences set forth in Table IA and/or Table IB, i.e. sequences that are present in both Table IV and Table IA or Table IB may be of particular interest for metabolic assays.


In another embodiment, an mRNA sample from heart tissue, preferably from one or more chambers affected by pressure overload, is analyzed for the genetic signature indicating pressure overload, and diagnostic of a tendency to heart failure. Expression signatures typically utilize a panel of genetic sequences, e.g. a microarray format; multiplex amplification, etc., coupled with analysis of the results to determine if there is a statistically significant match with a disease signature.


Functional modulation of pressure overload associated genes and their products provides a point of intervention to block the pathophysiologic processes of hypertrophy and enlargement, and also provides therapeutic intervention in other cardiovascular system diseases with similar pathophysiologies. These genes and their products can also be used to prevent, attenuate or reduce damage in prophylactic strategies in patients at high-risk of heart failure. Genes whose expression is altered during development of hypertrophy or enlargement may be cardiodamaging. Agent(s) that inhibit the activity or expression of cardiodamaging genes can be used as a therapeutic or prophylactic agent. The agent that acts to decrease such gene product activity can be an anti-sense or RNAi nucleic acid that includes a segment corresponding a cardiodamaging gene, or any agent that acts as a direct or indirect inhibitor of the gene product, e.g. a pharmacological agonist, or partial agonist.


Disease Conditions

Heart failure is a general term that describes the final common pathway of many disease processes. Heart failure is usually caused by a reduction in the efficiency of cardiac muscle contraction. However, mechanical overload with normal or elevated cardiac contraction can also cause heart failure. This mechanical overload may be due to arterial hypertension, or stenosis or leakage of the aortic, mitral, or pulmonary valves, or other causes. The initial response to overload is usually hypertrophy (cellular enlargement) of the myocardium to increase force production, returning cardiac output (CO) to normal levels. Typically, a hypertrophic heart has impaired relaxation, a syndrome referred to as diastolic dysfunction. In the natural history of the disease, compensatory hypertrophy in the face of ongoing overload is followed by thinning, dilation, and enlargement, resulting in systolic dysfunction, also commonly known as heart failure. This natural progression typically occurs over the course of months to many years in humans, depending on the severity of the overload stimulus. Intervention at the hypertrophy stage can slow or prevent the progression to the clinically significant systolic dysfunction stage. Thus, diagnosis in the early hypertrophy stage provides unique therapeutic opportunities. The most common cause of congestive heart failure is coronary artery disease, which can cause a myocardial infarction (heart attack), which forces the heart to carry out the same work with fewer heart cells. The result is a pathophysiological state where the heart is unable to pump out enough blood to meet the nutrient and oxygen requirements of metabolizing tissues or cells.


in LV failure, CO declines and pulmonary venous pressure increases. Elevated pulmonary capillary pressure to levels that exceed the oncotic pressure of the plasma proteins (about 24 mm Hg) leads to increased lung water, reduced pulmonary compliance, and a rise in the O2 cost of the work of breathing. Pulmonary venous hypertension and edema resulting from LV failure significantly alter pulmonary mechanics and, thereby, ventilation/perfusion relationships. When pulmonary venous hydrostatic pressure exceeds plasma protein oncotic pressure, fluid extravasates into the capillaries, the interstitial space, and the alveoli.


Increased heart rate and myocardial contractility, arteriolar constriction in selected vascular beds, venoconstriction, and Na and water retention compensate in the early stages for reduced ventricular performance. Adverse effects of these compensatory efforts include increased cardiac work, reduced coronary perfusion, increased cardiac preload and afterload, fluid retention resulting in congestion, myocyte loss, increased K excretion, and cardiac arrhythmia.


The mechanism by which an asymptomatic patient with cardiac dysfunction develops overt CHF is unknown, but it begins with renal retention of Na and water, secondary to decreased renal perfusion. Thus, as cardiac function deteriorates, renal blood flow decreases in proportion to the reduced CO, the GFR falls, and blood flow within the kidney is redistributed. The filtration fraction and filtered Na decrease, but tubular resorption increases.


Although symptoms and signs, for example exertional dyspnea, orthopnrea, edema, tachycardia, pulmonary rales, a third heart sound, jugular venous distention, etc. have a diagnostic specificity of 70 to 90%, the sensitivity and predictive accuracy of conventional tests are low. Elevated levels of B-type natriuretic peptide may be diagnostic. Adjunctive tests include CBC, blood creatinine, BUN, electrolytes (eg, Mg, Ca), glucose, albumin, and liver function tests. ECG may be performed in all patients with HF, although findings are not specific.


Patients diagnosed as being at risk for heart failure by the methods of the invention may be appropriately treated to reduce the risk of heart failure. Drug treatment of systolic dysfunction primarily involves diuretics, ACE inhibitors, digitalis, and β-blockers; most patients are treated with at least two of these classes. Addition of hydralazine and isosorbide dinitrate to standard triple therapy of HF may improve hemodynamics and exercise tolerance and reduce mortality in refractory patients. The angiotensin II receptor blocker losartan has effects similar to those of ACE inhibitors.


Digitalis preparations have many actions, including weak inotropism, and blockade of the atrioventricular node. Digoxin is the most commonly prescribed digitalis preparation. Digitoxin, an alternative in patients with known or suspected renal disease, is largely excreted in the bile and is thus not influenced by abnormal renal function.


With careful administration of β-blockers, some patients, especially those with idiopathic dilated cardiomyopathy, will improve clinically and may have reduced mortality. Carvedilol, a 3rd-generation nonselective β-blocker, is also a vasodilator with α blockade and an antioxidant activity. Vasodilators such as nitroglycerin or nitroprusside improve ventricular function by reducing systolic ventricular wall stress, aortic impedance, ventricular chamber size, and valvular regurgitation.


Arterial hypertension, or the elevation of systolic and/or diastolic BP, either primary or secondary, is frequently associated with pressure overload of the heart, and is an important risk factor for heart failure. Hypertensive patients may be analyzed by the diagnostic methods of the invention, in order to determine whether there is a concurrent development of hypertrophy, diastolic dysfunction, and a tendency to heart failure. Criteria for hypertension is typically over about 140 mm Hg systolic blood pressure, and/or diastolic blood pressure of greater than about 90 mm Hg.


Primary (essential) hypertension is of unknown etiology; its diverse hemodynamic and pathophysiologic derangements are unlikely to result from a single cause. Heredity is a predisposing factor, but the exact mechanism is unclear. The pathogenic mechanisms can lead to increased total peripheral vascular resistance by inducing vasoconstriction and to increased cardiac output.


While no early pathologic changes occur in primary hypertension, ultimately, generalized arteriolar sclerosis develops. Left ventricular hypertrophy and, eventually, dilation develop gradually. Coronary, cerebral, aortic, renal, and peripheral atherosclerosis are more common and more severe in hypertensives because hypertension accelerates atherogenesis.


Valvular disease, including stenosis or insufficiency of the aortic, mitral, pulmonary, or tricuspid valves, is also frequently associated with overload of the heart, and is another important risk factor for heart failure. Patients with valvular disease may be analyzed by the diagnostic methods of the invention, in order to determine whether other is a concurrent development of hypertrophy, diastolic dysfunction, and a tendency to heart failure. Valvular disease is typically diagnosed by echocardiographic measurement of significant valvular stenoses or insufficiencies. Valvular heart disease has many etiologies, including but not limited to rheumatic heart disease, congenital valve defects, endocarditis, aging, etc. The pathogenic mechanism whereby valvular disease leads to heart failure is the obstruction of blood outflow from various chambers of the heart, thus increasing load.


Cardiomyopathy refers to a structural or functional abnormality of the ventricular myocardium. Cardiomyopathy has many causes. Pathophysiologic classification (dilated congestive, hypertrophic, or restrictive cardiomyopathy) by means of history, physical examination, and invasive or noninvasive testing may be performed. If no cause can be found, cardiomyopathy is considered primary or idiopathic.


Dilated congestive cardiomyopathies include disorders of myocardial function with heart failure, in which ventricular dilation and systolic dysfunction predominate. The most common identifiable cause in temperate zones is diffuse coronary artery disease with diffuse ischemic myopathy. Most commonly, at presentation there is chronic myocardial fibrosis with diffuse loss of myocytes. Diagnosis depends on the characteristic history and physical examination and exclusion of other causes of ventricular failure. The ECG may show sinus tachycardia, low-voltage QRS, and nonspecific ST segment depression with low-voltage or inverted T waves.


Hypertrophic cardiomyopathies are congenital or acquired disorders characterized by marked ventricular hypertrophy with diastolic dysfunction that may develop in the absence of increased afterload. The cardiac muscle is abnormal with cellular and myofibrillar disarray, although this finding is not specific to hypertrophic cardiomyopathy. The interventricular septum may be hypertrophied more than the left ventricular posterior wall (asymmetric septal hypertrophy). In the most common asymmetric form of hypertrophic cardiomyopathy, there is marked hypertrophy and thickening of the upper interventricular septum below the aortic valve. During systole, the septum thickens and the anterior leaflet of the mitral valve, already abnormally oriented due to the abnormal shape of the ventricle, is sucked toward the septum, producing outflow tract obstruction. Clinical manifestations may occur alone or in any combination: Chest pain is usually typical angina related to exertion. Syncope is usually exertional and due to a combination of cardiomyopathy, arrhythmia, outflow tract obstruction, and poor diastolic filling of the ventricle. Dyspnea on exertion results from poor diastolic compliance of the left ventricle, which leads to a rapid rise in left ventricular end-diastolic pressure as flow increases. Outflow tract obstruction, by lowering cardiac output, may contribute to the dyspnea.


Restrictive cardiomyopathies are characterized by rigid, noncompliant ventricular walls that resist diastolic filling of one or both ventricles, most commonly the left. The cause is usually unknown. Amyloidosis involving the myocardium is usually systemic, as is iron infiltration in hemochromatosis. Sarcoidosis and Fabry's disease involve the myocardium, and nodal conduction tissue can be involved. Löffler's disease (a subcategory of hypereosinophilic syndrome with primary cardiac involvement) is a cause of restrictive cardiomyopathy. It occurs in the tropics. It begins as an acute arteritis with eosinophilia, with subsequent thrombus formation on the endocardium, chordae, and atrioventricular valves, progressing to fibrosis. Endocardial fibrosis occurs in temperate zones and involves only the left ventricle. The main hemodynamic consequence of these pathologic states is diastolic dysfunction with a rigid, noncompliant chamber with a high filling pressure. Systolic function may deteriorate if compensatory hypertrophy is inadequate in cases of infiltrated or fibrosed chambers. Mural thrombosis and systemic emboli can complicate the restrictive or obliterative variety.


Identification of Genes Associated With Pressure Overload

In order to identify pressure overload associated genes, tissue was taken from the chambers of the heart following transverse aortic constriction, or from control, unaffected tissue. RNA, either total or mRNA, is isolated from such tissues. See, for example, Sambrook et al., 1989, Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, New York; and Ausubel, F. M. et al., eds., 1987-1993, Current Protocols in Molecular Biology, John Wiley & Sons, Inc., New York, both of which are incorporated herein by reference in their entirety. Differentially expressed genes are detected by comparing gene expression levels between the experimental and control conditions. Transcripts within the collected RNA samples that represent differentially expressed genes may be identified by utilizing a variety of methods known to those of skill in the art, including differential screening, subtractive hybridization, differential display, or hybridization to an array comprising a plurality of gene sequences.


“Differential expression” as used herein refers to both quantitative as well as qualitative differences in the genes' temporal and/or tissue expression patterns. Thus, a differentially expressed gene may have its expression activated or inactivated in normal versus disease conditions, or in control versus experimental conditions. Preferably, a regulated gene will exhibit an expression pattern within a given tissue or cell type that is detectable in either control or disease subjects, but is not detectable in both. Detectable, as used herein, refers to an RNA expression pattern or presence of polypeptide product that is detectable via the standard techniques of differential display, reverse transcription-(RT-) PCR and/or Northern analyses, ELISA, RIA, metabolic assays, etc., which are well known to those of skill in the art. Generally, differential expression means that there is at least a 20% change, and in other instances at least a 2-, 3-, 5- or 10-fold difference between disease and control tissue expression. The difference usually is one that is statistically significant, meaning that the probability of the difference occurring by chance (the P-value) is less than some predetermined level (e.g., 5%). Usually the confidence level (P value) is <0.05, more typically <0.01, and in other instances, <0.001.


Table I provides a list of sequences that have significantly altered expression in hypertrophic cardiomyopathy, which genes may be induced or repressed as indicated in the table. Table IA provides a subset of upregulated genes of interest. Table IB provides a further subset of upregulated sequences of interest. The sequences of Table IA or Table IB may be further sub-divided according to their representation in Tables II, III or IV. In some embodiments, the sequences of interest have a “fold change” as set forth in Table I, of at least about 4; of a least about 5, of at least about 6, or more.


Nucleic Acids

The sequences of pressure overload associated genes find use in diagnostic and prognostic methods, for the recombinant production of the encoded polypeptide, and the like. A list of pressure overload associated genetic sequences is provided in Table I, and in the sub-tables thereof. The nucleic acids of the invention include nucleic acids having a high degree of sequence similarity or sequence identity to one of the sequences provided in Table 1, and also include homologs, particularly human homologs, examples of which are provided in Tables II, III and IV. Sequence identity can be determined by hybridization under stringent conditions, for example, at 50° C. or higher and 0.1×SSC (9 mM NaCl/0.9 mM Na citrate). Hybridization methods and conditions are well known in the art, see, e.g., U.S. Pat. No. 5,707,829. Nucleic acids that are substantially identical to the provided nucleic acid sequence, e.g. allelic variants, genetically altered versions of the gene, etc., bind to one of the sequences provided in Table I and sub-tables thereof under stringent hybridization conditions. Further specific guidance regarding the preparation of nucleic acids is provided by Fleury et al. (1997) Nature Genetics 15:269-272; Tartaglia et al., PCT Publication No. WO 96/05861; and Chen et al., PCT Publication No. WO 00/06087, each of which is incorporated herein in its entirety.


The genes listed in Table I and sub-tables thereof may be obtained using various methods well known to those skilled in the art, including but not limited to the use of appropriate probes to detect the genes within an appropriate cDNA or genomic DNA library, antibody screening of expression libraries to detect cloned DNA fragments with shared structural features, direct chemical synthesis, and amplification protocols. Libraries are preferably prepared from nerve cells. Cloning methods are described in Berger and Kimmel, Guide to Molecular Cloning Techniques, Methods in Enzymology, 152, Academic Press, Inc. San Diego, Calif.; Sambrook, et al. (1989) Molecular Cloning—A Laboratory Manual (2nd ed) Vols. 1-3, Cold Spring Harbor Laboratory, Cold Spring Harbor Press, N.Y.; and Current Protocols (1994), a joint venture between Greene Publishing Associates, Inc. and John Wiley and Sons, Inc.


The sequence obtained from clones containing partial coding sequences or non-coding sequences can be used to obtain the entire coding region by using the RACE method (Chenchik et al. (1995) CLONTECHniques (X) 1: 5-8). Oligonucleotides can be designed based on the sequence obtained from the partial clone that can amplify a reverse transcribed mRNA encoding the entire coding sequence. Alternatively, probes can be used to screen cDNA libraries prepared from an appropriate cell or cell line in which the gene is transcribed. Once the target nucleic acid is identified, it can be isolated and cloned using well-known amplification techniques. Such techniques include the polymerase chain reaction (PCR) the ligase chain reaction (LCR), Qβ-replicase amplification, the self-sustained sequence replication system (SSR) and the transcription based amplification system (TAS). Such methods include, those described, for example, in U.S. Pat. No. 4,683,202 to Mullis et al.; PCR Protocols A Guide to Methods and Applications (Innis et al. eds) Academic Press Inc. San Diego, Calif. (1990); Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86: 1173; Guatelli et al. (1990) Proc. Natl. Acad. Sci. USA 87: 1874; Lomell et al. (1989) J. Clin. Chem. 35: 1826; Landegren et al. (1988) Science 241: 1077-1080; Van Brunt (1990) Biotechnology 8: 291-294; Wu and Wallace (1989) Gene 4: 560; and Barringer et al. (1990) Gene 89: 117.


As an alternative to cloning a nucleic acid, a suitable nucleic acid can be chemically synthesized. Direct chemical synthesis methods include, for example, the phosphotriester method of Narang et al. (1979) Meth. Enzymol. 68: 90-99; the phosphodiester method of Brown et al. (1979) Meth. Enzymol. 68: 109-151; the diethylphosphoramidite method of Beaucage et al. (1981) Tetra. Left., 22: 1859-1862; and the solid support method of U.S. Pat. No. 4,458,066. Chemical synthesis produces a single stranded oligonucleotide. This can be converted into double stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template. While chemical synthesis of DNA is often limited to sequences of about 100 bases, longer sequences can be obtained by the ligation of shorter sequences. Alternatively, subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes.


The nucleic acids can be cDNAs or genomic DNAs, as well as fragments thereof. The term “cDNA” as used herein is intended to include all nucleic acids that share the arrangement of sequence elements found in native mature mRNA species, where sequence elements are exons and 3′ and 5′ non-coding regions. Normally mRNA species have contiguous exons, with the intervening introns, when present, being removed by nuclear RNA splicing, to create a continuous open reading frame encoding a polypeptide of the invention.


A genomic sequence of interest comprises the nucleic acid present between the initiation codon and the stop codon, as defined in the listed sequences, including all of the introns that are normally present in a native chromosome. It can further include the 3′ and 5′ untranslated regions found in the mature mRNA. It can further include specific transcriptional and translational regulatory sequences, such as promoters, enhancers, etc., including about 1 kb, but possibly more, of flanking genomic DNA at either the 5′ or 3′ end of the transcribed region. The genomic DNA flanking the coding region, either 3′ or 5′, or internal regulatory sequences as sometimes found in introns, contains sequences required for proper tissue, stage-specific, or disease-state specific expression, and are useful for investigating the up-regulation of expression in tumor cells.


Probes specific to the nucleic acid of the invention can be generated using the nucleic acid sequence disclosed in Table I and sub-tables thereof. The probes are preferably at least about 18 nt, 25 nt, 50 nt or more of the corresponding contiguous sequence of one of the sequences provided in Table I and sub-tables thereof, and are usually less than about 2, 1, or 0.5 kb in length. Preferably, probes are designed based on a contiguous sequence that remains unmasked following application of a masking program for masking low complexity, e.g. BLASTX. Double or single stranded fragments can be obtained from the DNA sequence by chemically synthesizing oligonucleotides in accordance with conventional methods, by restriction enzyme digestion, by PCR amplification, etc. The probes can be labeled, for example, with a radioactive, biotinylated, or fluorescent tag.


The nucleic acids of the subject invention are isolated and obtained in substantial purity, generally as other than an intact chromosome. Usually, the nucleic acids, either as DNA or RNA, will be obtained substantially free of other naturally-occurring nucleic acid sequences, generally being at least about 50%, usually at least about 90% pure and are typically “recombinant,” e.g., flanked by one or more nucleotides with which it is not normally associated on a naturally occurring chromosome.


The nucleic acids of the invention can be provided as a linear molecule or within a circular molecule, and can be provided within autonomously replicating molecules (vectors) or within molecules without replication sequences. Expression of the nucleic acids can be regulated by their own or by other regulatory sequences known in the art. The nucleic acids of the invention can be introduced into suitable host cells using a variety of techniques available in the art, such as transferrin polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated DNA transfer, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, gene gun, calcium phosphate-mediated transfection, and the like.


For use in amplification reactions, such as PCR, a pair of primers will be used. The exact composition of the primer sequences is not critical to the invention, but for most applications the primers will hybridize to the subject sequence under stringent conditions, as known in the art. It is preferable to choose a pair of primers that will generate an amplification product of at least about 50 nt, preferably at least about 100 nt. Algorithms for the selection of primer sequences are generally known, and are available in commercial software packages. Amplification primers hybridize to complementary strands of DNA, and will prime towards each other. For hybridization probes, it may be desirable to use nucleic acid analogs, in order to improve the stability and binding affinity. The term “nucleic acid” shall be understood to encompass such analogs.


Polypeptides

Polypeptides encoded by pressure overload associated genes are of interest for screening methods, as reagents to raise antibodies, as therapeutics, and the like. Such polypeptides can be produced through isolation from natural sources, recombinant methods and chemical synthesis. In addition, functionally equivalent polypeptides may find-use, where the equivalent polypeptide may be a homolog, e.g. a human homolog, may contain deletions, additions or substitutions of amino acid residues that result in a silent change, thus producing a functionally equivalent gene product. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. “Functionally equivalent”, as used herein, refers to a protein capable of exhibiting a substantially similar in vivo activity as the polypeptide encoded by an pressure overload associated gene, as provided in Table I and sub-tables thereof.


Peptide fragments find use in a variety of methods, where fragments are usually at least about 10 amino acids in length, about 20 amino acids in length, about 50 amino acids in length, or longer, up to substantially full length. Fragments of particular interest include fragments comprising an epitope, which can be used to raise specific antibodies. Soluble fragment of cell surface proteins are also of interest, e.g. truncated at transmembrane domains.


The polypeptides may be produced by recombinant DNA technology using techniques well known in the art. Methods that are well known to those skilled in the art can be used to construct expression vectors containing coding sequences and appropriate transcriptional/translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic recombination. Alternatively, RNA capable of encoding the polypeptides of interest may be chemically synthesized.


Typically, the coding sequence is placed under the control of a promoter that is functional in the desired host cell to produce relatively large quantities of the gene product. An extremely wide variety of promoters are well-known, and can be used in the expression vectors of the invention, depending on the particular application. Ordinarily, the promoter selected depends upon the cell in which the promoter is to be active. Other expression control sequences such as ribosome binding sites, transcription termination sites and the like are also optionally included. Constructs that include one or more of these control sequences are termed “expression cassettes.” Expression can be achieved in prokaryotic and eukaryotic cells utilizing promoters and other regulatory agents appropriate for the particular host cell. Exemplary host cells include, but are not limited to, E. coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines.


In mammalian host cells, a number of viral-based expression systems may be used, including retrovirus, lentivirus, adenovirus, adeno associated virus, and the like. In cases where an adenovirus is used as an expression vector, the coding sequence of interest can be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing differentially expressed or pathway gene protein in infected hosts.


Specific initiation signals may also be required for efficient translation of the genes. These signals include the ATG initiation codon and adjacent sequences. In cases where a complete gene, including its own initiation codon and adjacent sequences, is inserted into the appropriate expression vector, no additional translational control signals may be needed. However, in cases where only a portion of the gene coding sequence is inserted, exogenous translational control signals must be provided. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc.


In addition, a host cell strain may be chosen that modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells that possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERO, BHK, HeLa, COS, MDCK, 293, 3T3, W138, etc.


For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines that stably express the differentially expressed or pathway gene protein may be engineered. Rather than using expression vectors that contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements, and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines that express the target protein. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that affect the endogenous activity of the differentially expressed or pathway gene protein. A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase, hypoxanthine-guanine phosphoribosyltransferase, and adenine phosphoribosyltransferase genes. Antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate; gpt, which confers resistance to mycophenolic acid; neo, which confers resistance to the aminoglycoside G-418; and hygro, which confers resistance to hygromycin.


The polypeptide may be labeled, either directly or indirectly. Any of a variety of suitable labeling systems may be used, including but not limited to, radioisotopes such as 125I; enzyme labeling systems that generate a detectable calorimetric signal or light when exposed to substrate; and fluorescent labels. Indirect labeling involves the use of a protein, such as a labeled antibody, that specifically binds to the polypeptide of interest. Such antibodies include but are not limited to polyclonal, monoclonal, chimeric, single chain, Fab fragments and fragments produced by an Fab expression library.


Once expressed, the recombinant polypeptides can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, ion exchange and/or size exclusivity chromatography, gel electrophoresis and the like (see, generally, R. Scopes, Protein Purification, Springer—Verlag, N.Y. (1982), Deutscher, Methods in Enzymology Vol. 182: Guide to Protein Purification, Academic Press, Inc. N.Y. (1990)).


As an option to recombinant methods, polypeptides and oligopeptides can be chemically synthesized. Such methods typically include solid-state approaches, but can also utilize solution based chemistries and combinations or combinations of solid-state and solution approaches. Examples of solid-state methodologies for synthesizing proteins are described by Merrifield (1964) J. Am. Chem. Soc. 85:2149; and Houghton (1985) Proc. Natl. Acad. Sci., 82:5132. Fragments of a CARDIOPROTECTIVE protein can be synthesized and then joined together. Methods for conducting such reactions are described by Grant (1992) Synthetic Peptides: A User Guide, W.H. Freeman and Co., N.Y.; and in “Principles of Peptide Synthesis,” (Bodansky and Trost, ed.), Springer-Verlag, Inc. N.Y., (1993).


Arrays

Arrays provide a high throughput technique that can assay a large number of polynucleotides or polypeptides in a sample. In one aspect of the invention, an array is constructed comprising one or more of the pressure overload associated genes, gene products, binding members specific for the gene product, etc., as set forth in Table I and sub-tables thereof, preferably comprising at least 4 distinct genes or gene products, at least about 8, at least 10, at least about 15, at least about 25, or more of these sequences, which array may further comprise other sequences known to be up- or down-regulated in heart tissue.


This technology can be used as a tool to test for differential expression. Arrays can be created by spotting polynucleotide probes, antibodies, polypeptides, etc. onto a substrate (e.g., glass, nitrocellulose, etc.) in a two-dimensional matrix or array having bound probes. The probes can be bound to the substrate by either covalent bonds or by non-specific interactions, such as hydrophobic interactions. Techniques for constructing arrays and methods of using these arrays are described in, for example, Schena et al. (1996) Proc Natl Acad Sci USA. 93(20):10614-9; Schena et al. (1995) Science 270(5235):467-70; Shalon et al. (1996) Genome Res. 6(7):639-45, U.S. Pat. No. 5,807,522, EP 799 897; WO 97/29212; WO 97/27317; EP 785 280; WO 97/02357; U.S. Pat. No. 5,593,839; U.S. Pat. No. 5,578,832; EP 728 520; U.S. Pat. No. 5,599,695; EP 721 016; U.S. Pat. No. 5,556,752; WO 95/22058; and U.S. Pat. No. 5,631,734.


The probes utilized in the arrays can be of varying types and can include, for example, synthesized probes of relatively short length (e.g., a 20-mer or a 25-mer), cDNA (full length or fragments of gene), amplified DNA, fragments of DNA (generated by restriction enzymes, for example), reverse transcribed DNA, peptides, proteins, antibodies or fragments thereof, and the like. Arrays can be utilized in detecting differential expression levels.


Arrays can be used to, for example, examine differential expression of genes. For example, arrays can be used to detect differential expression of pressure overload associated genes, where expression is compared between a test cell and control cell. Exemplary uses of arrays are further described in, for example, Pappalarado et al. (1998) Sem. Radiation Oncol. 8:217; and Ramsay. (1998) Nature Biotechnol. 16:40. Furthermore, many variations on methods of detection using arrays are well within the skill in the art and within the scope of the present invention. For example, rather than immobilizing the probe to a solid support, the test sample can be immobilized on a solid support which is then contacted with the probe. Additional discussion regarding the use of microarrays in expression analysis can be found, for example, in Duggan, et al., Nature Genetics Supplement 21:10-14 (1999); Bowtell, Nature Genetics Supplement 21:25-32 (1999); Brown and Botstein, Nature Genetics Supplement 21:33-37 (1999); Cole et al., Nature Genetics Supplement 21:38-41 (1999); Debouck and Goodfellow, Nature Genetics Supplement 21:48-50 (1999); Bassett, Jr., et al., Nature Genetics Supplement 21:51-55 (1999); and Chakravarti, Nature Genetics Supplement 21:56-60 (1999).


For detecting expression levels, usually nucleic acids are obtained from a test sample, and either directly labeled, or reversed transcribed into labeled cDNA. Alternatively, a protein sample, e.g. a serum sample, may be used, and labeled following binding to the array. The test sample containing the nucleic acids or proteins is then contacted with the array. After allowing a period sufficient for any nucleic acid or protein present in the sample to bind to the probes, the array is typically subjected to one or more washes to remove unbound sample and to minimize nonspecific binding to the probes of the arrays. Binding of labeled sequences is detected using any of a variety of commercially available scanners and accompanying software programs.


For example, if the nucleic acids from the sample are labeled with fluorescent labels, hybridization intensity can be determined by, for example, a scanning confocal microscope in photon counting mode. Appropriate scanning devices are described by e.g., U.S. Pat. No. 5,578,832 to Trulson et al., and U.S. Pat. No. 5,631,734 to Stern et al. and are available from Affymetrix, Inc., under the GeneChip™ label. Some types of label provide a signal that can be amplified by enzymatic methods (see Broude, et al., Proc. Natl. Acad. Sci. U.S.A. 91, 3072-3076 (1994)). A variety of other labels are also suitable including, for example, radioisotopes, chromophores, magnetic particles and electron dense particles.


Those locations on the probe array that are bound to sample are detected using a reader, such as described by U.S. Pat. No. 5,143,854, WO 90/15070, and U.S. Pat. No. 5,578,832. For customized arrays, the hybridization pattern can then be analyzed to determine the presence and/or relative amounts or absolute amounts of known species in samples being analyzed as described in e.g., WO 97/10365.


Specific Binding Members

The term “specific binding member” or “binding member” as used herein refers to a member of a specific binding pair, i.e. two molecules, usually two different molecules, where one of the molecules (i.e., first specific binding member) through chemical or physical means specifically binds to the other molecule (i.e., second specific binding member). The complementary members of a specific binding pair are sometimes referred to as a ligand and receptor; or receptor and counter-receptor. For the purposes of the present invention, the two binding members may be known to associate with each other, for example where an assay is directed at detecting compounds that interfere with the association of a known binding pair. Alternatively, candidate compounds suspected of being a binding partner to a compound of interest may be used.


Specific binding pairs of interest include carbohydrates and lectins; complementary nucleotide sequences; peptide ligands and receptor; effector and receptor molecules; hormones and hormone binding protein; enzyme cofactors and enzymes; enzyme inhibitors and enzymes; lipid and lipid-binding protein; etc. The specific binding pairs may include analogs, derivatives and fragments of the original specific binding member. For example, a receptor and ligand pair may include peptide fragments, chemically synthesized peptidomimetics, labeled protein, derivatized protein, etc.


In a preferred embodiment, the specific binding member is an antibody. The term “antibody” or “antibody moiety” is intended to include any polypeptide chain-containing molecular structure with a specific shape that fits to and recognizes an epitope, where one or more non-covalent binding interactions stabilize the complex between the molecular structure and the epitope. The specific or selective fit of a given structure and its specific epitope is sometimes referred to as a “lock and key” fit. The archetypal antibody molecule is the immunoglobulin, and all types of immunoglobulins, IgG, IgM, IgA, IgE, IgD, etc., from all sources, e.g. human, rodent, rabbit, cow, sheep, pig, dog, other mammal, chicken, other avians, etc., are considered to be “antibodies.” Antibodies utilized in the present invention may be polyclonal antibodies, although monoclonal antibodies are preferred because they may be reproduced by cell culture or recombinantly, and can be modified to reduce their antigenicity.


Polyclonal antibodies can be raised by a standard protocol by injecting a production animal with an antigenic composition, formulated as described above. See, e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In one such technique, an antigen comprising an antigenic portion of the protein target is initially injected into any of a wide variety of mammals (e.g., mice, rats, rabbits, sheep or goats). When utilizing an entire protein, or a larger section of the protein, antibodies may be raised by immunizing the production animal with the protein and a suitable adjuvant (e.g., Freund's, Freund's complete, oil-in-water emulsions, etc.) When a smaller peptide is utilized, it is advantageous to conjugate the peptide with a larger molecule to make an immunostimulatory conjugate. Commonly utilized conjugate proteins that are commercially available for such use include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). In order to raise antibodies to particular epitopes, peptides derived from the full sequence may be utilized. Alternatively, in order to generate antibodies to relatively short peptide portions of the protein target, a superior immune response may be elicited if the polypeptide is joined to a carrier protein, such as ovalbumin, BSA or KLH. The peptide-conjugate is injected into the animal host, preferably according to a predetermined schedule incorporating one or more booster immunizations, and the animals are bled periodically. Polyclonal antibodies specific for the polypeptide may then be purified from such antisera by, for example, affinity chromatography using the polypeptide coupled to a suitable solid support.


Alternatively, for monoclonal antibodies, hybridomas may be formed by isolating the stimulated immune cells, such as those from the spleen of the inoculated animal. These cells are then fused to immortalized cells, such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The immortal cell line utilized is preferably selected to be deficient in enzymes necessary for the utilization of certain nutrients. Many such cell lines (such as myelomas) are known to those skilled in the art, and include, for example: thymidine kinase (TK) or hypoxanthine-guanine phosphoriboxyl transferase (HGPRT). These deficiencies allow selection for fused cells according to their ability to grow on, for example, hypoxanthine aminopterinthymidine medium (HAT).


Preferably, the immortal fusion partners utilized are derived from a line that does not secrete immunoglobulin. The resulting fused cells, or hybridomas, are cultured under conditions that allow for the survival of fused, but not unfused, cells and the resulting colonies screened for the production of the desired monoclonal antibodies. Colonies producing such antibodies are cloned, expanded, and grown so as to produce large quantities of antibody, see Kohler and Milstein, 1975 Nature 256:495 (the disclosures of which are hereby incorporated by reference).


Large quantities of monoclonal antibodies from the secreting hybridomas may then be produced by injecting the clones into the peritoneal cavity of mice and harvesting the ascites fluid therefrom. The mice, preferably primed with pristane, or some other tumor-promoter, and immunosuppressed chemically or by irradiation, may be any of various suitable strains known to those in the art. The ascites fluid is harvested from the mice and the monoclonal antibody purified therefrom, for example, by CM Sepharose column or other chromatographic means. Alternatively, the hybridomas may be cultured in vitro or as suspension cultures. Batch, continuous culture, or other suitable culture processes may be utilized. Monoclonal antibodies are then recovered from the culture medium or supernatant.


Monoclonal antibodies against the protein targets of the invention may be currently available from commercial sources. These antibodies are suitable for use in the compositions of the present invention.


In addition, the antibodies or antigen binding fragments may be produced by genetic engineering. In this technique, as with the standard hybridoma procedure, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using PCR amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors. A combinatorial library is constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones which co-express a heavy and light chain (resembling the Fab fragment or antigen binding fragment of an antibody molecule). The vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.). When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.


In addition to entire immunoglobulins (or their recombinant counterparts), immunoglobulin fragments comprising the epitope binding site (e.g., Fab′, F(ab′)2, or other fragments) are useful as antibody moieties in the present invention. Such antibody fragments may be generated from whole immunoglobulins by ficin, pepsin, papain, or other protease cleavage. “Fragment,” or minimal immunoglobulins may be designed utilizing recombinant immunoglobulin techniques. For instance “Fv” immunoglobulins for use in the present invention may be produced by linking a variable light chain region to a variable heavy chain region via a peptide linker (e.g., poly-glycine or another sequence which does not form an alpha helix or beta sheet motif).


In addition, derivatized immunoglobulins with added chemical linkers, detectable moieties, such as fluorescent dyes, enzymes, substrates, chemiluminescent moieties and the like, or specific binding moieties, such as streptavidin, avidin, or biotin, and the like may be utilized in the methods and compositions of the present invention. For convenience, the term “antibody” or “antibody moiety” will be used throughout to generally refer to molecules which specifically bind to an epitope of the protein targets, although the term will encompass all immunoglobulins, derivatives, fragments, recombinant or engineered immunoglobulins, and modified immunoglobulins, as described above.


Diagnostic and Prognostic Methods

The differential expression of pressure overload associated genes indicates that these sequences can serve as markers for diagnosis, and in prognostic evaluations to detect individuals at risk for cardiac pathologies, including atrial enlargement, ventricular hypertrophy, heart failure, etc. Prognostic methods can also be utilized to monitor an individual's health status prior to and after an episode, as well as in the assessment of the severity of the episode and the likelihood and extent of recovery.


In general, such diagnostic and prognostic methods involve detecting an altered level of expression of pressure overload associated genes or gene products in the cells or tissue of an individual or a sample therefrom, to generate an expression profile. A variety of different assays can be utilized to detect an increase in pressure overload associated gene expression, including both methods that detect gene transcript and protein levels. More specifically, the diagnostic and prognostic methods disclosed herein involve obtaining a sample from an individual and determining at least qualitatively, and preferably quantitatively, the level of a pressure overload associated genes product expression in the sample. Usually this determined value or test value is compared against some type of reference or baseline value.


The term expression profile is used broadly to include a genomic expression profile, e.g., an expression profile of mRNAs, or a proteomic expression profile, e.g., an expression profile of one or more different proteins. Profiles may be generated by any convenient means for determining differential gene expression between two samples, e.g. quantitative hybridization of mRNA, labeled mRNA, amplified mRNA, cRNA, etc., quantitative PCR, ELISA for protein quantitation, and the like.


The expression profile may be generated from a biological sample using any convenient protocol. While a variety of different manners of generating expression profiles are known, such as those employed in the field of differential gene expression analysis, one representative and convenient type of protocol for generating expression profiles is array based gene expression profile generation protocols. Following obtainment of the expression profile from the sample being assayed, the expression profile is compared with a reference or control profile to make a diagnosis regarding the susceptibility phenotype of the cell or tissue from which the sample was obtained/derived. Typically a comparison is made with a set of cells from an unaffected, normal source. Additionally, a reference or control profile may be a profile that is obtained from a cell/tissue known to be predisposed to heart failure, and therefore may be a positive reference or control profile.


In certain embodiments, the obtained expression profile is compared to a single reference/control profile to obtain information regarding the phenotype of the cell/tissue being assayed. In yet other embodiments, the obtained expression profile is compared to two or more different reference/control profiles to obtain more in depth information regarding the phenotype of the assayed cell/tissue. For example, the obtained expression profile may be compared to a positive and negative reference profile to obtain confirmed information regarding whether the cell/tissue has the phenotype of interest.


The difference values, i.e. the difference in expression in the presence and absence of radiation may be performed using any convenient methodology, where a variety of methodologies are known to those of skill in the array art, e.g., by comparing digital images of the expression profiles, by comparing databases of expression data, etc. Patents describing ways of comparing expression profiles include, but are not limited to, U.S. Pat. Nos. 6,308,170 and 6,228,575, the disclosures of which are herein incorporated by reference. Methods of comparing expression profiles are also described above. A statistical analysis step is then performed to obtain the weighted contribution of the set of predictive genes.


In one embodiment of the invention, blood samples, or samples derived from blood, e.g. plasma, serum, etc. are assayed for the presence of polypeptides encoded by pressure overload associated genes, e.g. cell surface and, of particular interest, secreted polypeptides. Such polypeptides may be detected through specific binding members. The use of antibodies for this purpose is of particular interest. Various formats find use for such assays, including antibody arrays; ELISA and RIA formats; binding of labeled antibodies in suspension/solution and detection by flow cytometry, mass spectroscopy, and the like. Detection may utilize one or a panel of specific binding members, e.g. specific for at least about 2, at least about 3, at least about 5, at least about 10 or more different gene products. A subset of genes and gene products of interest for serologic assays are provided in Table II.


In another embodiment, in vivo imaging is utilized to detect the presence of pressure overload associated gene on heart tissue. Such methods may utilize, for example, labeled antibodies or ligands specific for cell surface pressure overload associated gene products. Included for such methods are gene products differentially expressed on chambers of the heart, which can be localized by in situ binding of a labeled reagent. In these embodiments, a detectably-labeled moiety, e.g., an antibody, ligand, etc., which is specific for the polypeptide is administered to an individual (e.g., by injection), and labeled cells are located using standard imaging techniques, including, but not limited to, magnetic resonance imaging, computed tomography scanning, and the like. Detection may utilize one or a cocktail of imaging reagents e.g. imaging reagents specific for at least about 2, at least about 3, at least about 5, at least about 10 or more different gene products. A subset of genes and gene products of interest for imaging assays are provided in Table III.


In another embodiment, metabolic tests are performed, e.g. with a labeled substrate, to determine the level of enzymatic activity of a pressure overload associated gene product. Gene products of interest for such assays include enzymes whose reaction product is readily detected, e.g. in blood samples. It is shown herein, for example, that oxidative phosphorylation is markedly downregulated during atrial enlargement, and provides a marker for risk of heart failure. A subset of genes and gene products of interest for metabolic assays are provided in Table IV. Assays may be directed to one or more metabolic activities


In another embodiment, an mRNA sample from heart tissue, preferably from one or more chambers affected by pressure overload, is analyzed for the genetic signature indicating pressure overload, and diagnostic of a tendency to heart failure. Expression signatures typically utilize a panel of genetic sequences, e.g. a microarray format; multiplex amplification, etc., coupled with analysis of the results to determine if there is a statistically significant match with a disease signature.


Nucleic acids or binding members such as antibodies that are specific for polypeptides derived from the sequence of one of the sequences provided in Table I and sub-tables thereof can be used to screen patient samples for increased expression of the corresponding mRNA or protein. Samples can be obtained from a variety of sources. For example, since the methods are designed primarily to diagnosis and assess risk factors for humans, samples are typically obtained from a human subject. However, the methods can also be utilized with samples obtained from various other mammals, such as primates, e.g. apes and chimpanzees, mice, cats, rats, and other animals. Such samples are referred to as a patient sample.


Samples can be obtained from the tissues or fluids of an individual, as well as from cell cultures or tissue homogenates. For example, samples can be obtained from whole blood, heart tissue biopsy, serum, saliva, tears, urine, fecal material, sweat, buccal, skin, etc. Also included in the term are derivatives and fractions of such cells and fluids. Where cells are analyzed, the number of cells in a sample will often be at least about 102, usually at least 103 and may be about 104 or more. The cells may be dissociated, in the case of solid tissues, or tissue sections may be analyzed. Alternatively a lysate of the cells may be prepared.


Diagnostic samples are collected any time after an individual is suspected to have cardiomyopathy, atrial enlargement, ventricular hypertrophy, etc. or has exhibited symptoms that predict such pathologies. In prophylactic testing, samples can be obtained from an individual who present with risk factors that indicate a susceptibility to heart failure, which risk factors include high blood pressure, obesity, diabetes, etc. as part of a routine assessment of the individual's health status.


The various test values determined for a sample from an individual believed to suffer pressure overload, cardiac hypertrophy, diastolic dysfunction, and/or, a tendency to heart failure typically are compared against a baseline value to assess the extent of increased or decreased expression, if any. This baseline value can be any of a number of different values: In some instances, the baseline value is a value established in a trial using a healthy cell or tissue sample that is run in parallel with the test sample. Alternatively, the baseline value can be a statistical value (e.g., a mean or average) established from a population of control cells or individuals. For example, the baseline value can be a value or range that is characteristic of a control individual or control population. For instance, the baseline value can be a statistical value or range that is reflective of expression levels for the general population, or more specifically, healthy individuals not susceptible to stroke. Individuals not susceptible to stroke generally refer to those having no apparent risk factors correlated with heart failure, such as high blood pressure, high cholesterol levels, diabetes, smoking and high salt diet, for example.


Nucleic Acid Screening Methods


Some of the diagnostic and prognostic methods that involve the detection of a pressure overload associated gene transcript begin with the lysis of cells and subsequent purification of nucleic acids from other cellular material, particularly mRNA transcripts. A nucleic acid derived from an mRNA transcript refers to a nucleic acid for whose synthesis the mRNA transcript, or a subsequence thereof, has ultimately served as a template. Thus, a cDNA reverse transcribed from an mRNA, an RNA transcribed from that cDNA, a DNA amplified from the cDNA, an RNA transcribed from the amplified DNA, are all derived from the mRNA transcript and detection of such derived products is indicative of the presence and/or abundance of the original transcript in a sample. Thus, suitable samples include, but are not limited to, mRNA transcripts of pressure overload associated genes, cDNA reverse transcribed from the mRNA, cRNA transcribed from the cDNA, DNA amplified from pressure overload associated nucleic acids, and RNA transcribed from amplified DNA.


A number of methods are available for analyzing nucleic acids for the presence of a specific sequence, e.g. upregulated expression. The nucleic acid may be amplified by conventional techniques, such as the polymerase chain reaction (PCR), to provide sufficient amounts for analysis. The use of the polymerase chain reaction is described in Saiki et al. (1985) Science 239:487, and a review of techniques may be found in Sambrook, et al. Molecular Cloning: A Laboratory Manual, CSH Press 1989, pp. 14.2-14.33.


A detectable label may be included in an amplification reaction. Suitable labels include fluorochromes, e.g. fluorescein isothiocyanate (FITC), rhodamine, Texas Red, phycoerythrin, allophycocyanin, 6-carboxyfluorescein(6-FAM),2,7-dimethoxy4,5-dichloro-6-carboxyfluorescein (JOE), 6-carboxy-X-rhodamine (ROX), 6-carboxy-2,4,7,4,7-hexachlorofluorescein (HEX), 5-carboxyfluorescein (5-FAM) or N,N,N,N-tetramethyl-6-carboxyrhodamine (TAMRA), radioactive labels, e.g. 32P, 35S, 3H; etc. The label may be a two stage system, where the amplified DNA is conjugated to biotin, haptens, etc. having a high affinity binding partner, e.g. avidin, specific antibodies, etc., where the binding partner is conjugated to a detectable label. The label may be conjugated to one or both of the primers. Alternatively, the pool of nucleotides used in the amplification is labeled, so as to incorporate the label into the amplification product.


The sample nucleic acid, e.g. amplified, labeled, cloned fragment, etc. is analyzed by one of a number of methods known in the art. Probes may be hybridized to northern or dot blots, or liquid hybridization reactions performed. The nucleic acid may be sequenced by dideoxy or other methods, and the sequence of bases compared to a wild-type sequence. Single strand conformational polymorphism (SSCP) analysis, denaturing gradient gel electrophoresis (DGGE), and heteroduplex analysis in gel matrices are used to detect conformational changes created by DNA sequence variation as alterations in electrophoretic mobility. Fractionation is performed by gel or capillary electrophoresis, particularly acrylamide or agarose gels.


In situ hybridization methods are hybridization methods in which the cells are not lysed prior to hybridization. Because the method is performed in situ, it has the advantage that it is not necessary to prepare RNA from the cells. The method usually involves initially fixing test cells to a support (e.g., the walls of a microtiter well) and then permeabilizing the cells with an appropriate permeabilizing solution. A solution containing labeled probes for a pressure overload associated gene is then contacted with the cells and the probes allowed to hybridize with the nucleic acids. Excess probe is digested, washed away and the amount of hybridized probe measured. This approach is described in greater detail by Harris, D. W. (1996) Anal. Biochem. 243:249-256; Singer, et al. (1986) Biotechniques 4:230-250; Haase et al. (1984) Methods in Virology, vol. VII, pp. 189-226; and Nucleic Acid Hybridization: A Practical Approach (Hames, et al., eds., 1987).


A variety of so-called “real time amplification” methods or “real time quantitative PCR” methods can also be utilized to determine the quantity of pressure overload associated gene mRNA present in a sample. Such methods involve measuring the amount of amplification product formed during an amplification process. Fluorogenic nuclease assays are one specific example of a real time quantitation method that can be used to detect and quantitate pressure overload associated gene transcripts. In general such assays continuously measure PCR product accumulation using a dual-labeled fluorogenic oligonucleotide probe—an approach frequently referred to in the literature simply as the “TaqMan” method.


The probe used in such assays is typically a short (ca. 20-25 bases) polynucleotide that is labeled with two different fluorescent dyes. The 5′ terminus of the probe is typically attached to a reporter dye and the 3′ terminus is attached to a quenching dye, although the dyes can be attached at other locations on the probe as well. For measuring a pressure overload associated gene transcript, the probe is designed to have at least substantial sequence complementarity with a probe binding site on a pressure overload associated gene transcript. Upstream and downstream PCR primers that bind to regions that flank the pressure overload associated gene are also added to the reaction mixture.


When the probe is intact, energy transfer between the two fluorophors occurs and the quencher quenches emission from the reporter. During the extension phase of PCR, the probe is cleaved by the 5′ nuclease activity of a nucleic acid polymerase such as Taq polymerase, thereby releasing the reporter dye from the polynucleotide-quencher complex and resulting in an increase of reporter emission intensity that can be measured by an appropriate detection system.


One detector which is specifically adapted for measuring fluorescence emissions such as those created during a fluorogenic assay is the ABI 7700 manufactured by Applied Biosystems, Inc. in Foster City, Calif. Computer software provided with the instrument is capable of recording the fluorescence intensity of reporter and quencher over the course of the amplification. These recorded values can then be used to calculate the increase in normalized reporter emission intensity on a continuous basis and ultimately quantify the amount of the mRNA being amplified.


Additional details regarding the theory and operation of fluorogenic methods for making real time determinations of the concentration of amplification products are described, for example, in U.S. Pat. No. 5,210,015 to Gelfand, U.S. Pat. No. 5,538,848 to Livak, et al., and U.S. Pat. No. 5,863,736 to Haaland, as well as Heid, C. A., et al., Genome Research, 6:986-994 (1996); Gibson, U. E. M, et al., Genome Research 6:995-1001 (1996); Holland, P. M., et al., Proc. Natl. Acad. Sci. USA 88:7276-7280, (1991); and Livak, K. J., et al., PCR Methods and Applications 357-362 (1995), each of which is incorporated by reference in its entirety.


Polypeptide Screening Methods


Screening for expression of the subject sequences may be based on the functional or antigenic characteristics of the protein. Various immunoassays designed to quantitate proteins encoded by the sequences corresponding to the sequences provided in Table I and sub-tables thereof may be used in screening. Functional, or metabolic, protein assays have proven to be effective screening tools. The activity of the encoded protein in oxidative phosphorylation assays, etc., may be determined by comparison with unaffected individuals.


Detection may utilize staining of cells or histological sections, performed in accordance with conventional methods, using antibodies or other specific binding members that specifically bind to the pressure overload associated polypeptides. The antibodies or other specific binding members of interest, e.g. receptor ligands, are added to a cell sample, and incubated for a period of time sufficient to allow binding to the epitope, usually at least about 10 minutes. The antibody may be labeled with radioisotopes, enzymes, fluorescers, chemiluminescers, or other labels for direct detection. Alternatively, a second stage antibody or reagent is used to amplify the signal. Such reagents are well known in the art. For example, the primary antibody may be conjugated to biotin, with horseradish peroxidase-conjugated avidin added as a second stage reagent. Final detection uses a substrate that undergoes a color change in the presence of the peroxidase. The absence or presence of antibody binding may be determined by various methods, including flow cytometry of dissociated cells, microscopy, radiography, scintillation counting, etc.


An alternative method for diagnosis depends on the in vitro detection of binding between antibodies and the polypeptide corresponding to a sequence of Table I and sub-tables thereof in a blood sample, cell lysate, etc. Measuring the concentration of the target protein in a sample or fraction thereof may be accomplished by a variety of specific assays. A conventional sandwich type assay may be used. For example, a sandwich assay may first attach specific antibodies to an insoluble surface or support. The particular manner of binding is not crucial so long as it is compatible with the reagents and overall methods of the invention. They may be bound to the plates covalently or non-covalently, preferably non-covalently.


The insoluble supports may be any compositions to which polypeptides can be bound, which is readily separated from soluble material, and which is otherwise compatible with the overall method. The surface of such supports may be solid or porous and of any convenient shape. Examples of suitable insoluble supports to which the receptor is bound include beads, e.g. magnetic beads, membranes and microtiter plates. These are typically made of glass, plastic (e.g. polystyrene), polysaccharides, nylon or nitrocellulose. Microtiter plates are especially convenient because a large number of assays can be carried out simultaneously, using small amounts of reagents and samples.


Patient sample lysates are then added to separately assayable supports (for example, separate wells of a micromiter plate) containing antibodies. Preferably, a series of standards, containing known concentrations of the test protein is assayed in parallel with the samples or aliquots thereof to serve as controls. Preferably, each sample and standard will be added to multiple wells so that mean values can be obtained for each. The incubation time should be sufficient for binding, generally, from about 0.1 to 3 hr is sufficient. After incubation, the insoluble support is generally washed of non-bound components. Generally, a dilute non-ionic detergent medium at an appropriate pH, generally 7-8, is used as a wash medium. From one to six washes may be employed, with sufficient volume to thoroughly wash non-specifically bound proteins present in the sample.


After washing, a solution containing a second antibody is applied. The antibody will bind to one of the proteins of interest with sufficient specificity such that it can be distinguished from other components present. The second antibodies may be labeled to facilitate direct, or indirect quantification of binding. Examples of labels that permit direct measurement of second receptor binding include radiolabels, such as 3H or 125I, fluorescers, dyes, beads, chemiluminescers, colloidal particles, and the like. Examples of labels that permit indirect measurement of binding include enzymes where the substrate may provide for a colored or fluorescent product. In a preferred embodiment, the antibodies are labeled with a covalently bound enzyme capable of providing a detectable product signal after addition of suitable substrate. Examples of suitable enzymes for use in conjugates include horseradish peroxidase, alkaline phosphatase, malate dehydrogenase and the like. Where not commercially available, such antibody-enzyme conjugates are readily produced by techniques known to those skilled in the art. The incubation time should be sufficient for the labeled ligand to bind available molecules. Generally, from about 0.1 to 3 hr is sufficient, usually 1 hr sufficing.


After the second binding step, the insoluble support is again washed free of non-specifically bound material, leaving the specific complex formed between the target protein and the specific binding member. The signal produced by the bound conjugate is detected by conventional means. Where an enzyme conjugate is used, an appropriate enzyme substrate is provided so a detectable product is formed.


Other immunoassays are known in the art and may find use as diagnostics. Ouchterlony plates provide a simple determination of antibody binding. Western blots may be performed on protein gels or protein spots on filters, using a detection system specific for the pressure overload associated polypeptide as desired, conveniently using a labeling method as described for the sandwich assay.


In some cases, a competitive assay will be used. In addition to the patient sample, a competitor to the targeted protein is added to the reaction mix. The competitor and the pressure overload associated polypeptide compete for binding to the specific binding partner. Usually, the competitor molecule will be labeled and detected as previously described, where the amount of competitor binding will be proportional to the amount of target protein present. The concentration of competitor molecule will be from about 10 times the maximum anticipated protein concentration to about equal concentration in order to make the most sensitive and linear range of detection.


The detection methods can be provided as part of a kit. Thus, the invention further provides kits for detecting the presence of an mRNA corresponding to a sequence of Table I, II, or III, and/or a polypeptide encoded thereby, in a biological sample. Procedures using these kits can be performed by clinical laboratories, experimental laboratories, medical practitioners, or private individuals. The kits of the invention for detecting a polypeptide comprise a moiety that specifically binds the polypeptide, which may be a specific antibody. The kits of the invention for detecting a nucleic acid comprise a moiety that specifically hybridizes to such a nucleic acid. The kit may optionally provide additional components that are useful in the procedure, including, but not limited to, buffers, developing reagents, labels, reacting surfaces, means for detection, control samples, standards, instructions, and interpretive information.


Imaging In Vivo


In some embodiments, the methods are adapted for imaging use in vivo, e.g., to locate or identify sites where pressure overload associated genes are expressed. In these embodiments, a detectably-labeled moiety, e.g., an antibody, which is specific for the pressure overload associated polypeptide is administered to an individual (e.g., by injection), and labeled cells are located using standard imaging techniques, including, but not limited to, magnetic resonance imaging, computed tomography scanning, and the like.


For diagnostic in vivo imaging, the type of detection instrument available is a major factor in selecting a given radionuclide. The radionuclide chosen must have a type of decay that is detectable by a given type of instrument. In general, any conventional method for visualizing diagnostic imaging can be utilized in accordance with this invention. Another important factor in selecting a radionuclide for in vivo diagnosis is that its half-life be long enough that it is'still detectable at the time of maximum uptake by the target tissue, but short enough that deleterious radiation of the host is minimized. A currently used method for labeling with 99mTc is the reduction of pertechnetate ion in the presence of a chelating precursor to form the labile 99mTc-precursor complex, which, in turn, reacts with the metal binding group of a bifunctionally modified chemotactic peptide to form a 99mTc-chemotactic peptide conjugate.


The detectably labeled antibody is used in conjunction with imaging techniques, in order to analyze the expression of the target. In one embodiment, the imaging method is one of PET or SPECT, which are imaging techniques in which a radionuclide is synthetically or locally administered to a patient. The subsequent uptake of the radiotracer is measured over time and used to obtain information about the targeted tissue. Because of the high-energy (γ-ray) emissions of the specific isotopes employed and the sensitivity and sophistication of the instruments used to detect them, the two-dimensional distribution of radioactivity may be inferred from outside of the body.


Among the most commonly used positron-emitting nuclides in PET are included 11C, 13N, 15O, and 18F. Isotopes that decay by electron capture and/or y emission are used in SPECT, and include 123I and 99mTc.


Time Course Analyses


Certain prognostic methods of assessing a patient's risk of heart failure involve monitoring expression levels for a patient susceptible to heart failure, to track whether there is a change in expression of a pressure overload associated gene over time. An increase in expression over time can indicate that the individual is at increased risk for heart failure. As with other measures, the expression level for the patient at risk for heart failure is compared against a baseline value. The baseline in such analyses can be a prior value determined for the same individual or a statistical value (e.g., mean or average) determined for a control group (e.g., a population of individuals with no apparent neurological risk factors). An individual showing a statistically significant increase in pressure overload associated expression levels over time can prompt the individual's physician to take prophylactic measures to lessen the individual's potential for heart failure. For example, the physician can recommend certain life style changes (e.g., medication, improved diet, exercise program) to reduce the risk of heart failure.


Databases of Expression Profiles

Also provided are databases of expression profiles of phenotype determinative genes. Such databases will typically comprise expression profiles of various cells/tissues having susceptible phenotypes, negative expression profiles, etc., where such profiles are further described below.


The expression profiles and databases thereof may be provided in a variety of media to facilitate their use. “Media” refers to a manufacture that contains the expression profile information of the present invention. The databases of the present invention can be recorded on computer readable media, e.g. any medium that can be read and accessed directly by a computer. Such media include, but are not limited to: magnetic storage media, such as floppy discs, hard disc storage medium, and magnetic tape; optical storage media such as CD-ROM; electrical storage media such as RAM and ROM; and hybrids of these categories such as magnetic/optical storage media. One of skill in the art can readily appreciate how any of the presently known computer readable mediums can be used to create a manufacture comprising a recording of the present database information. “Recorded” refers to a process for storing information on computer readable medium, using any such methods as known in the art. Any convenient data storage structure may be chosen, based on the means used to access the stored information. A variety of data processor programs and formats can be used for storage, e.g. word processing text file, database format, etc.


As used herein, “a computer-based system” refers to the hardware means, software means, and data storage means used to analyze the information of the present invention. The minimum hardware of the computer-based systems of the present invention comprises a central processing unit (CPU), input means, output means, and data storage means. A skilled artisan can readily appreciate that any one of the currently available computer-based system are suitable for use in the present invention. The data storage means may comprise any manufacture comprising a recording of the present information as described above, or a memory access means that can access such a manufacture.


A variety of structural formats for the input and output means can be used to input and output the information in the computer-based systems of the present invention. Such presentation provides a skilled artisan with a ranking of similarities and identifies the degree of similarity contained in the test expression profile.


Therapeutic/Prophylactic Treatment Methods

Agents that modulate activity of pressure overload associated genes provide a point of therapeutic or prophylactic intervention. Numerous agents are useful in modulating this activity, including agents that directly modulate expression, e.g. expression vectors, antisense specific for the targeted gene; and agents that act on the protein, e.g. specific antibodies and analogs thereof, small organic molecules that block catalytic activity, etc.


The genes, gene fragments, or the encoded protein or protein fragments are useful in therapy to treat disorders associated with defects in expression. From a therapeutic point of view, modulating activity may have a therapeutic effect on a number of degenerative disorders. For example, expression can be upregulated by introduction of an expression vector, enhancing expression, providing molecules that mimic the activity of the targeted polypeptide, etc.


Antisense molecules can be used to down-regulate expression in cells. The antisense reagent may be antisense oligonucleotides (ODN), particularly synthetic ODN having chemical modifications from native nucleic acids, or nucleic acid constructs that express such antisense molecules as RNA. The antisense sequence is complementary to the mRNA of the targeted gene, and inhibits expression of the targeted gene products. Antisense molecules inhibit gene expression through various mechanisms, e.g. by reducing the amount of mRNA available for translation, through activation of RNAse H, or steric hindrance. One or a combination of antisense molecules may be administered, where a combination may comprise multiple different sequences.


Antisense molecules may be produced by expression of all or a part of the target gene sequence in an appropriate vector, where the transcriptional initiation is oriented such that an antisense strand is produced as an RNA molecule. Alternatively, the antisense molecule is a synthetic oligonucleotide. Antisense oligonucleotides will generally be at least about 7, usually at least about 12, more usually at least about 20 nucleotides in length, and not more than about 500, usually not more than about 50, more usually not more than about 35 nucleotides in length, where the length is governed by efficiency of inhibition, specificity, including absence of cross-reactivity, and the like.


Antisense oligonucleotides may be chemically synthesized by methods known in the art (see Wagner et al. (1993) supra. and Milligan et al., supra.) Preferred oligonucleotides are chemically modified from the native phosphodiester structure, in order to increase their intracellular stability and binding affinity. A number of such modifications have been described in the literature, which alter the chemistry of the backbone, sugars or heterocyclic bases.


In one embodiment of the invention, RNAi technology is used. As used herein, RNAi technology refers to a process in which double-stranded RNA is introduced into cells expressing a candidate gene to inhibit expression of the candidate gene, i.e., to “silence” its expression. The dsRNA is selected to have substantial identity with the candidate gene. In general such methods initially involve transcribing a nucleic acids containing all or part of a candidate gene into single- or double-stranded RNA. Sense and anti-sense RNA strands are allowed to anneal under appropriate conditions to form dsRNA. The resulting dsRNA is introduced into cells via various methods. Usually the dsRNA consists of two separate complementary RNA strands. However, in some instances, the dsRNA may be formed by a single strand of RNA that is self-complementary, such that the strand loops back upon itself to form a hairpin loop. Regardless of form, RNA duplex formation can occur inside or outside of a cell.


dsRNA can be prepared according to any of a number of methods that are known in the art, including in vitro and in vivo methods, as well as by synthetic chemistry approaches. Examples of such methods include, but are not limited to, the methods described by Sadher et al. (Biochem. Int. 14:1015, 1987); by Bhaltacharyya (Nature 343:484, 1990); and by Livache, et al. (U.S. Pat. No. 5,795,715), each of which is incorporated herein by reference in its entirety. Single-stranded RNA can also be produced using a combination of enzymatic and organic synthesis or by total organic synthesis. The use of synthetic chemical methods enable one to introduce desired modified nucleotides or nucleotide analogs into the dsRNA. dsRNA can also be prepared in vivo according to a number of established methods (see, e.g., Sambrook, et al. (1989) Molecular Cloning: A Laboratory Manual, 2nd ed.; Transcription and Translation (B. D. Hames, and S. J. Higgins, Eds., 1984); DNA Cloning, volumes I and II (D. N. Glover, Ed., 1985); and Oligonucleotide Synthesis (M. J. Gait, Ed., 1984, each of which is incorporated herein by reference in its entirety).


A number of options can be utilized to deliver the dsRNA into a cell or population of cells. For instance, RNA can be directly introduced intracellularly. Various physical methods are generally utilized in such instances, such as administration by microinjection (see, e.g., Zernicka-Goetz, et al. (1997) Development 124:1133-1137; and Wianny, et al. (1998) Chromosoma 107: 430-439). Other options for cellular delivery include permeabilizing the cell membrane and electroporation in the presence of the dsRNA, liposome-mediated transfection, or transfection using chemicals such as calcium phosphate. A number of established gene therapy techniques can also be utilized to introduce the dsRNA into a cell. By introducing a viral construct within a viral particle, for instance, one can achieve efficient introduction of an expression construct into the cell and transcription of the RNA encoded by the construct.


Compound Screening

Compound screening may be performed using an in vitro model, a genetically altered cell or animal, or purified protein corresponding to any one of the provided pressure overload associated genes. One can identify ligands or substrates that bind to, inhibit, modulate or mimic the action of the encoded polypeptide.


The polypeptides include those encoded by the provided genetic sequences, as well as nucleic acids that, by virtue of the degeneracy of the genetic code, are not identical in sequence to the disclosed nucleic acids, and variants thereof. Variant polypeptides can include amino acid (aa) substitutions, additions or deletions. The amino acid substitutions can be conservative amino acid substitutions or substitutions to eliminate non-essential amino acids, such as to alter a glycosylation site, a phosphorylation site or an acetylation site, or to minimize misfolding by substitution or deletion of one or more cysteine residues that are not necessary for function. Variants can be designed so as to retain or have enhanced biological activity of a particular region of the protein (e.g., a functional domain and/or, where the polypeptide is a member of a protein family, a region associated with a consensus sequence). Variants also include fragments of the polypeptides disclosed herein, particularly biologically active fragments and/or fragments corresponding to functional domains. Fragments of interest will typically be at least about 10 aa to at least about 15 aa in length, usually at least about 50 aa in length, and can be as long as 300 aa in length or longer, but will usually not exceed about 500 aa in length, where the fragment will have a contiguous stretch of amino acids that is identical to a polypeptide encoded by a pressure overload associated gene, or a homolog thereof.


Transgenic animals or cells derived therefrom are also used in compound screening. Transgenic animals may be made through homologous recombination, where the normal locus corresponding to a pressure overload associated gene is altered. Alternatively, a nucleic acid construct is randomly integrated into the genome. Vectors for stable integration include plasmids, retroviruses and other animal viruses, YACs, and the like. A series of small deletions and/or substitutions may be made in the coding sequence to determine the role of different domains. Of interest is the use of pressure overload associated genes to construct transgenic animal models for heart failure. Specific constructs of interest include antisense sequences that block expression of the targeted gene and expression of dominant negative mutations. A detectable marker, such as lac Z may be introduced into the locus of interest, where up-regulation of expression will result in an easily detected change in phenotype. One may also provide for expression of the target gene or variants thereof in cells or tissues where it is not normally expressed or at abnormal times of development. By providing expression of the target protein in cells in which it is not normally produced, one can induce changes in cell behavior.


Compound screening identifies agents that modulate function of the pressure overload associated gene. Of particular interest are screening assays for agents that have a low toxicity for human cells. A wide variety of assays may be used for this purpose, including labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, and the like. Knowledge of the 3-dimensional structure of the encoded protein, derived from crystallization of purified recombinant protein, could lead to the rational design of small drugs that specifically inhibit activity. These drugs may be directed at specific domains.


The term “agent” as used herein describes any molecule, e.g. protein or pharmaceutical, with the capability of altering or mimicking the physiological function of a pressure overload associated associated gene. Generally a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a differential response to the various concentrations. Typically one of these concentrations serves as a negative control, i.e. at zero concentration or below the level of detection.


Candidate agents encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 50 and less than about 2,500 daltons. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Candidate agents are also found among biomolecules including peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof.


Candidate agents are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides and oligopeptides. Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural or synthetically produced libraries and compounds are readily modified through conventional chemical, physical and biochemical means, and may be used to produce combinatorial libraries. Known pharmacological agents may be subjected to directed or random chemical modifications, such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs. Test agents can be obtained from libraries, such as natural product libraries or combinatorial libraries, for example. A number of different types of combinatorial libraries and methods for preparing such libraries have been described, including for example, PCT publications WO 93/06121, WO 95/12608, WO 95/35503, WO 94/08051 and WO 95/30642, each of which is incorporated herein by reference.


Where the screening assay is a binding assay, one or more of the molecules may be joined to a label, where the label can directly or indirectly provide a-detectable signal. Various labels include radioisotopes, fluorescers, chemiluminescers, enzymes, specific binding molecules, particles, e.g. magnetic particles, and the like. Specific binding molecules include pairs, such as biotin and streptavidin, digoxin and antidigoxin, etc. For the specific binding members, the complementary member would normally be labeled with a molecule that provides for detection, in accordance with known procedures.


A variety of other reagents may be included in the screening assay. These include reagents like salts, neutral proteins, e g. albumin, detergents, etc that are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Reagents that improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc. may be used. The mixture of components are added in any order that provides for the requisite binding. Incubations are performed at any suitable temperature, typically between 4 and 40° C. Incubation periods are selected for optimum activity, but may also be optimized to facilitate rapid high-throughput screening. Typically between 0.1 and 1 hours will be sufficient.


Preliminary screens can be conducted by screening for compounds capable of binding to a pressure overload associated gene product, as at least some of the compounds so identified are likely inhibitors. The binding assays usually involve contacting a protein with one or more test compounds and allowing sufficient time for the protein and test compounds 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. The protein utilized in such assays can be naturally expressed, cloned or synthesized.


Compounds that are initially identified by any of the foregoing screening methods can be further tested to validate the apparent activity. 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 an pressure overload associated gene is in fact differentially regulated. The animal models utilized in validation studies generally are mammals. Specific examples of suitable animals include, but are not limited to, primates, mice, and rats.


Active test agents identified by the screening methods described herein can serve as lead compounds for the synthesis of analog compounds. Typically, the analog compounds are synthesized to have an electronic configuration and a molecular conformation similar to that of the lead compound. Identification of analog compounds can be performed through use of techniques such as self-consistent field (SCF) analysis, configuration interaction (CI) analysis, and normal mode dynamics analysis. Computer programs for implementing these techniques are available. See, e.g., Rein et al., (1989) Computer-Assisted Modeling of Receptor-Ligand Interactions (Alan Liss, New York).


Once analogs have been prepared, they can be screened using the methods disclosed herein to identify those analogs that exhibit an increased ability to modulate gene product activity. Such compounds can then be subjected to further analysis to identify those compounds that appear to have the greatest potential as pharmaceutical agents. Alternatively, analogs shown to have activity through the screening methods can serve as lead compounds in the preparation of still further analogs, which can be screened by the methods described herein. The cycle of screening, synthesizing analogs and re-screening can be repeated multiple times.


Compounds identified by the screening methods described above and analogs thereof can serve as the active ingredient in pharmaceutical compositions formulated for the treatment of various disorders, including a propensity for heart failure. The compositions can also include various other agents to enhance delivery and efficacy. The compositions can also include various agents to enhance delivery and stability of the active ingredients.


Thus, for example, the compositions can also include, depending on the formulation desired, pharmaceutically-acceptable, non-toxic carriers of diluents, which are defined as vehicles commonly used to formulate pharmaceutical compositions for animal or human administration. The diluent is selected so as not to affect the biological activity of the combination. Examples of such diluents are distilled water, buffered water, physiological saline, PBS, Ringer's solution, dextrose solution, and Hank's solution. In addition, the pharmaceutical composition or formulation can include other carriers, adjuvants, or non-toxic, nontherapeutic, nonimmunogenic stabilizers, excipients and the like. The compositions can also include additional substances to approximate physiological conditions, such as pH adjusting and buffering agents, toxicity adjusting agents, wetting agents and detergents.


The composition can also include any of a variety of stabilizing agents, such as an antioxidant for example. When the pharmaceutical composition includes a polypeptide, the polypeptide can be complexed with various well-known compounds that enhance the in vivo stability of the polypeptide, or otherwise enhance its pharmacological properties (e.g., increase the half-life of the polypeptide, reduce its toxicity, enhance solubility or uptake). Examples of such modifications or complexing agents include sulfate, gluconate, citrate and phosphate. The polypeptides of a composition can also be complexed with molecules that enhance their in vivo attributes. Such molecules include, for example, carbohydrates, polyamines, amino acids, other peptides, ions (e.g., sodium, potassium, calcium, magnesium, manganese), and lipids.


Further guidance regarding formulations that are suitable for various types of administration can be found in Remington's Pharmaceutical Sciences, Mace Publishing Company, Philadelphia, Pa., 17th ed. (1985). For a brief review of methods for drug delivery, see, Langer, Science 249:1527-1533 (1990).


The pharmaceutical compositions can be administered for prophylactic and/or therapeutic treatments. Toxicity and therapeutic efficacy of the active ingredient can be determined according to standard pharmaceutical procedures in cell cultures and/or experimental animals, including, for example, determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred.


The data obtained from cell culture and/or animal studies can be used in formulating a range of dosages for humans. The dosage of the active ingredient typically lines within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage can vary within this range depending upon the dosage form employed and the route of administration utilized.


The pharmaceutical compositions described herein can be administered in a variety of different ways. Examples include administering a composition containing a pharmaceutically acceptable carrier via oral, intranasal, rectal, topical, intraperitoneal, intravenous, intramuscular, subcutaneous, subdermal, transdermal, and intrathecal methods.


Formulations suitable for parenteral administration, such as, for example, by intraarticular (in the joints), intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives.


The components used to formulate the pharmaceutical compositions are preferably of high purity and are substantially free of potentially harmful contaminants (e.g., at least National Food (NF) grade, generally at least analytical grade, and more typically at least pharmaceutical grade). Moreover, compositions intended for in vivo use are usually sterile. To the extent that a given compound must be synthesized prior to use, the resulting product is typically substantially free of any potentially toxic agents, particularly any endotoxins, which may be present during the synthesis or purification process. Compositions for parental administration are also sterile, substantially isotonic and made under GMP conditions.


Experimental

The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric.


All publications and patent applications cited in this specification are herein incorporated by reference as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference.


The present invention has been described in terms of particular embodiments found or proposed by the present inventor to comprise preferred modes for the practice of the invention. It will be appreciated by those of skill in the art that, in light of the present disclosure, numerous modifications and changes can be made in the particular embodiments exemplified without departing from the intended scope of the invention. For example, due to codon redundancy, changes can be made in the underlying DNA sequence without affecting the protein sequence. Moreover, due to biological functional equivalency considerations, changes can be made in protein structure without affecting the biological action in kind or amount. All such modifications are intended to be included within the scope of the appended claims.


The mammalian heart responds to pressure overload by undergoing left ventricular hypertrophy (LVH) and left atrial enlargement (LAE). The response to pressure overload is mediated in large part by alterations in gene transcription, and previous studies using standard molecular biological, computational, and, recently, microarray techniques have identified a number of genes involved in the pathophysiology of LVH. Many of the differentially expressed genes identified in these earlier studies are involved in cytoskeletal and matrix remodeling, myosin isoform switching (MHCα to MHCβ), TGFβ signaling, and a general reactivation of fetal gene expression patterns. Transcriptional downregulation of components of the fatty acid oxidation pathway in the hypertrophic LV has also been noted, though there has been little previous evidence of alterations in other energy metabolism pathways.


While previous studies have examined transcriptional changes in the LV, almost no attention has been paid to the changes which occur in the other heart chambers in response to pressure overload.


Transverse aortic constriction (TAC) was used to induce LVH and LAE in young adult mice, and then performed genome-wide transcriptional profiling on each of the four heart chambers from TAC and sham operated animals. Transcription of thousands of genes is significantly altered in the hypertrophic LV and enlarged LA, with an unexpectedly dramatic shift in the transcriptional profile of the TAC LA. No significant transcriptional changes are seen in the right atrium or right ventricle. Using Gene Ontology group enrichment analysis, we identified biological process groups with significant changes in group-wide expression, and found major new and unexpected changes in energy metabolism, cell cycle regulation, and signaling pathways in the LA and LV which may profoundly affect our understanding of the molecular basis of the heart's response to pressure overload.


Materials and Methods


Animal surgery, RNA preparation and hybridization. Twenty male FVB mice, age 8 weeks, underwent transverse aortic constriction performed as described by Nakamura et al. (2001) Am J Physiol Heart Circ Physiol. 281:H1104-12; and Rockman et al. (1991) Proc Natl Acad Sci USA. 1991;88:8277-81. Twenty male age matched littermates underwent the identical surgical procedure without placement of the aortic band and served as sham-operated controls.


Hearts were harvested 20 days after operation. Chambers from 15 TAC and 15 sham hearts were divided into three independent pools for RNA isolation (5 mice per pool) to obtain sufficient RNA to perform three biological replicate microarray hybridizations for each chamber. Heart harvest, chamber dissection, RNA preparation, and array hybridizations were performed as previously described in Tabibiazar et al. (2003) Circ Res.


Microarray construction. The Mouse Transcriptome Microarray used in this study was constructed in our laboratory in collaboration with the Stanford Functional Genomics Facility. Briefly, the microarray is composed of 43,200 mouse cDNA probes representing ˜25,000 unique genes and ESTs. It is composed of the National Institutes of Aging 15 k developmental gene set, the Riken 22 k gene set, and approximately 5,000 other unique clones chosen for their biological interest.


Data acquisition, processing, and statistical analysis. Image acquisition, processing, and normalization of the mouse cDNA microarray data was performed as described previously. Microarray experiments were performed using three biological replicates for each tissue and control. Features with values significantly above background in at least two out of three biological replicates were used for two-group statistical comparisons.


The Significance Analysis of Microarrays (SAM) algorithm was employed to identify genes with statistically different expression levels between TAC and sham for each of the chambers. Hierarchical clustering was performed using a set of variable genes (ANOVA, p<0.005 across all experiments) as described by Tabibiazar et al. (2003), supra. Heat maps were prepared using Heatmap Builder, Version 1. The approach to data analysis is summarized in FIG. 1.


Statistical analysis of over- and under-representation within Gene Ontology categories was performed by applying Fisher's exact test to SAM flagged genes using GoMiner analysis software.


Quantitative real-time reverse transcriptase-polymerase chain reaction. Primers and probes for 9 representative genes were obtained from Applied Biosystems' Assays-on-Demand. Quantitative rtPCR was performed as described by Tabibiazar et al. (2003), supra.


Results


Induction of cardiac hypertrophy. Hearts were harvested 20 days after operative intervention at a point when LV hypertrophy and echocardiographic indices had reached equilibrium (Nakamura et al. (2001) Am J Physiol Heart Circ Physiol. 281:H1104-12). Transverse aortic constriction induced an increase in heart weight of ˜50% (TAC 0.192±0.03 g, sham 0.133±0.007 g, p<0.03), and an increase in heart to body weight ratio of 11% (TAC 5.27+/-0.69, sham 4.72+/-0.32, p<0.03), as expected. On inspection, the left atria and left ventricles of TAC operated animals were visibly greatly enlarged, and the left ventricular wall thickness was increased.


Overview of gene expression patterns—clustering analysis. Twenty-four heart chamber mRNA samples derived from 30 individual animals were labeled and hybridized in triplicate to microarrays containing 42,300 elements, totaling over 1 million gene expression measurements. Hierarchical clustering of the data revealed a large change in the transcriptional profile of the TAC left atria, (FIG. 2) resulting in their clustering more closely with ventricles than with atria. The remainder of the atrial samples clustered as expected, with the sham LA tissues in one subgroup, and TAC and sham RA tissues in another. Left ventricles from TAC mice formed a distinct subcluster within the ventricular group, while the TAC RV and sham RV and LV cluster more closely together, suggesting there is little transcriptional change from the ventricular baseline in these tissues. These clustering results show that the most significant changes in transcription take place in the LA and LV, the two heart chambers most directly affected by increased afterload.


Differential gene expression in the left atria and left ventrcles of TAC mice. Using SAM, we identified 891 upregulated and 1001 downregulated genes in the TAC LA (false detection rate (FDR) <0.01) (FIG. 3a). A heatmap of these variable genes highlights genes whose expression in the TAC LA was similar to the ventricular pattern (FIG. 4). In the LV, SAM identified 42 upregulated and 532 downregulated genes (FDR<0.20)(FIG. 3b). Overall, the differentially regulated genes, and their direction of change in expression, are similar in the LA and LV. SAM analysis of RV and RA data demonstrated that there are no significant differences in gene expression in these tissues. T-tests identified only a small number of genes in the RA and RV with differential expression that trended toward significance.


GO functional group enrichment analysis of differentially regulated genes demonstrates coordinated regulation of biological processes. We applied Fisher's exact test to the 8773 unique GO annotated genes on the array to identify statistically significantly enriched and depleted GO groups in the TAC LA and LV. (FIG. 5). In the TAC LA, among the most significantly upregulated processes were signaling pathway activation, blood vessel development/angiogenesis, cell matrix and adhesion, and cytoskeletal organization. Downregulated processes were dominated in both the TAC LA and LV by energy pathways, including downregulation of genes involved in fatty acid oxidation, the TCA cycle, and oxidative phosphorylation. Because of the small number of upregulated genes in the TAC LV, statistical GO group analysis was not considered to be valid.


Transcriptional regulation of signaling pathways. The physiological stresses of pressure overload must be transduced into molecular signals to actuate compensatory mechanisms in cardiac cells. Deciphering which genes and pathways are involved in this transduction is of central importance, since they are some of the most interesting targets for further investigation and, potentially, drug development. In this study, we have identified many specifically regulated genes from a number of signaling pathways that have not previously been implicated in the pressure overload response.


Signaling through the transforming growth factor-β superfamily pathways is thought to modulate the cardiac response to stress, but the role of many of the downstream molecules has not been well characterized. We found significant increases in the transcription of TGF-β82, BMP2, BMP4, BMP receptor 1A, and endoglin, a component of the TGF-β receptor complex involved in angiogenesis and vessel identity. In addition, transcription of many downstream genes, including TGF-β induced transcript 1, latent transforming growth factor-β binding protein 3, activin receptor-like kinase 1, and SMADs 2, 5, 6, and 7 was significantly increased in the TAC LA, implicating them in the pressure response.


G-protein coupled receptor (GPCR) signaling pathways play a key role in the cardiac response to pressure overload. The most striking finding was the 3.6-fold downregulation of regulator of G-protein signaling 2 (RGS2) in both the LA and LV of banded mice. This gene is critically important in the regulation of blood pressure and vascular smooth muscle relaxation. Expression of the related genes RGS 3, 4, and 5 was significantly upregulated (˜2-fold) in the TAC LA but not LV. Other modifiers of GPCR signaling, the Rho small GTPases, are also specifically regulated in pressure overload. Expression of Rho A2, C, D, and G is highly significantly increased, and Rho GDP dissociation inhibitor alpha, which disrupts cardiac morphogenesis when overexpressed in the heart, is upregulated by 2.5-fold. In total, 7 of 28 annotated Rho signal transduction genes and 22 of 181 small GTPase signal transduction genes are upregulated, suggesting that this signaling pathway is integrally involved in the pressure overload response.


Transcription of several pathways involved in cell-cell signaling and physiological regulation is also dramatically impacted in pressure overload. For example, many components of angiogenic signaling pathways including VEGF A, VEGF C, VEGF-D (fos induced growth factor), neuropilin, TIE 1 tyrosine kinase receptor, angiopoietin 2, endoglin, PDGF receptor beta polypeptide, MCAM, protein O-fucosyltransferase 1, integrin alpha V, endothelial PAS domain protein 1 (HIF 2 alpha), and hypoxia inducible factor 1a are upregulated in the LA, as is chemokine receptor CXCR 4, a transcript directly induced by HIF. Altered hemodynamics in the LA also leads to regulation of a number of vasoactive peptides; transcription of endothelin receptor b was upregulated by 2-fold, while transcription of endothelin itself was downregulated 2-fold. Angiotensin converting enzyme (3,4-fold), angiotensin receptor-like 1 (Apelin receptor)(2,3-fold), adrenomedullin (2.5fold), and myotrophin (3,4-fold) were also upregulated in the LA, suggesting that the left atrium may be especially important in sensing and responding to volume conditions.


Transcriptional Regulation of Downstream Processes


Matrix and cytoskeletal remodeling. In response to the signals documented above, the pressure overloaded heart undergoes substantial tissue and cellular remodeling. Since much of this remodeling is maladaptive, and drugs which interrupt the process promote survival, (Jessup and Brozena (2003) N Engl J Med. 348:2007-18) it is important to understand which specific genes are involved. Many matrix and cell adhesion genes are highly differentially regulated, with expression differences from 5-15 fold. Expression of specific collagens is upregulated (types I, III, IV, V, VI, VIII, XV, XVI, XVIII) or downregulated (types II, IX, XI, XIV, as are specific MMPs (2 and 23 upregulated, 3, 8, 13, and 16 downregulated). One of the most highly regulated ECM genes is osteoblast specific factor 2, which has also been identified in other surveys of pressure overload. In all, more than 40 cell adhesion genes are upregulated in the TAC LA (FIG. 5).


Dynamic cytoskeletal remodeling also occurs in response to pressure overload. Transcription of a large number of actins and other cytoskeletal proteins is highly upregulated in the TAC tissues, including beta cytoplasmic actin, catenin beta, cofilin 1 (non-muscle), alpha actinin 1, coronin, dynein cytoplasmic light chain 1, thymosin beta 4 and 10, tropomodulin 3, calponin 2, destfin, drebrin, epithelial protein lost in neoplasm, vinculin, LIM and SH-3 protein 1, actin related protein complex 2/3 subunits 1B and 3, glia maturation factor beta, moesin, and the atypical, myosins Ic, Va, and X (FIG. 1a). Transcription of several actin related genes including α2 smooth muscle actin, γ-cytoplasmic actin, and four-and-a-half LIM domains 1 is also upregulated in the TAC LV. In the overabundance analyses, 30 of 298 annotated cytoskeletal and structural genes are upregulated in the TAC LA (FIG. 5). This highly specific regulation of a broad range of matrix and cytoskeletal genes demonstrates that the significant remodeling that is taking place is following a precise molecular script.


There are many points at which this maladaptive process be interrupted, such as specific inhibition of matrix metalloproteinases or potentiation of TIMPs, which can provide treatment of new aspects of the disease process.


Precisely regulated expression of cell cycle factors. Another prominent downstream target of signaling in pressure overload is the cell cycle machinery. Over 30 of 328 cell cycle genes are upregulated in the TAC LA; importantly, these genes are a clearly delineated subset of the G1 cell cycle machinery. Transcription of the early G1 cyclins D1 and D2 is elevated 2.4-to 4.7-fold in both the TAC LA and LV while there is no change in the late G1 cyclin E, necessary for entry into S-phase, or cyclin B, necessary for the G2/M phase transition. Inhibition of cyclin D expression or the downstream E2F in primary cardiomyocyte culture has been shown to prevent the development of cardiomyocyte hypertrophy. Thus, it appears that cyclin D/CDK activity without cell cycle progression promotes the hypertrophic response by facilitating increased transcription of prohypertrophic genes. Our finding that this mechanism is active in vivo in the LA and LV indicates that targeted inhibition of D-type cyclin activity provides another therapeutic approach to hypertrophy.


Altered regulation of energy metabolism. One of the most prominent and interesting targets of signaling in the pressure overloaded heart is energy metabolism. In both the LA and LV, there is a major downregulation of mitochondrial oxidative phosphorylation, the TCA cycle, and fatty acid oxidation in the TAC LA and LV. Transcription of over 40 genes associated with complexes (I-V) of the mitochondrial oxidative phosphorylation and respiratory chain machinery is dramatically downregulated, as are 7 TCA cycle genes and a large number of lipid metabolism and fatty acid oxidation pathway genes. (FIGS. 5, 6) These metabolic alterations have profound implications in a signaling feedback mechanism which may perpetuate hypertrophy.


Differential expression of hundreds of uncharacterized ESTs. A major benefit of performing microarray analyses is the ability to recognize new, uncharacterized genes which may be involved in disease processes. We have identified over 200 upregulated and 400 downregulated ESTs which respond to pressure overload. Further analysis of these novel genes can provide unique insights into the biology of the cardiac response to stress.


Quantitative realtime polymerase chain reaction confirmation of array results. Quantitative realtime polymerase chain reaction (qRT-PCR) was performed using primers for nine representative genes involved in the major processes discussed to verify that array results represent true expression differences. Each of the genes was shown to be regulated similarly in the qRT-PCR and array measurements, with the qRT-PCR data showing slightly larger measured differences in most cases (FIG. 7).


Heart failure is the leading cause of morbidity in western cultures. Commonly, the disease process begins with the development of LVH and LAE due to an increase in afterload, often as the result of systemic hypertension or aortic valve disease. We have used microarray profiling of the TAC mouse model of pressure overload to obtain a more comprehensive view of the genes and processes involved in the heart's response to increased afterload.


Previous studies of cardiac pressure overload have focused on only one heart chamber, the left ventricle, and have used significantly smaller microarrays. By using more comprehensive microarrays and improved statistical techniques to analyze transcription in the LV, we have been able identify important and previously unrecognized genes, pathways, and processes which mediate changes in the hypertrophic LV.


While the LV takes the brunt of the pressure insult, we know that during pressure overload the left atrium faces physiological challenges due to mitral regurgitation and increased wall stress which result in enlargement and remodeling. Many of the most important clinical complications of hypertrophic cardiomyopathy, valvulvar heart disease, and congestive heart failure are due to atrial enlargement, and include atrial fibrillation and other electrophysiological disturbances, as well as hemodynamic compromise caused by decreased ventricular filling. Knowing which genes and processes are associated with the atrial response may give us important clues about how to intervene in this disease process, but no studies have previously examined the transcriptional changes in the left atrium in this setting. Surprisingly, the transcriptional changes in the enlarged LA are tremendous, and much greater in scope and magnitude than the changes in the LV at this timepoint.


Similarly, no previous studies have examined whether increased pulmonary capillary wedge pressure or systemic neurohumoral changes due to left sided stresses induce transcriptional changes in the right ventricle and atrium. By examining transcription in the RA and RV, we have shown that at this point in the process, which is characterized by substantial left ventricular hypertrophy and left atrial enlargement, transcription in the RA and RV is essentially unchanged.


Our findings provide answers to a number of intriguing questions about the biology of heart failure. We know that physiological stresses such as stretch, shear, and hypoxia must be transduced into cellular signals. The data indicate that a number of different pathways are utilized in specific ways. For example, we see evidence for activation of TGFβ superfamily pathways from the extracellular space (TGFβ2, BMP2 and 4), to cell surface receptors (endoglin, BMP receptor 1a , ACVRL), to downstream transcription factors (SMADs). While the participation of TGFβ itself in the response to pressure overload has been suspected for some time, this is the first demonstration that BMPs and their receptors are involved. Mutations in the BMP pathways may be responsible for inherited cardiomyopathies, and whether targeted myocardial overexpression predisposes the heart to hypertrophy. If so, components of these BMP pathways may be tempting targets for the development of drugs aimed at interrupting the hypertrophic response.


Another unique observation from these investigations is that angiogenic signaling pathways are upregulated in the TAC LA, from extracellular VEGFs A, C and D, to receptors (Tie1, neuropilins), to transcription factors (Hif1α). This is likely the result of increased workload that leads to myocardial hypoxia followed a by robust angiogenic response.


Energy generation in the normal adult myocardium is primarily dependent on oxidative metabolism of long-chain fatty acids through the TCA cycle and mitochondrial oxidative phosphorylation, all of which we find to be dramatically transcriptionally downregulated in both the LA and LV. Though a metabolic substrate switch from fatty acids to glucose in LV hypertrophy is a well known phenomenon, there has been little previous evidence of altered expression of mitochondrial respiratory chain genes with only a few instances of decreased transcription (COX I and IV, adenine nucleotide transporter 1, F1ATPase α and β) or protein levels (ANT1, F1 ATPase α and β cytochrome c oxidase, cytochrome b5) in stressed hearts reported. We find that transcription of more than 40 genes coding for multiple components of all five complexes of the respiratory chain is dramatically downregulated in both the TAC LA and LV (FIG. 5). This concerted metabolic switch from oxygen intensive fatty acid oxidation and oxidative phosphorylation (4.1 mole ATP/1 mole O2) to glycolysis (6.3 mole ATP/1 mole O2) probably represents a response to relative hypoxia resulting from increased myocardial work and increased oxygen extraction. This response, however, leads to lower energy production in the form of ATP.


What are the potential effects of this energy deficit on the myocardium? We know that a number of mutations in disparate energy pathway genes such as the mitochondrial fatty acid importer CD36, very long chain acyl-CoA dehydrogenase, adenine nucleotide translocator-1, and mitochondrial tRNA result in inefficient ATP production and lead to hypertrophic cardiomyopathy. Another major class of inherited cardiomyopathies is due to sarcomeric protein mutations, many of which result in inefficient ATP utilization. This has led to the development of a model in which end-systolic ATP depletion prevents effective cytosolic calcium clearance by the SERCA2 pump, which is exquisitely sensitive to ATP levels. Prolonged cytosolic calcium transients then activate calcium sensitive mediators such as calcineurin, calmodulin, and CaM kinase, leading to hypertrophic stimulation.


The dramatic downregulation of oxidative phosphorylation observed herein certainly also leads to decreased ATP production in accordance with this model. The likely proximate cause for downregulation of ox-phos in the pressure overloaded and hypoxic tissues is to prevent the production of immediately toxic reactive oxygen species; unfortunately, this leads to a cycle-of hypertrophy, increased oxygen demand, ATP depletion, and further hypertrophic signaling. (FIG. 8)


The response to cardiac pressure overload requires the coordinated regulation of transcription of thousands of genes in the left atrium and left ventricle. Microarray transcription profiling and rigorous and innovative statistical techniques are used to identify the specific genes and the general biological processes which are modulated in a standard mouse model of LV hypertrophy and LA enlargement. Transcriptional patterns demonstrate significant alterations in energy metabolism, cell cycle regulation, remodeling, and signaling transduction. This study provides important insights into the pathophysiology of LVH and LAE, and identifies numerous new targets diagnosis and therapy.

TABLE I Significant Genes List - Significantly Altered Expression in Hypertrophic CardiomyopathyS0 percentile0.03False Significant Number (Median, 90 percentile)(19.57943, 55.64681)False Discovery Rate (Median, 90 percentile)(1.03485, 2.94116)Pi0Hat0.51525Gene NameGene IDScore(d)Fold Change768 Positive Significant Genes_Upregulated**CD8 antigen, beta chainBG0731404.9359527441.62458**DNA segment, Chr 1, ERATO Doi 471, expressedBG0676256.6797787652.17829**ESTs, Weakly similar to CG1_HUMAN CG1 PROTEIN [H. sapiens]BG0723355.6395965212.12391**expressed sequence AI324259AA0308955.8626702012.27914**expressed sequence AW986256AW9083124.5473792871.76174**guanine nucleotide binding protein, alpha 13BG0731655.2984555371.78085**itchyBG0740975.9587783111.78255**lymphoid blast crisis-like 1BG0633255.4819568981.83237**N-acetylated alpha-linked acidic dipeptidase 2BG06930310.260355692.13623**ribophorin 2, related sequence 1BG0657244.2799429551.63117**RIKEN cDNA 1110005E01 geneBG0729566.3204816992.65102**RIKEN cDNA 2210419I08 geneBG0726304.4432890312.74871**RIKEN cDNA 9130023P14 geneBG0738474.8989542832.03363**secreted acidic cysteine rich glycoproteinBG0650134.3057564255.37944**selected mouse cDNA on the XBG0753335.407568341.96253a disintegrin and metalloproteinase domain 15 (metargidin)AI8413536.4185645331.69879A kinase (PRKA) anchor protein 2AV0246849.3399684192.37728A20 binding inhibitor of NF-kappaB activation-2AV0519794.8336062331.36115actin related protein 2/3 complex, subunit 1B (41 kDa)AV0002465.3396448423.15358actin related protein 2/3 complex, subunit 3 (21 kDa)AV1037304.3571796621.72106actin, alpha 1, skeletal muscleAV0858824.6807155632.52776actin, alpha 2, smooth muscle, aortaAA8159934.7421462642.50123adaptor protein complex AP-1, sigma 1AV1339375.1159431931.75715adenylate cyclase 7BG0631675.8365995361.97081ADP-ribosylation factor 2AV0308604.9708111161.83182ADP-ribosylation factor 4AV1030434.8592849261.70300ADP-ribosylation-like factor 6 interacting protein 5AV0329925.2543197011.99125adrenomedullinBG06346121.135581622.44953aldehyde dehydrogenase family 1, subfamily A1BG0739395.3621745262.10401alpha actinin 4AA0002578.7322574662.60533alpha glucosidase 2, alpha neutral subunitBG0747476.5054084982.20388amyloid beta (A4) precursor proteinAV0289859.7912833592.57737amyloid beta (A4) precursor protein-binding, family B, member 2BG0749984.7029429151.59024amyloid beta (A4) precursor-like protein 2AV0702185.0991191451.98500anaphase-promoting complex subunit 5AV1624324.7603793672.04115angiopoietin 2BG1763098.3074414711.96272angiotensin converting enzymeAV0434046.7656848233.37500angiotensin receptor-like 1AV0251465.1371129842.30047ankyrin repeat hooked to zinc finger motifAV2336125.2586310252.31219annexin A3AV2183195.5801067362.46726annexin A5AV08797110.634866692.44345annexin A7AV0831206.6299515331.67612antigen identified by monoclonal antibody MRC OX-2AV0704199.0740599593.86021aquaporin 1AV0259414.6160399591.60363ATPase, Cu++ transporting, alpha polypeptideAV1737444.5462599881.99187ATPase, H+ transporting, lysosomal 34 kD, V1 subunit DAU0445668.4324529132.47791ATPase, H+ transporting, lysosomal 70 kD, V1 subunit A, isoform 1AV0315024.3003543421.50397ATP-binding cassette, sub-family G (WHITE), member 1U349204.752515492.19022basiginBG0645254.7676616511.91891Bcl-2-related ovarian killer proteinAV0864754.8640637283.01715beclin 1 (coiled-coil, myosin-like BCL2-interacting protein)AV1045355.1498919521.43711benzodiazepine receptor, peripheralAV0879216.3399808321.76235beta-2 microglobulinX018384.8188601521.51526biglycanAV1708264.230505289.77739binder of Rho GTPase 4AV0337545.4359252441.57561biregional cell adhesion molecule-related/down-regulated by oncogene AV1404586.2230503151.90841block of proliferation 1AV0551764.4628627682.03097bone morphogenetic protein 1BG0728095.0762005261.75397bone morphogenetic protein 2AV0870366.3125345381.97717bone morphogenetic protein 4AA49872426.255316225.68709bone morphogenetic protein receptor, type 1AD162504.8025500911.70860bridging integrator 3AV0410005.0211496271.50525calcium binding protein P22BG0698926.0384261912.12398calcium binding protein, intestinalAV0891055.4240736352.85345calcium channel, voltage-dependent, beta 3 subunitBG0729646.2616202082.92954calponin 2AV02519910.465797773.67100calreticulinAV1059535.7812495152.81549calumeninAV1037728.5567601912.53735capping protein alpha 1AV0011056.7597275092.71943caspase 6AV0784094.7123057581.66628catalase 1AV0062024.7894019281.58530catenin betaAA1162874.6257275473.51804cathepsin DX528866.0734588642.36142CCR4-NOT transcription complex, subunit 8AV0862274.3230851011.52705CD 81 antigenAV1718675.3452114321.62394CD24a antigenBG0760694.4898260522.69550CD34 antigenAI8932335.2423687891.99835Cd63 antigenAI8383027.5161415281.57199CD97 antigenAI3258514.6128992551.49007cell line NK14 derived transforming oncogeneAV0850727.2678965681.89454cellular retinoic acid binding protein IAV1095554.2848205486.21775chemokine (C-X-C) receptor 4D8774711.406529674.14082cholinergic receptor, nicotinic, epsilon polypeptideAV0432796.3256481182.37315citrate synthaseAV0063204.3199281461.74608CLIP associating protein 1AV0437987.8703309612.45765coagulation factor II (thrombin) receptorBG0675696.3608241213.46932coatomer protein complex, subunit gamma 1AV0312244.968232251.90246cofilin 1, non-muscleAV1707884.4185025623.52909cut-like 1 (Drosophila)AV1382334.6992082381.90631cyclin D1AA1117228.1050679064.69475cyclin D2AV1128214.8042903492.37763cyclin-dependent kinase 9 (CDC2-related kinase)BG0734234.4476157051.37304cyclin-dependent kinase inhibitor 1A (P21)AA1843684.9258945782.03325cystatin CAV1499874.5976035641.69061cytochrome P450, 2j6AV1474465.6230331931.75987damage specific DNA binding protein 1 (127 kDa)BG0635435.1594144261.74271degenerative spermatocyte homolog (Drosophila)AV0371855.9574626071.73960destrinBG0734284.3487985052.67946diaphanous homolog 1 (Drosophila)U969635.8386596071.91987diaphorase 1 (NADH)BG0670954.8990454944.08856dimethylarginine dimethylaminohydrolase 2BG0737325.1374106471.81856DNA segment, Chr 10, ERATO Doi 398, expressedBG0750706.1436263371.70405DNA segment, Chr 17, human D6S45AV1336294.2118821151.59857DNA segment, Chr 5, Bucan 26 expressedAV0696145.8649801761.33431DNA segment, Chr 6, Wayne State University 116, expressedAV0257474.177340881.78077DNA segment, Chr 6, Wayne State University 157, expressedBG0633194.7787910531.37298DNA segment, Chr 6, Wayne State University 176, expressedBG0741745.066590141.61445DNA segment, Chr 8, Brigham & Women's Genetics 1112 expressedAV08374112.394913864.11124DnaJ (Hsp40) homolog, subfamily B, member 11AV1034294.7624158791.59127dolichyl-di-phosphooligosaccharide-protein glycotransferaseBG0741385.6146407751.93040downstream of tyrosine kinase 1BG0757754.5185200783.49959drebrin 1AI8933886.852116332.36141dual adaptor for phosphotyrosine and 3-phosphoinositides 1AV0261924.4552310012.98196E26 avian leukemia oncogene 1, 5′ domainBG0650724.661684271.92560ectonucleotide pyrophosphatase/phosphodiesterase 1BG0656404.8207206242.12344elastinAV0192104.3120300379.08198ELAV (embryonic lethal, abnormal vision, Drosophila)-like 1 (Hu antigetext missing or illegible when filedAV0662116.8790631541.62078ELK3, member of ETS oncogene familyBE6244285.1076547562.38162elongation of very long chain fatty acids (FEN1/Elo2, SUR4/Elo3, yeastext missing or illegible when filedAV0505184.4184127432.30385embiginAV1403024.4843608695.19130endoglinAV0865316.4719406952.94673endothelial cell-selective adhesion moleculeAV1042135.0500520511.60966endothelial PAS domain protein 1AV0244018.2859110893.72721endothelin receptor type BAA6463226.1459207182.12895enhancer of rudimentary homolog (Drosophila)AV1096136.5537467081.82896enigma homolog (R. norvegicus)AV0328324.9442560523.43678epithelial membrane protein 1X9840313.587388415.24265epithelial protein lost in neoplasmAV1115314.5314932831.48848ESTAW55096019.855260249.11485ESTAW54758322.958663377.72500ESTAV0250404.9576879726.04194ESTAW5491664.5954407533.33061ESTAW5540826.2755688313.30960ESTS783554.6084235033.25394ESTAV1094534.8192808142.92748ESTAW5409954.4188975932.81516ESTAW5582275.7084518762.56659ESTAW5462565.044883132.47766ESTAV0870395.1667332392.46773ESTAW5443496.5847703272.44220ESTAV0399677.7239500242.43554ESTAW5364214.602875712.31306ESTAV1114658.7817512482.25221ESTAV0884108.1096310882.25135ESTAV1409016.2336437712.22461ESTAV0004467.4387183412.15361ESTAV1715844.4773964042.15320ESTBG07125511.228195322.05956ESTAW5577114.2129065272.05094ESTAW5374244.4625810952.00188ESTAV0426834.7436210751.97510ESTBG0630994.2927526011.91866ESTAV0839934.3286079761.88436ESTAV0585735.4084778711.87775ESTAV0703936.2506542381.86022ESTAV1115805.9311703641.85750ESTAW5521774.2656794711.83036ESTU201565.9930891171.81293ESTAV03634710.471398231.81269ESTAV0601654.4119553961.76104ESTAV0947064.4941659651.66259ESTAV0396384.5035347711.65226ESTAW5507054.5194307751.64943ESTAV0343327.5966717531.62595ESTW3339611.403484291.61638ESTAV0111665.1542008111.52498ESTBI0764645.4487885391.48872ESTAI8407885.9131833121.47325ESTAW5482084.1802857671.45699ESTAV3115824.5335203811.45416ESTAV1067364.2426649311.43099ESTAV0154644.4656243841.38793ESTAV0571585.3712587361.37442EST AA087124AV0879184.8839991331.86715EST, Moderately similar to A57474 extracellular matrix protein 1 precutext missing or illegible when filedAV0874997.9211722152.38462ESTsAV0244124.737821188.19962ESTsBG07346111.902786784.05199ESTsAV0337984.6725112852.61520ESTsBG0645805.6266686372.59721ESTsBG0678798.667299162.54050ESTsBG0762766.3001566682.48193ESTsBG0717398.8476367722.45591ESTsAV03240312.615140852.31331ESTsAV0784004.8370852552.27415ESTsBG0737998.2808668892.22741ESTsBG0764044.6342042512.19874ESTsAV0146074.3076536992.06730ESTsBG0737136.5611394631.99167ESTsBG0714227.4244098351.98279ESTsBI0768125.2050043141.85616ESTsAV0137225.1343252711.84817ESTsAV0117684.6423196571.81806ESTsBG0685975.1066510081.80365ESTsBG0700874.3929893251.71777ESTsAW5483604.4471217981.70141ESTsAU0401595.2024469481.64202ESTsAV0592384.7876214261.56132ESTsBG0716745.5509820711.54806ESTs, Highly similar to KIAA0356 [H. sapiens]AU0430345.5165541071.52378ESTs, Highly similar to tyrosine phosphatase [H. sapiens]AV0858164.5753619732.50854ESTs, Moderately similar to AAK1 RAT 5′-AMP-ACTIVATED PROTEINAV1096235.9114068412.27280ESTs, Moderately similar to AF188634 1 F protein [D. melanogaster]AV0833754.5686490071.95386ESTs, Moderately similar to KIAA0337 [H. sapiens]BG0746914.8253375151.56164ESTs, Moderately similar to S12207 hypothetical protein [M. musculus]AV0249816.2770676031.92645ESTs, Moderately similar to T17285 hypothetical protein DKFZp434N0text missing or illegible when filedBG0702704.1757522571.47554ESTs, Moderately similar to T46312 hypothetical protein DKFZp434J1text missing or illegible when filedBG0639815.6142339321.55378ESTs, Weakly similar to ATPase, class 1, member a; ATPase 8A2, p ttext missing or illegible when filedAV0219425.9487329022.18491ESTs, Weakly similar to DnaJ (Hsp40) homolog, subfamily B, memberAV0554604.2183018951.86141ESTs, Weakly similar to SELX_MOUSE SELENOPROTEIN X 1 (SELEtext missing or illegible when filedAA0167994.249309292.59695ESTs, Weakly similar to TUBULIN ALPHA-2 CHAIN [M. musculus]BG0696377.6975919572.61021ESTs, Weakly similar to TYROSINE-PROTEIN KINASE JAK3 [M. musctext missing or illegible when filedBG0646474.8247349131.86704ESTs, Weakly similar to Y43F4B.7.p [Caenorhabditis elegans] [C. elegtext missing or illegible when filedAV0165347.0202277112.36673ESTs, Weakly similar to ZINC FINGER PROTEIN ZFP-90 [M. musculutext missing or illegible when filedAV0100284.6019682352.80189ETL1AV0258415.6470916481.71244eukaryotic translation initiation factor 4A1BG0638794.6503365042.14899eukaryotic translation initiation factor 4EAV0947289.891112672.36476expressed sequence AA408208BG0689114.941034431.20099expressed sequence AA408225BG0641805.3742916412.50821expressed sequence AA408783AV1404754.7638022822.25681expressed sequence AA409156BG0633668.9105556812.10904expressed sequence AA414969AV0248575.4588662682.29391expressed sequence AA517451BG0688285.0238119231.49100expressed sequence AA589574AV0132174.2832262371.80346expressed sequence AA960365BG0630686.8158639121.66690expressed sequence AA986889AV0599244.2345421232.92099expressed sequence AI115505AV0257307.4618923971.96667expressed sequence AI316797BG0726594.9145874252.36058expressed sequence AI448102AV0240964.734158261.77000expressed sequence AI450948AW5548404.3726188112.43030expressed sequence AI451006BG0649995.008904082.04887expressed sequence AI452336AV0250474.3247323411.54836expressed sequence AI480459BG0727984.5422528471.93882expressed sequence AI481106AV0250424.892094322.42812expressed sequence AI504145AV0337046.2522826031.96397expressed sequence AI645998AV0588926.1531401911.71074expressed sequence AI790744BG0753634.483674781.83228expressed sequence AI836219AV0694616.4734748921.26115expressed sequence AI852829AV0099187.8945298712.08611expressed sequence AL024047AV1032904.737226551.67508expressed sequence AU022349BG0742574.175946531.59209expressed sequence AU022349AV1404714.3306679961.40070expressed sequence AU022549AV0377694.7346431122.21919expressed sequence AU024550AV0263418.6587170091.91059expressed sequence AV218468AV1622144.8459397832.30456expressed sequence AW146116AV0872204.9221118161.82565expressed sequence AW229038BG0734796.0742720865.58416expressed sequence AW547365BG0755204.7085529851.82784expressed sequence AW553532BG0745255.2083906151.92628expressed sequence C79946C799464.4430937263.00389expressed sequence C80501BG06682014.537127281.78010expressed sequence C86807BG0675805.8131080821.63424expressed sequence C87251AV0109135.4347879751.62230expressed sequence R74732BG0729845.0284484071.92281expressed sequence R74732AV0517215.1349837851.74936extracellular matrix protein 1AV0850199.8871519662.46146F-box only protein 25AV0494384.6945423331.44710fibrillin 1AA0003504.8735261083.58211fibroblast growth factor receptor 1AW4765375.2838370411.38006fibronectin 1BG0728788.3925832879.10080fibulin 2BG0732279.5348087355.40206FK506 binding protein 9AV0594456.4059507641.82419flightless I homolog (Drosophila)AV1031214.9230747192.02616follistatin-like 3BG0632944.934406512.16520frizzled-related proteinAV08965010.880583626.12984frizzled-related proteinAV08965015.649073145.14052FXYD domain-containing ion transport regulator 6AV0860025.732587123.32687G1 to phase transition 1BG0665354.9376954031.78801GA repeat binding protein, beta 1AV0410525.785172922.14048gamma-aminobutyric acid (GABA-B) receptor, 1AI8384684.5373018021.60145glia maturation factor, betaBG0664384.2879513781.91477glucose regulated protein, 58 kDaAV0739975.1383444342.95017glutathione S-transferase, mu 2BG0765048.9324826551.89118glycoprotein galactosyltransferase alpha 1, 3BG0670284.3692359792.77433glycoprotein m6bAV0333944.3915930982.33415GPI-anchored membrane protein 1AV0258624.6234710432.55428granule cell differentiation protein - MyotrophinAV0389576.0964803983.36270granulinAV0014645.8344973422.84047growth arrest and DNA-damage-inducible 45 alphaAV0350815.530172671.97603guanine nucleotide binding protein, alpha inhibiting 2BG0720925.462625112.36297guanine nucleotide binding protein, beta 1BG0634474.4680781372.09860guanosine diphosphate (GDP) dissociation inhibitor 1AV1141805.315722241.87795guanosine diphosphate (GDP) dissociation inhibitor 3AV1417294.3365249331.59962guanylate cyclase 1, soluble, beta 3AV02940412.250968252.41285H2A histone family, member YC759714.8262838051.60582hairy/enhancer-of-split related with YRPW motif-likeBG0637967.737427052.82845Harvey rat sarcoma oncogene, subgroup RAA12346610.696445021.67121heterogeneous nuclear ribonucleoprotein CAW5517786.0866513324.39239heterogeneous nuclear ribonucleoprotein KAV1115385.4204546462.03602histocompatibility 2, D region locus 1X002464.7963009971.83908histone deacetylase 1AV0236216.3994711461.72915HLS7-interacting protein kinaseBG0647337.5363866452.10383homer, neuronal immediate early gene, 3AV0418504.3336533161.39983human immunodeficiency virus type I enhancer binding protein 1AI8478325.4667294031.52844hypothetical protein MGC32441AV1037425.6970470991.61848hypothetical protein MGC7474AV0258404.4174515051.54831hypothetical protein, MGC: 6943AV0039214.3890904491.53375hypoxia inducible factor 1, alpha subunitAV06868515.091486842.53258immunoglobulin kappa chain variable 4 (V4)AV1338635.619714921.92740immunoglobulin superfamily containing leucine-rich repeatAV0848444.4893858613.04893inhibitor of DNA binding 2BG0714215.6455257342.61535inositol 1,4,5-triphosphate receptor5AI5266305.5005241881.77221insulin-like growth factor binding protein 5AV0126174.2106171151.98780insulin-like growth factor binding protein 7AV01385111.61364273.03200integral membrane protein 2BAV0104014.7611310481.49528integrin alpha 6AV0782954.481858862.35403integrin beta 1 (fibronectin receptor beta)BG0744229.1789228652.31509integrin beta 5BF1004147.0427856824.40899interferon (alpha and beta) receptor 2AV0065146.2068461711.36667interleukin 17 receptorAV0745868.8874844872.61352interleukin 6 signal transducerBG0703874.9052769933.42328kit ligandAV0315404.3597208072.07255lactate dehydrogenase 1, A chainAV0949455.6108288082.11934lamin AAV0571354.4517454881.91029laminin, gamma 1AA0597795.2851435062.71396latent transforming growth factor beta binding protein 3AV0571007.6910669712.61620lectin, galactose binding, soluble 8AV0429649.3420707281.55241leptin receptorAV0546664.2459773321.75594leukemia-associated geneAV1341665.3347526192.63905leukotriene B4 receptor 1AV1041524.9169319942.25628LIM and SH3 protein 1AV0949745.8273898712.57319LIM-domain containing, protein kinaseAV3063595.7368473231.49652low density lipoprotein receptor-related protein 1BG0753618.6287982352.60739LPS-induced TNF-alpha factorAV0513864.3489123582.73900lymphocyte antigen 6 complex, locus AAV1622704.197676612.80421lymphocyte antigen 6 complex, locus EAV0364544.268294691.80785lysyl oxidase-likeAV0949986.1689912933.19925macrophage migration inhibitory factorAV0990904.4450567691.46008MAD homolog 6 (Drosophila)AA4515015.167840273.86816manic fringe homolog (Drosophila)AV1170357.326469132.04230mannosidase 1, alphaAV02621910.738471632.23747matrilin 2AV1565344.5770388741.52149matrix metalloproteinase 2M843247.7276684892.67602matrix metalloproteinase 23BG0678075.4245313011.87576melanoma cell adhesion moleculeBG0753776.1567320113.94572membrane-bound transcription factor protease, site 1BG0729084.8106234161.93507mesenchyme homeobox 1AV30702311.159998652.72770mesothelinBG0743446.3696365181.59146metastasis associated 1-like 1AV0485894.9239775792.01067methionine aminopeptidase 2AV0582435.4619748982.45077methyl-CpG binding domain protein 1AV0292557.6619526992.16378microfibrillar associated protein 5AV1130976.3738837832.56881microtubule-associated protein 4AV0251336.0333479491.84371milk fat globule-EGF factor 8 proteinAV0944986.9516384452.53495milk fat globule-EGF factor 8 proteinAV0883584.2839897291.84505mitogen activated protein kinase 1D109394.8742685571.57936mitogen activated protein kinase 3BE1970336.3984202631.53070moesinBG0666326.707793981.86464MORF-related gene XAV0949895.6332287622.01584Mus musculus, clone IMAGE: 2647796, mRNAAV0168906.3389162121.87032Mus musculus, clone IMAGE: 2647796, mRNABG0703576.0471909141.74898Mus musculus, clone IMAGE: 2647796, mRNAAV01117510.45111731.64082Mus musculus, clone IMAGE: 3597827, mRNA, partial cdsBG0710666.3126655332.57700Mus musculus, clone IMAGE: 3597827, mRNA, partial cdsAV0902534.4079334091.70877Mus musculus, clone IMAGE: 4913219, mRNA, partial cdsAI8377644.1909990251.74159Mus musculus, clone IMAGE: 5066061, mRNA, partial cdsAV0259274.4878324071.99689Mus musculus, clone IMAGE: 5251262, mRNA, partial cdsAV0434964.8108082642.82307Mus musculus, clone MGC: 19042 IMAGE: 4188988, mRNA, complete text missing or illegible when filedAV0734894.2214234021.62803Mus musculus, clone MGC: 27672 IMAGE: 4911158, mRNA, complete text missing or illegible when filedAV0574404.8180776481.96209Mus musculus, clone MGC: 36911 IMAGE: 4945500, mRNA, complete text missing or illegible when filedBG0679724.5672566411.61513Mus musculus, clone MGC: 37634 IMAGE: 4990983, mRNA, complete text missing or illegible when filedBG0639585.1753201482.15206Mus musculus, clone MGC: 6357 IMAGE: 3493883, mRNA, complete ctext missing or illegible when filedBG0740054.3098674062.13653Mus musculus, clone MGC: 7530 IMAGE: 3492114, mRNA, complete ctext missing or illegible when filedBG0746844.7623693581.93980Mus musculus, clone MGC: 7734 IMAGE: 3498403, mRNA, complete ctext missing or illegible when filedBG0735004.3419239162.21105Mus musculus, Similar to cytoskeleton-associated protein 4, clone IMABG0737725.4513410063.42885Mus musculus, Similar to gene overexpressed in astrocytoma, clone Itext missing or illegible when filedBG0656936.477349462.38394Mus musculus, Similar to huntingtin interacting protein 1, clone MGC: 2BG0747307.3732820711.94462Mus musculus, Similar to hypothetical protein BC014916, clone MGC: 3AU0409655.6335413642.13415Mus musculus, Similar to hypothetical protein FLJ12806, clone MGC: 6AV0139634.7282900732.06908Mus musculus, Similar to hypothetical protein FLJ20244, clone MGC: 3BG0646256.8056281051.67661Mus musculus, Similar to hypothetical protein FLJ20335, clone MGC: 2AV0417954.2383851.55944Mus musculus, Similar to hypothetical protein MGC2555, clone MGC: 2AV0898165.34967144110.06282Mus musculus, Similar to hypothetical protein MGC3178, clone MGC: 2BG0656416.1638534713.84895Mus musculus, Similar to KIAA1741 protein, clone IMAGE: 5133740, mBG0665594.2771838061.72731Mus musculus, Similar to KIAA1741 protein, clone IMAGE: 5133740, mAV0740725.1880664361.54141Mus musculus, Similar to pituitary tumor-transforming 1 interacting protext missing or illegible when filedBG0666216.4398633452.07579Mus musculus, Similar to Protein P3, clone MGC: 38638 IMAGE: 53558AV1622864.4528937862.08569Mus musculus, Similar to Rho GTPase activating protein 1, clone MGCtext missing or illegible when filedAV0090028.6883946732.37995Mus musculus, Similar to xylosylprotein beta1, 4-galactosyltransferase,BG0646734.4070483661.51119myeloid-associated differentiation markerBG0726327.7854898251.99411myosin lcAW5437484.9399765441.62146myosin VaX573774.1799711642.18490myosin XBG0654534.2076724521.44525myristoylated alanine rich protein kinase C substrateBG0725848.4868134723.67023N-acetylated alpha-linked acidic dipeptidase 2BG0665635.2957227611.55776nestinBG0662284.9274944322.81873neural proliferation, differentiation and control gene 1AV0610817.403036821.97029neuroblastoma ras oncogeneBG0742194.6310122682.22671neuroblastoma, suppression of tumorigenicity 1AI32588613.276530712.60809neuropilinAV0058257.4207964984.00358nidogen 1BG0636164.8742315121.63136Niemann Pick type C2BG0728105.8717340282.05727nischarinAV0247794.6277852181.86577nitric oxide synthase 2, inducible, macrophageM926496.0981823171.74329NK2 transcription factor related, locus 5 (Drosophila)AA5305754.457797652.08311N-myc downstream regulated 3AV0023956.6651007291.93402non-POU-domain-containing, octamer binding proteinBG0640064.6216858671.97153Notch gene homolog 1, (Drosophila)BF1821584.6674601872.06267Notch gene homolog 3, (Drosophila)BF1367704.6918727972.76353novel nuclear protein 1AV0308236.4128982311.45599nuclear factor of kappa light chain gene enhancer in B-cells 1, p105AV0115397.6274799071.72959nucleobindinBG0671016.4717838362.20795O-linked N-acetylglucosamine (GlcNAc) transferase (UDP-N-acetylglutext missing or illegible when filedAV0260794.760439051.79532origin recognition complex, subunit 2 homolog (S. cerevisiae)AV0325824.7127792511.52315osteoblast specific factor 2 (fasciclin I-like)AV0848766.696001794.83838parathyroid hormone receptorAV1457184.4026416052.07806parotid secretory proteinBG0749154.3538774831.96222PDZ and LIM domain 1 (elfin)AV0937724.2604726852.39615peptidylprolyl isomerase ABG0651644.336694641.87201peptidylprolyl isomerase C-associated proteinAV0595205.4486079352.69065peripheral myelin protein, 22 kDaAV1138887.60045721.83675phosphatase and tensin homologAI8407614.4688426631.49890phosphatidylinositol glycan, class QAV0060194.3106239651.57576phosphatidylinositol transfer proteinAV0860459.1230166341.84353phosphofructokinase, liver, B-typeBG0649305.9283862142.36933phosphoglycerate mutase 1BG0648234.7379738131.87748phosphoprotein enriched in astrocytes 15BG0640354.2682304322.97109platelet derived growth factor receptor, beta polypeptideAV1129834.5531282013.77585platelet-activating factor acetylhydrolase, isoform 1b, alpha1 subunitAV0901945.2889647221.60210pleckstrin homology, Sec7 and coiled/coil domains 3AV0532705.5770331882.02770plexin B2AW5440294.4228707651.98924poly A binding protein, cytoplasmic 1AV1127244.7823711553.15594polycystic kidney disease 1 homologAV2348825.3585027172.22470polydomain proteinAI3271337.8585406073.84128procollagen C-proteinase enhancer proteinAV0845618.9957933123.95693procollagen C-proteinase enhancer proteinBG0748517.0054563023.30109procollagen, type IV, alpha 1AV0093004.7996314326.90333procollagen, type IV, alpha 2BG0747186.5569557078.64733procollagen, type XVAV0155954.2556153271.63778procollagen-proline, 2-oxoglutarate 4-dioxygenase (proline 4-hydroxylatext missing or illegible when filedAW5482584.726989982.16626programmed cell death 10AV1349454.450107461.49911proline arginine-rich end leucine-rich repeatBG0697455.2962555084.80791prolyl 4-hydroxylase, beta polypeptideBG0737504.8548481832.62046prosaposinBE3077244.2814580181.86208prostaglandin-endoperoxide synthase 2AV0256656.861888361.97886protective protein for beta-galactosidaseAV0880114.4087579051.91973protein kinase C and casein kinase substrate in neurons 2BG0741855.124878671.71964protein kinase C, deltaAA2768445.7113029042.37450protein kinase C, etaAI7878445.0599467311.93754protein kinase, cAMP dependent regulatory, type I, alphaBG0752404.7511716392.91943protein phosphatase 1, regulatory (inhibitor) subunit 14BAV0877564.956783781.55296protein tyrosine phosphatase, non-receptor type 2AA6930539.432344092.53086protein tyrosine phosphatase, receptor type, EBG0700834.6708954341.80602protein tyrosine phosphatase, receptor type, SBG0746635.1194715621.71380proteolipid protein 2AI8932124.6400451231.95153protocadherin 13BG0730004.6675313231.89233protocadherin alpha 1AV0330497.6685423321.68190PTK2 protein tyrosine kinase 2BG0651374.2021135441.69356purine-nucleoside phosphorylaseAU0425114.4504853861.59343Rab6 interacting protein 1AW5549764.296558281.83268RAB7, member RAS oncogene familyBG0742928.1904469142.03505RAD51 homolog (S. cerevisiae)AV1404834.5334218421.88562radixinAV0402474.4430389782.29201ras homolog 9 (RhoC)AV1403336.4583080621.82988ras homolog A2AA0087935.6502164521.97274ras homolog D (RhoD)AU0413578.3692737141.74085ras homolog G (RhoG)AV1042845.7542367271.75346RAS p21 protein activator 3AV0903294.5157345771.43582Ras suppressor protein 1BG0646124.2236892791.66992regulator of G-protein signaling 19 interacting protein 1AV0861285.4785963422.14051regulator of G-protein signaling 3AU0405966.4499981231.32466regulator of G-protein signaling 4AV0883799.0802814452.31400regulator of G-protein signaling 5AV0129996.012594022.00387reticulon 4AV0842198.2279190392.29694retinal short-chain dehydrogenase/reductase 1BG0733417.3344943251.84661retinoblastoma binding protein 7AW5440814.9118624413.01012retinoid-inducible serine caroboxypetidaseAV0838677.6546428121.89865retinol binding protein 1, cellularAV1401848.1944349322.71765reversion-inducing-cysteine-rich protein with kazal motifsAV0243966.2046988092.25801Rho guanine nucleotide exchange factor (GEF) 3AV0250234.8119213982.10195Rho interacting protein 3AV0745659.039902222.07373rhotekinAV1708784.9138112751.99649ribosomal protein L13aAV0299547.604343091.79277ribosomal protein L35AW5587198.6481991661.79930ribosome binding protein 1BG0636384.4223863812.03374RIKEN cDNA 0610013I17 geneAW5387667.4350567381.78394RIKEN cDNA 0610031J06 geneBG0641275.8476271561.61255RIKEN cDNA 0610039A15 geneAV1337824.2648729531.68391RIKEN cDNA 0610040B21 geneAV1401894.3913546321.62500RIKEN cDNA 0610040B21 geneBG0738894.7688515181.58153RIKEN cDNA 0610041E09 geneAV0175825.4841905231.75496RIKEN cDNA 0710001O03 geneAV0327345.0073780392.30051RIKEN cDNA 1100001D10 geneBG0645655.819064331.83095RIKEN cDNA 1110003M08 geneAV0072764.8432929952.03155RIKEN cDNA 1110006G06 geneAV0563874.2435064731.74607RIKEN cDNA 1110007A10 geneBG0636825.6125595722.02026RIKEN cDNA 1110007A14 geneAV0585249.4246894621.84586RIKEN cDNA 1110007F23 geneAV08335225.740860999.37273RIKEN cDNA 1110007F23 geneBG07457310.539622378.20649RIKEN cDNA 1110020C13 geneAV0714249.6576209021.67480RIKEN cDNA 1110020C13 geneBG0679624.5515735981.64600RIKEN cDNA 1110059L23 geneAV1337065.930343921.95157RIKEN cDNA 1110067B02 geneAV0167654.5686608851.62828RIKEN cDNA 1110070A02 geneAV0485564.5450634282.14508RIKEN cDNA 1190017B18 geneAV0203464.2031684521.41632RIKEN cDNA 1200002H13 geneAV0917074.5728212081.60106RIKEN cDNA 1200003O06 geneAV0865204.3567323742.11517RIKEN cDNA 1200013F24 geneBG0642854.9638570291.46712RIKEN cDNA 1200015A22 geneAV0880975.4862131831.89786RIKEN cDNA 1200015E15 geneBG0733185.4150483112.58596RIKEN cDNA 1200015E15 geneAV0816636.7475033442.47340RIKEN cDNA 1200015E15 geneAV1339987.3019864862.26073RIKEN cDNA 1200015G06 geneBG0759835.6379313951.36193RIKEN cDNA 1300012G16 geneBG0741424.6673581991.78865RIKEN cDNA 1300013C10 geneAV0253696.1208946012.76926RIKEN cDNA 1300018J16 geneAI8385684.8284164663.43289RIKEN cDNA 1500019E20 geneBG0752904.5709073791.56867RIKEN cDNA 1600013L13 geneAV0840404.9563925521.78135RIKEN cDNA 1600019O04 geneAV0365916.6747974851.66154RIKEN cDNA 1600025D17 geneAV0936685.1070665571.47692RIKEN cDNA 1810004P07 geneAV0603195.0371441152.13161RIKEN cDNA 1810009F10 geneAV0601945.7654965464.45887RIKEN cDNA 1810013K23 geneAV1414994.9979258211.60819RIKEN cDNA 1810048P08 geneAV1035105.5259459882.01813RIKEN cDNA 1810049K24 geneAV0582504.2039744922.26156RIKEN cDNA 1810061M12 geneAV0601805.1351662581.83261RIKEN cDNA 1810073N04 geneBG0751304.7478374212.97518RIKEN cDNA 2010012O16 geneAV0659624.195709012.00840RIKEN cDNA 2010209O12 geneBG0675254.8732731831.71182RIKEN cDNA 2210404D11 geneBG0752424.3950093471.71187RIKEN cDNA 2210412K09 geneAV0874104.1785206261.36176RIKEN cDNA 2210417O06 geneBG0637004.9025428541.82425RIKEN cDNA 2300002L21 geneAV0880225.0288589181.63333RIKEN cDNA 2310003C10 geneAV0835284.2033097991.68513RIKEN cDNA 2310003C10 geneAV0854184.2710311251.54570RIKEN cDNA 2310008D10 geneAV0863277.0295771342.03788RIKEN cDNA 2310008M10 geneAV0845536.2275597291.57439RIKEN cDNA 2310010I22 geneAV0860496.0789433461.64346RIKEN cDNA 2310010I22 geneBG0757214.2680186581.53406RIKEN cDNA 2310028N02 geneAV0871815.0217759511.85309RIKEN cDNA 2310047O13 geneAV0564954.769900361.63158RIKEN cDNA 2310058J06 geneBG0713346.6845672022.01084RIKEN cDNA 2410001H17 geneAV0851044.6015655961.72648RIKEN cDNA 2410004M09 geneAV0853874.7214143491.72715RIKEN cDNA 2410006F12 geneAV1401165.9177431281.71626RIKEN cDNA 2410008K03 geneAV1037914.433800251.43239RIKEN cDNA 2410043F08 geneBG0636198.4451390442.28280RIKEN cDNA 2410043F08 geneAV1127359.0859752151.93280RIKEN cDNA 2500002L14 geneAV1033485.5940341541.57808RIKEN cDNA 2500002L14 geneBG0715044.4433761611.40983RIKEN cDNA 2510025F08 geneAV1338384.6835647781.90121RIKEN cDNA 2510049I19 geneAV0655384.4587397411.25154RIKEN cDNA 2600001C03 geneAV1092576.6001918431.75703RIKEN cDNA 2600015J22 geneAI8478834.5091261032.02467RIKEN cDNA 2610001A11 geneAV1113204.2315682492.73739RIKEN cDNA 2610001E17 geneBG0741585.4799869021.93419RIKEN cDNA 2610002H11 geneBG0673324.2388356214.00913RIKEN cDNA 2610002H11 geneAV1115264.4892915613.74398RIKEN cDNA 2610007A16 geneBG0633735.3502419391.76553RIKEN cDNA 2610007K22 geneBG0639034.5374433231.74250RIKEN cDNA 2610009E16 geneBG0706144.4597549311.78302RIKEN cDNA 2610027H02 geneBG0730644.8553514961.90289RIKEN cDNA 2610040E16 geneAV0946304.2156933031.44224RIKEN cDNA 2610042L04 geneAV1340217.5692495962.12844RIKEN cDNA 2610209F03 geneAV0400104.8078608461.52011RIKEN cDNA 2610301D06 geneAV0949214.5995290291.48585RIKEN cDNA 2610301D06 geneBG0727794.1936651791.27258RIKEN cDNA 2610306D21 geneBG0673974.202663681.41549RIKEN cDNA 2610528A15 geneBG0735209.8826010011.87944RIKEN cDNA 2700083B06 geneAV0506825.3413266241.42328RIKEN cDNA 2810002E22 geneAV1337555.0137795452.42777RIKEN cDNA 2810404D13 geneAV1349535.0742033891.71177RIKEN cDNA 2810417D08 geneAV1417034.8501269491.89762RIKEN cDNA 2810482I07 geneAV0249735.1797443061.54763RIKEN cDNA 3110023E09 geneAV0539554.549990421.87698RIKEN cDNA 3110079L04 geneAV1401928.1786776071.66774RIKEN cDNA 3230402E02 geneAV1404389.698222291.91583RIKEN cDNA 4432404K01 geneAV0254216.8844705492.73483RIKEN cDNA 4833439O17 geneBG0755824.7505543651.76219RIKEN cDNA 4921531N22 geneAV0523796.9303397731.83146RIKEN cDNA 4921531N22 geneAV0604785.1991229271.77508RIKEN cDNA 4930415K17 geneAV0325995.2401943871.73203RIKEN cDNA 5031406P05 geneAV0612766.4116751281.56308RIKEN cDNA 5033421K01 geneBG0707134.7821364511.43323RIKEN cDNA 5133400A03 geneBG0705514.3532828771.71061RIKEN cDNA 5430400P17 geneAA0600866.0446442271.82388RIKEN cDNA 5730403E06 geneAV0205514.3476324961.84263RIKEN cDNA 5730414C17 geneAV0167434.3691818422.10883RIKEN cDNA 5730461F13 geneBG0754366.3519811251.92385RIKEN cDNA 5730518J08 geneAV0563504.2496857481.61971RIKEN cDNA 5730591C18 geneAV0859424.8676120341.87048RIKEN cDNA 6030455P07 geneBG0762435.9791460532.90914RIKEN cDNA 6330414G21 geneBG0765054.8139301932.19023RIKEN cDNA 6720474K14 geneAV0859664.8225925982.07363RIKEN cDNA 9130005N14 geneAV0606654.2523583292.54257RIKEN cDNA B430104H02 geneAV0002139.1386944632.32483RIKEN cDNA C330007P06 geneAV0294195.7221928261.77950ring finger protein 13AV0724795.9891103491.56109RNA polymerase II 1AV0183434.4897079811.82930roundabout homolog 1 (Drosophila)AV1283285.5245116391.85130roundabout homolog 4 (Drosophilia)BE3777234.9819174212.15467RuvB-Iike protein 2AV1093404.24469861.65863S-adenosylmethionine decarboxylase 1AV1219395.7076038491.64498sarcoglycan, epsilonBG0728504.3707507461.50031scavenger receptor class B1U377994.503589522.46176secreted acidic cysteine rich glycoproteinAW9887415.5492928926.14126secreted frizzled-related sequence protein 2AV0217124.2384241773.26213sema domain, immunoglobulin domain (Ig), short basic domain, secretBG0743825.0283184712.13790septin 2AV1168327.2123024842.33584serine (or cysteine) proteinase inhibitor, clade F (alpha-2 antiplasmin, text missing or illegible when filedBG0746978.8566835333.35898serine (or cysteine) proteinase inhibitor, clade H (heat shock protein 47text missing or illegible when filedAV1045224.2587402415.50558serine (or cysteine) proteinase inhibitor, clade I (neuroserpin), memberAV0520909.7902290282.31567serine palmitoyltransferase, long chain base subunit 1AV0624629.240350251.73956serine protease inhibitor 6AV0357854.3080109441.41468serum/glucocorticoid regulated kinaseAI3155894.3592686232.04271serum-inducible kinaseAV0569428.6884481073.20116SH3 domain protein D19BG0763184.832865731.72859shroomBG0728344.4600512792.66437sialyltransferase 1 (beta-galactoside alpha-2,6-sialyltransferase)D161066.3920863961.92378sialyltransferase 4C (beta-galactosidase alpha-2,3-sialytransferase)AI3856506.6103583531.97374signal transducer and activator of transcription 6L476506.3159081471.91050signal transducing adaptor molecule (SH3 domain and ITAM motif) 2AV0468594.3271581681.76305signal-induced proliferation associated gene 1AV0884794.5504089612.31046small GTPase, homolog (S. cerevisiae)BG0673564.5865038571.50828solute carrier family 29 (nucleoside transporters), member 1BG0757394.3376486071.39981sorting nexin 4AV0557224.4735357941.46762sprouty homolog 4 (Drosophila)AA4994326.4382401382.13976SRY-box containing gene 18AA2612405.1110049321.78753stanniocalcin 2AV0944164.4057140111.46040stromal cell derived factor 1BG0735934.247230612.11053stromal cell derived factor 4AV0487804.8020356071.43164superoxide dismutase 3, extracellularU382617.2502319723.29160suppressor of white apricot homolog 2-pendingAV1621954.9943556971.70716surfeit gene 4AV0745054.8155698011.79779survival motor neuronAV1339876.5397975821.39888SWI/SNF related, matrix associated, actin dependent regulator of chrotext missing or illegible when filedAV2985694.3553701182.60646syndecan 3BG0642656.6135303182.88308synovial sarcoma translocation, Chromosome 18AV0333105.4088084581.80124syntaxin binding protein 2BG0757535.0042339581.65309TAR (HIV) RNA binding protein 2AV0408476.4230862552.01946thymic stromal-derived lymphopoietin, receptorAV0708058.5470828062.02117torsin family 3, member AAV0578277.4778878672.27552transcription factor 4AV0001628.3459578912.23130transcription factor Dp 1AV0530814.3294994651.34063transcription factor E2aAA0308856.5253074061.75147transcription factor UBFAV0953174.8952256791.62658transforming growth factor beta 1 induced transcript 1AV0064799.7581349352.79512transforming growth factor, beta 2AV1358945.1735850052.73350transient receptor protein 2AV0025975.3334473662.68369transmembrane domain protein regulated in adipocytes 40 kDaAV0839475.0886653021.28986transmembrane protein with EGF-like and two follistatin-like domains 1text missing or illegible when filedAA0234935.2068121361.93718tropomodulin 3AV0264095.074818451.77695tubby like protein 4AW5526944.5306300761.78186tubby-like protein 3AV1396485.6163403121.85776tubulin, alpha 1AV0936326.1935758863.07888tubulin, alpha 4AA4087257.1555366992.13397tubulin, beta 5AV10961411.65738261.99179tumor necrosis factorX026116.4289306941.53428tumor necrosis factor receptor superfamily, member 1aL263496.3924311792.39873tumor necrosis factor, alpha-induced protein 1 (endothelial)AV0245704.3702954611.75306tumor-associated calcium signal transducer 1AV0898356.7910925173.32950tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation prtext missing or illegible when filedAV1042666.1002876291.55178tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation prtext missing or illegible when filedU573116.5739288531.87425tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation prtext missing or illegible when filedAV1304518.3508389322.79631tyrosine kinase receptor 1AA8389966.0502551883.70273U1 small nuclear ribonucleoprotein 70 kDa polypeptide AAV0354035.2183651941.76839ubiquitin carboxy-terminal hydrolase L1BG0740094.7580722342.59745UDP-GlcNAc:betaGal beta-1,3-N-acetylglucosaminyltransferase 1BG0629944.7841750931.63427UDP-glucuronate decarboxylase 1BG0736974.6518570391.53280UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosamAI8931814.619606551.98472UDP-N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosamBG0711005.2513305782.12686Unsequenced EST4131076.2732916557.53126Unsequenced EST4132734.318071475.78325Unsequenced EST41239418.329987634.03427Unsequenced EST4114674.3578342253.38896Unsequenced EST4117554.9518499413.34666Unsequenced EST4127454.5685019363.27897Unsequenced EST4321514.7747386022.87892Unsequenced EST4326034.3331426232.85312Unsequenced EST4310066.5627122842.77119Unsequenced EST4113509.5059711572.72549Unsequenced EST4116094.713549522.66098Unsequenced EST4122465.6339664392.61787Unsequenced EST4115055.9011912932.55842Unsequenced EST4320105.5575445122.54505Unsequenced EST4109934.9397338612.50496Unsequenced EST4127014.2090835292.47011Unsequenced EST4118856.1868817292.40448Unsequenced EST4120214.9028119742.39953Unsequenced EST4107614.9246404472.39667Unsequenced EST4316515.2378760412.38955Unsequenced EST1994505.7806256752.37856Unsequenced EST4125884.7950049182.37853Unsequenced EST4119238.3969406532.33231Unsequenced EST4108404.4578495852.31171Unsequenced EST4307325.5978871322.30696Unsequenced EST4126754.8150149542.22233Unsequenced EST4109685.1538446672.19677Unsequenced EST4125945.8240246832.19605Unsequenced EST4107465.9736937512.18081Unsequenced EST4318888.6084871662.15587Unsequenced EST4319205.6822013442.12745Unsequenced EST4107434.4397384152.12029Unsequenced EST1971048.3831058662.09296Unsequenced EST4309194.7942147492.08514Unsequenced EST4317066.3041177432.08389Unsequenced EST4106548.3519530222.05228Unsequenced EST2069565.2377841012.04248Unsequenced EST1933064.9455156692.02954Unsequenced EST4310725.6846025652.00932Unsequenced EST4130096.6148546171.99915Unsequenced EST4114124.8680300261.99180Unsequenced EST4310506.6994117151.98252Unsequenced EST41061912.577064051.97239Unsequenced EST4110134.9604711911.96703Unsequenced EST4116356.1187631051.95047Unsequenced EST4317675.5210765311.94831Unsequenced EST4114645.027327441.94358Unsequenced EST4105456.371479161.89709Unsequenced EST4113295.2942068791.88701Unsequenced EST4119694.924257491.86985Unsequenced EST4112854.35703541.86488Unsequenced EST4323267.9668937381.84998Unsequenced EST4124474.2604731961.83558Unsequenced EST4310824.9376321661.82592Unsequenced EST4315406.4283369191.82275Unsequenced EST1965525.7931220781.81776Unsequenced EST4107894.5502755421.81343Unsequenced EST4128034.1765852061.80861Unsequenced EST4115614.6059001031.80665Unsequenced EST4130424.6761826481.78983Unsequenced EST4122205.1676733031.78385Unsequenced EST2079145.1733033611.76367Unsequenced EST4129584.8712330651.72164Unsequenced EST4107735.1077334231.71129Unsequenced EST4320244.4327351421.70615Unsequenced EST4120114.7423937591.69693Unsequenced EST4114724.4904876261.69603Unsequenced EST4117654.5565595151.69434Unsequenced EST4123374.7701087211.69362Unsequenced EST4106984.3406164921.69179Unsequenced EST4135914.590163151.68542Unsequenced EST4123134.4908100171.67931Unsequenced EST4109206.6212272611.66619Unsequenced EST4126126.3541303711.65767Unsequenced EST4130969.6495324091.65344Unsequenced EST4113095.8556581631.65342Unsequenced EST4319824.4285550851.63322Unsequenced EST4112224.5243971031.63149Unsequenced EST4122104.3570356561.60479Unsequenced EST4135826.1724753521.59892Unsequenced EST4131815.2478393381.59329Unsequenced EST4322735.2849281811.57465Unsequenced EST4112294.6060223571.55993Unsequenced EST4328896.860445121.54569Unsequenced EST4112404.9313890881.54312Unsequenced EST4112564.3706218351.53806Unsequenced EST4311975.5535582021.51658Unsequenced EST4113844.2265029781.51562Unsequenced EST43306411.815172121.44531Unsequenced EST4115764.5571994971.41029Unsequenced EST4306834.3957447111.40057Unsequenced EST2072095.4622933971.39444Unsequenced EST4132866.1468958591.38486Unsequenced EST4119044.6539021771.37670Unsequenced EST3338704.9732077011.33528Unsequenced EST4131724.5876548571.20891uridine phosphorylaseD444644.4074207843.33647valosin containing proteinBG0743074.5825293171.50710vanilloid receptor-like protein 1BG0645105.545982921.95257vascular endothelial growth factor AAW9131888.8325649992.38847vascular endothelial growth factor CBE3769686.237015221.95868vasodilator-stimulated phosphoproteinAW5388715.1717912681.99901vinculinAI3857124.2034578511.61965v-rel reticuloendotheliosis viral oncogene homolog A, (avian)AV0952044.4436518961.71953WD repeat domain 1BG0648395.0535852282.13577zinc finger protein 103AV2247475.2364480711.82055zinc finger protein 106AV0719155.0828271542.05709zinc finger protein 36AV1031954.4441076552.24632zyxinAV1660886.2730238841.64875896 Negative Significant Genes - Repressed in Hypertrophic Cardiomyopathy**DNA segment, Chr 13, ERATO Doi 332, expressedBG066890−5.3960620550.45499**DNA segment, Chr 2, ERATO Doi 542, expressedBG073740−6.9954984830.57935**DNA segment, Chr 2, Wayne State University 85, expressedBG062980−4.1367513310.61115**DNA segment, Chr 8, Brigham & Women's Genetics 1112 expressedtext missing or illegible when filedBG064137−4.1747140820.64681**ESTsBG074866−5.8132634090.54492**guanine nucleotide binding protein, alpha 13BG068913−5.7452503430.64597**methionine aminopeptidase 2BG074258−5.8801704540.70541**Mus musculus, clone IMAGE: 5361283, mRNA, partial cdsAA072842−4.131612740.58861**proteasome (prosome, macropain) 26S subunit, ATPase 3AA163174−5.0404965670.46827**RIKEN cDNA 2310075M17 geneAI840674−5.8234261430.68802**RIKEN cDNA 3110052N05 geneBG072585−4.2036530880.68898**RIKEN cDNA 3930401B19 geneBG076041−4.2219662320.69199**RIKEN cDNA 6720463E02 geneBG067712−5.5273622470.42232**RIKEN cDNA 6720475J19 geneBG071484−7.6746854750.26086**RNA polymerase II 4 (14 kDa subunit)BG073536−4.4079899350.64966**small nuclear ribonucleoprotein NAI841348−4.562478460.50950**succinate-Coenzyme A ligase, GDP-forming, beta subunitBG075548−4.4440811730.49038**suppressor of initiator codon mutations, related sequence 1 (S. ceretext missing or illegible when filedBG064153−5.4348024110.46790**ubiquinol-cytochrome c reductase core protein 1AI841290−4.5543384090.519116-pyruvoyl-tetrahydropterin synthaseBG072031−4.9029290920.56213acetyl-Coenzyme A dehydrogenase, long-chainBG066557−9.0909096760.40106acetyl-Coenzyme A dehydrogenase, medium chainAI840666−8.3984906970.43686acyl-Coenzyme A dehydrogenase, very long chainAI839605−6.187629280.59203acylphosphatase 2, muscle typeAA120674−7.6579832390.33130adaptor-related protein complex AP-4, sigma 1BG069322−4.1389287160.48502adenylate cyclase 6AA727732−5.8707400660.47590ADP-ribosylation-like 3AV134034−4.982472190.45712ADP-ribosylation-like 4AA003086−4.4520969780.45981adrenergic receptor kinase, beta 1BG072616−5.9513118240.60538aldo-keto reductase family 1, member B3 (aldose reductase)AV133992−5.0293525660.74821aminolevulinate, delta-, dehydrataseBG063937−4.2459917220.51637amino-terminal enhancer of splitAA968065−4.9428478250.72701angiopoietinBF538875−4.8817300930.32339apoptotic chromatin condensation inducer in the nucleusBG071714−4.626237290.47419ATP synthase, H+ transporting mitochondrial F1 complex, beta subunittext missing or illegible when filedAV006369−4.6955307880.53925ATP synthase, H+ transporting, mitochondrial F0 complex, subunit b, istext missing or illegible when filedAI836064−6.4231439970.45158ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (stext missing or illegible when filedAV095153−7.4302155620.48878ATP synthase, H+ transporting, mitochondrial F0 complex, subunit c (stext missing or illegible when filedAV056821−4.4241026150.52819ATP synthase, H+ transporting, mitochondrial F0 complex, subunit f, istext missing or illegible when filedBG073062−4.4920011190.50909ATP synthase, H+ transporting, mitochondrial F0 complex, subunit gBG069449−6.6848656380.39574ATP synthase, H+ transporting, mitochondrial F1 complex, gamma poltext missing or illegible when filedBG072870−5.3478830740.52850ATP synthase, H+ transporting, mitochondrial F1 complex, O subunitAV133927−5.3526982530.47237ATP synthase, H+ transporting, mitochondrial F1F0 complex, subunit text missing or illegible when filedBG072635−4.8196183540.41437ATPase, Ca++ transporting, cardiac muscle, slow twitch 2AI837797−5.8345215020.53249ATPase, H+ transporting, lysosomal 70 kD, V1 subunit A, isoform 1AW545296−4.2807191240.75002AU RNA binding protein/enoyl-coenzyme A hydrataseAV095181−8.7829721740.53747baculoviral IAP repeat-containing 4AV073504−5.1300390530.68359bromodomain-containing 4AV085802−5.7866107270.71518cadherin EGE LAG seven-pass G-type receptor 2BG074441−4.1548793650.71952calcyclin binding proteinBG069742−8.6907063440.65713capping protein alpha 3AV039134−5.0815823570.42546carbonic anhydrase 14AV014385−5.821398140.40180carbonyl reductase 1AI323923−5.2607368150.63722carboxylesterase 3BG072503−9.8553394950.17436cardiac Abnormality/abnormal facies (CATCH22), microdeletion syndrctext missing or illegible when filedAV041840−9.984189610.40426carnitine palmitoyltransferase 2AV006197−5.3125561250.62582caspase 1AA672522−5.4828857520.50832caspase 14AJ007750−4.2707945280.59138catenin srcC77281−5.0608979450.55404cathepsin FAV085152−5.3255133550.51925Cbp/p300-interacting transactivator, with Glu/Asp-rich carboxy-terminatext missing or illegible when filedBG069399−4.2220382940.49555CDC-like kinaseBG065099−4.3903636210.71405cell division cycle 5-like (S. pombe)BG069455−4.1178208710.62771citrate lyase beta likeAV028854−4.1992254910.53480cleavage and polyadenylation specific factor 2, 100 kD subunitAV111435−4.8009131520.49169coagulation factor IIIAA879919−6.6867391140.58633cold inducible RNA binding proteinBG073558−14.83020430.37969complexin 2AV149907−4.7757027690.37946copper chaperone for superoxide dismutaseAV093569−5.2483575110.59552cornichon-like (Drosophila)AV150049−5.4324445460.56343creatine kinase, mitochondrial 2AV085004−4.7420662710.61057cysteine-rich protein 3AV087451−4.2665682190.39188cytochrome c oxidase subunit VIIbAV093625−8.9881388040.39401cytochrome c oxidase, subunit IVaAV005997−4.4874202890.41076cytochrome c oxidase, subunit VbAV088644−4.9495691160.46997cytochrome c oxidase, subunit VI a, polypeptide 2AV001082−4.8423707250.31139cytochrome c oxidase, subunit VI a, polypeptide 2AV030529−4.1525685570.33572cytochrome c oxidase, subunit VIcAV149855−9.1928279770.37223cytochrome c oxidase, subunit VIIa 1AV086493−4.3649239880.27457cytochrome c oxidase, subunit VIIa 3AV133935−5.9368471570.47440cytochrome c oxidase, subunit VIIa 3BG072912−4.121937310.53257cytochrome c oxidase, subunit VIIcBG063960−5.0998037280.37129cytochrome c oxidase, subunit XVII assembly protein homolog (yeast)AV081105−7.9387461280.46201cytochrome c, somaticAV086888−5.7221059980.42669cytochrome c-1AV093672−5.4465891490.68598cytochrome P450, 17AV042908−4.4265172750.37805DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 13 (RNA helicase A)AV106868−6.3749542180.67058DEAD/H (Asp-Glu-Ala-Asp/His) box polypeptide 20BG071005−4.1457614020.69357death associated protein 3BG065205−6.7849492320.48820deleted in polyposis 1AA032557−4.195679490.40696desmocollin 2BG063370−6.6376750790.34694diacylglycerol kinase, alpha (80 kDa)AV069373−4.8082131530.58075diacylglycerol O-acyltransferase 2BG072524−5.2166967410.26003diaphanous homolog 1 (Drosophila)AV134828−4.3499104060.64965DiGeorge syndrome critical region gene 6BG071919−4.999530280.52770dipeptidylpeptidase 4AA266854−5.0034759250.66937DNA fragmentation factor, 40 kD, beta subunitAV109088−4.250800840.65806DNA primase, p49 subunitAV113083−9.8218148430.49491DNA segment, Chr 14, ERATO Doi 574, expressedBG068808−7.4160072660.52173DNA segment, Chr 9, Wayne State University 149, expressedAV135842−4.1652739350.56300DnaJ (Hsp40) homolog, subfamily A, member 3AW540988−6.5427508440.45648DnaJ (Hsp40) homolog, subfamily A, member 3AV050059−6.3117083260.48336DnaJ (Hsp40) homolog, subfamily B, member 9AV041142−4.5949009760.65180DnaJ (Hsp40) homolog, subfamily C, member 1AV057225−5.4773006490.51634dodecenoyl-Coenzyme A delta isomerase (3,2 trans-enoyl-Coenyme Atext missing or illegible when filedAA108563−7.0174805030.35225down-regulated by Ctnnb1, aBG068535−4.5863020980.59629dynein, axon, heavy chain 11AA039110−4.6193234460.41136dystoninBG070533−4.5839001310.55822dystroglycan 1BE137475−4.9606126620.55724E2F transcription factor 6AV126035−4.4402661930.57132ectodermal-neural cortex 1BG065122−5.7052750170.55060endothelial monocyte activating polypeptide 2BG076119−4.9740866980.59151endothelin 1AA511462−4.9198911560.50725enigma homolog (R. norvegicus)AV086590−4.4959358820.46027enoyl coenzyme A hydratase 1, peroxisomalBG074113−6.805825810.36476Eph receptor A4AV089919−4.3441590520.34405ephrin A2AA036231−5.0714774250.55979ESTAV084337−15.846094550.22443ESTAV089256−7.8219457040.32354ESTAV088222−6.0008037560.34203ESTBG067237−5.606600020.37931ESTAV092327−10.73131560.40744ESTBG067593−5.3087337950.40771ESTAV104735−4.2348150340.41649ESTAV107204−4.798997250.41907ESTAV090230−4.5292610680.42529ESTAV032077−5.7396286120.44260ESTBI076847−5.2569432250.44584ESTBG066574−7.1273845510.45000ESTAW558245−5.4784093710.45389ESTAV089999−5.1906655010.45408ESTAW554432−5.8962144110.46163ESTAV006409−5.9640820520.46864ESTAV058135−4.5216495290.47454ESTAI836950−5.9372111880.47461ESTAV092810−5.2419361260.47602ESTAV112960−4.6176281520.47834ESTAW545825−6.7276695460.48212ESTAV085516−4.8426484770.48488ESTAW538191−5.1534589170.48631ESTAU024393−4.8952885830.49035ESTAI836065−4.77550920.49306ESTAA855859−4.3313059580.50195ESTBG068314−5.1992283340.50230ESTAV043406−6.098938170.51042ESTAV066234−4.2544846620.51985ESTAW537378−4.7049894360.52235ESTBI076614−5.1726715390.52412ESTC78728−4.3424690460.52937ESTAV106287−4.1571982490.53067ESTAV084802−5.1666395760.53424ESTAV113584−5.3642822010.53477ESTAV073557−4.5063253460.54223ESTAV058085−8.0959109620.54278ESTAV087849−6.6712096150.54694ESTAV087838−8.7691445580.54700ESTAV113429−6.644940740.54723ESTAI854089−4.2345235510.55638ESTAW539454−4.2985373330.56091ESTAV054545−6.946542870.56151ESTBG065742−13.009333010.56794ESTBG067648−8.6833961490.57773ESTAW537634−5.3245199080.57869ESTAW538620−5.0250493780.58142ESTAW554258−5.8324006460.59289ESTAW558391−4.2573655970.59868ESTAV065563−4.7683485450.60682ESTAW542440−4.4916839330.62565ESTAW558803−5.0203290840.63071ESTAW558059−4.2819107510.63476ESTBG067262−5.9228098480.63861ESTAW556930−4.2462412250.65183ESTBG069129−4.1372771320.66716ESTBG068320−4.215218660.67052ESTBG063124−4.3438591080.67655ESTAV124902−6.2444821470.68098ESTAV066141−4.2585301030.70579ESTAW546201−5.3343342060.71851ESTsAV013380−8.6751102870.12285ESTsAI839959−11.808272480.26051ESTsAV087279−10.847389740.37033ESTsBG074584−4.9918480580.41016ESTsBG071766−7.1404495390.41412ESTsBG064317−5.7237771220.42958ESTsBG071847−5.9281356780.43532ESTsAW558570−4.4801541950.45840ESTsBG069296−5.2409174480.46577ESTsAV028938−4.1515412410.48718ESTsAI840562−12.066835490.49094ESTsAV026027−4.5069395080.49232ESTsAV006522−4.6138198920.52324ESTsAV083513−4.8282515770.53129ESTsBG073031−4.5663062640.53403ESTsBG075173−5.0285065370.53874ESTsBG063906−8.0893709790.54039ESTsBG066954−4.7826154570.54260ESTsBG067242−6.823323780.54553ESTsBG072934−5.2283131950.54677ESTsAI854088−4.1595982390.55320ESTsBG073667−10.484927220.55826ESTsBG065948−4.8600616530.56492ESTsAV031990−6.5493274090.56848ESTsBG067986−7.074527910.58210ESTsBG067553−5.0004436360.59575ESTsAV033253−4.2130523140.59746ESTsBG066080−7.1788656260.60242ESTsAV094549−5.4484656010.61795ESTsBG069475−5.1979761150.63287ESTsBG073483−5.5808966250.63556ESTsAU043006−6.9020270480.63790ESTsAW557124−4.4003326720.67259ESTsBG071818−6.1647347240.67323ESTsAV087922−5.4635511980.68467ESTsBG073793−5.5562897840.69451ESTsAV029719−4.645728080.70854ESTsAU040991−4.6563300270.71007ESTsAV123079−4.4879538870.79323ESTsAA219953−4.9284763020.81818ESTs, Highly similar to NUMM MOUSE NADH-UBIQUINONE OXIDORtext missing or illegible when filedAV053614−4.8920193150.42037ESTs, Highly similar to SR68_HUMAN SIGNAL RECOGNITION PARTtext missing or illegible when filedAA044456−5.7791404150.63127ESTs, Moderately similar to CENC MOUSE CENTROMERE PROTEINtext missing or illegible when filedBG070887−6.9371331220.49208ESTs, Moderately similar to COXM MOUSE CYTOCHROME C OXIDAtext missing or illegible when filedBG073133−4.3826143290.38552ESTs, Moderately similar to hypothetical protein MGC2217 [Homo saptext missing or illegible when filedAV140202−5.8840985320.42443ESTs, Moderately similar to put. gag and pol gene product [M. musculutext missing or illegible when filedAU017598−4.669175380.61340ESTs, Moderately similar to T29098 microtubule-associated protein 4,AV085051−4.6521204470.41777ESTs, Moderately similar to TSC1_RAT HAMARTIN (TUBEROUS SCItext missing or illegible when filedBG073522−4.5283640310.57654ESTs, Moderately similar to unnamed protein product [H. sapiens]BG069242−5.8640255220.48855ESTs, Weakly similar to 17-beta hydroxysteroid dehydrogenase type 2AV012778−5.995460570.29569ESTs, Weakly similar to A48133 pre-mRNA splicing SRp75 [H. sapienstext missing or illegible when filedBG068996−8.427673350.41807ESTs, Weakly similar to COXD MOUSE CYTOCHROME C OXIDASEAV088683−4.6866505350.38315ESTs, Weakly similar to DIA3_MOUSE Diaphanous protein homolog 3BG066491−5.6035513570.42357ESTs, Weakly similar to F-actin binding protein b-Nexilin [R. norvegicustext missing or illegible when filedAU022020−5.0300694520.55649ESTs, Weakly similar to FOR4 MOUSE FORMIN 4 [M. musculus]BG068457−5.1274101890.51270ESTs, Weakly similar to proline rich protein 2 [Mus musculus] [M. musctext missing or illegible when filedBG068802−6.5783075440.63820ESTs, Weakly similar to S33477 hypothetical protein 1 —rat [R. norvegitext missing or illegible when filedBG063187−4.6662267940.59621ESTs, Weakly similar to S48081 GRSF-1 protein [H. sapiens]AV074326−4.3282781090.58441ESTs, Weakly similar to SNAP190 [H. sapiens]AV094673−4.3685909020.62151ESTs, Weakly similar to testis derived transcript 3 [Mus musculus] [M. rtext missing or illegible when filedBG065317−5.1445199480.39289ESTs, Weakly similar to TLM MOUSE TLM PROTEIN [M. musculus]AV092958−6.1504037410.45074eukaryotic translation elongation factor 1 delta (guanine nucleotide exctext missing or illegible when filedAA253918−4.1865699860.57143eukaryotic translation elongation factor 2BG067570−6.3710444440.65020eukaryotic translation initiation factor 2 alpha kinase 3AV095205−5.0593933190.56401eukaryotic translation initiation factor 3, subunit 2 (beta, 36 kD)AV094437−4.6015273120.45547excision repair cross-complementing rodent repair deficiency, complentext missing or illegible when filedBG063161−5.5470508720.63136expressed sequence AA407270BG063148−5.935660940.40575expressed sequence AA407270AV024203−5.7713682250.55519expressed sequence AA408168BG066580−7.7201424580.42173expressed sequence AA408877AV009485−7.3318433420.44266expressed sequence AA408877BG063884−7.5497362890.69757expressed sequence AA959758BG070652−6.2105695040.69281expressed sequence AA959857AV109470−6.1111992310.57250expressed sequence AA960047AV033573−4.6328110110.71552expressed sequence AI197390BG064453−4.4474293920.65801expressed sequence AI256693AV083357−7.0615942270.44924expressed sequence AI256693BG062933−6.840694010.50397expressed sequence AI314967BG075147−9.7004266660.58836expressed sequence AI315037AV014911−4.1689171280.46734expressed sequence AI414265BG063334−5.3740788730.35065expressed sequence AI428506AV032231−4.3120841530.46225expressed sequence AI428794BG076075−4.2283797090.69144expressed sequence AI450287BG065344−6.1678757560.74403expressed sequence AI451892AV032341−4.4050358520.58191expressed sequence AI452301BI076508−8.1972080430.54245expressed sequence AI462702BG068253−6.4183108830.57868expressed sequence AI480535AV083879−5.1870495080.47634expressed sequence AI504630AV015284−5.8883942360.56047expressed sequence AI595366AV086025−7.2092649220.54969expressed sequence AI604911BG063457−6.278693330.60458expressed sequence AI746547BG073543−4.3034743740.66202expressed sequence AI838773AV013448−5.4303202970.51111expressed sequence AU022809AU022809−6.8778202530.37946expressed sequence AU040217AV006387−4.6014371440.37921expressed sequence AU043990AV085893−4.610608750.61610expressed sequence AV006127AV006127−4.9684788140.55637expressed sequence AV028368AV010507−4.920032120.42417expressed sequence AW122032BG071778−5.4498358280.53237expressed sequence AW125446BG070892−6.5045251670.53458expressed sequence AW215868BG069736−4.2846513890.71600expressed sequence AW495846BG076492−4.4618761370.66865expressed sequence AW545363AV060425−4.6997713880.68385expressed sequence AW554339AW554339−4.9908965060.68667expressed sequence AW555814BG065375−5.7292643120.37042expressed sequence C76711C76711−4.6737010330.54362expressed sequence C78643C78643−4.9232709520.57835expressed sequence C79026BG066389−4.287483570.68151expressed sequence C81189BG066971−5.5973952750.41821expressed sequence C85317BG067152−5.1358346080.52423expressed sequence C86676BG069605−5.5669570460.59228expressed sequence C87882BG067895−5.3511812140.51928expressed sequence R74645AV032243−4.8370232480.46405Fas-activated serine/threonine kinaseBG074856−4.2170256130.45434fatty acid binding protein 3, muscle and heartAV006024−7.3087564310.40356fatty acid Coenzyme A ligase, long chain 2AV006061−4.9418667690.48297FBJ osteosarcoma oncogene BBG076079−7.0427463770.52580f-box and leucine-rich repeat protein 12BG067545−4.4002643810.77610fibroblast growth factor receptor 4AI385693−5.907856260.48522FK506 binding protein 3 (25 kD)AV134155−12.240598790.46456forkhead box C1A1415347−4.2995848930.64530four and a half LIM domains 2BG065614−4.8373224630.40643G protein-coupled receptor kinase 7AV005838−5.2825170480.50864galactokinaseAV108357−4.3910300160.47824gamma-glutamyl transpeptidaseAA162908−4.5629534330.41377gelsolinAV170949−7.8116444750.39819gene rich cluster, C8 geneC81126−7.150728210.68777genes associated with retinoid-IFN-induced mortality 19BG073545−6.9673461660.40268glioblastoma amplified sequenceAV082190−7.3365747110.44947glucocorticoid-induced leucine zipperW33468−4.3779773940.39408glutamate oxaloacetate transaminase 1, solubleBG066689−5.1131969580.41673glutamine synthetaseAV009064−5.4943225060.38899glutathione S-transferase, alpha 4AV084880−5.6202685080.49942glutathione S-transferase, mu 1BG074268−4.9049816350.48909glycosylphosphatidylinositol specific phospholipase D1AV086924−6.0858905140.44720granzyme BAV038272−4.6068810060.42438growth factor receptor bound protein 2-associated protein 1BG063323−4.1730212490.73731guanosine monophosphate reductaseAV103032−4.1214590060.49495H2A histone family, member YC75971−9.6329300020.29998heat shock 10 kDa protein 1 (chaperonin 10)AV055529−4.143886020.66410heat shock protein, 70 kDa 3AV223941−4.7178675230.42727heme oxygenase (decycling) 1AV083964−9.1301086620.57613hemoglobin, beta adult major chainAV108710−6.5753288420.48588histidine ammonia lyaseAV022721−5.3579605580.44637histidine rich calcium binding proteinBG073810−7.7233746490.29908histidine triad nucleotide binding proteinAA154889−4.9367982820.68692histocompatibility 47AV036651−7.3475033050.63359homeo box C4AA245472−4.463922460.41142homocysteine-inducible, endoplasmic reticulum stress-inducible, ubiqutext missing or illegible when filedAV086303−4.4507950310.32623hydroxysteroid (17-beta) dehydrogenase 10BG073539−5.7574172260.49471hypothetical protein, MGC: 6943AV085351−4.5478111080.62294hypothetical protein, MGC: 6989AV031846−4.9324528860.38973hypothetical protein, MGC: 7550AV087882−8.3759708890.61973immediate early responses 5BG069628−4.1584604060.56982immunoglobulin superfamily, member 7AV073565−7.8649778710.52541insulin-like growth factor binding protein 4AV005795−5.3684165820.18068insulin-like growth factor binding protein 5AV087798−6.3672473480.43614integrin binding sialoproteinAV171934−4.992909280.34304interferon activated gene 204AV015208−7.7013313190.64560interferon activated gene 205AV058630−8.0151909460.34982interferon-related developmental regulator 1AA107115−4.3669312880.67719iroquois related homeobox 4 (Drosophila)AV006035−6.230996420.58603isocitrate dehydrogenase 2 (NADP+), mitochondrialAV089252−5.2786872850.45360isocitrate dehydrogenase 3 (NAD+) alphaBG068774−4.554878210.45957isocitrate dehydrogenase 3 (NAD+) betaAA036340−4.1622693180.47460isovaleryl coenzyme A dehydrogenaseBG070984−8.7679356050.30518Janus kinase 1BG067874−7.254517750.65078Janus kinase 2AA153109−5.3075866450.64858keratin associated protein 6-2AV013499−5.5251318150.38744keratin complex 2, basic, gene 16AA738772−4.2660874470.51812keratin complex 2, basic, gene 18AV086522−4.9891884040.40787keratin complex 2, basic, gene 6gAV008410−5.4811040590.33635L-3-hydroxyacyl-Coenzyme A dehydrogenase, short chainAA122758−7.4892594260.44349lactate dehydrogenase 2, B chainAV171750−4.6525807190.33146leucine zipper-EF-hand containing transmembrane protein 1AV083103−4.8471707190.65147LIM domain binding 3AV088371−4.4011963680.41447lipin 1AV022047−4.9140163940.52166lipoprotein lipaseAV084650−4.8393341450.42555lipoprotein lipaseAV006290−11.424644590.42847low density lipoprotein receptor-related protein 2BG064854−4.2201868030.59503lurcher transcript 1BG074415−6.2442743610.41951lysosomal apyrase-like 1AV086322−6.7757812990.65322lysosomal membrane glycoprotein 2BG074453−6.2481535870.74154malate dehydrogenase, solubleAV093576−5.2029574560.32039MAP kinase-activated protein kinase 2AA030342−7.5979642060.59516MAP kinase-activated protein kinase 5AA616241−6.2811755940.51661maternal embryonic leucine zipper kinaseAV140411−5.560583330.51604membrane-associated protein 17AV060358−4.8062942560.39397methyl-CpG binding domain protein 4AV032932−4.6289185390.55652methylmalonyl-Coenzyme A mutaseAV031545−5.4679118030.50168microsomal glutathione S-transferase 3AV056432−4.3335913340.41688microtubule-associated protein tauBG066372−4.1169547260.42329mitochondrial ribosomal protein 64AV094889−4.4905030040.63412mitochondrial ribosomal protein L15BG064987−5.2291426030.54936mitochondrial ribosomal protein L16BG075780−4.1488724640.60350mitochondrial ribosomal protein L23BG071604−7.0592491110.49751mitochondrial ribosomal protein L39AV150063−6.9431795030.67150mitochondrial ribosomal protein L43AV094774−4.9689394330.69126mitochondrial ribosomal protein S17BG071752−5.2272577810.42507mitochondrial ribosomal protein S25BG065867−6.4630010450.47504mitochondrial ribosomal protein S31AV058185−4.9433289850.52131mitogen activated protein binding protein interacting proteinAV134069−5.0845043280.63511mitogen-activated protein kinase kinase kinase 7 interacting protein 2AV011185−5.2697668340.51165MLN51 proteinAW556296−6.2391036870.56037Mus musculus 10 day old male pancreas cDNA, RIKEN full-length enritext missing or illegible when filedAV058496−9.8671615290.43027Mus musculus 10, 11 days embryo whole body cDNA, RIKEN full-lengtext missing or illegible when filedBG075565−6.1736633430.72665Mus musculus brain and reproductive organ-expressed protein (Bre) mtext missing or illegible when filedAV073509−4.8835818120.51095Mus musculus methyl-CpG binding domain protein 3-like protein 2 (Mbtext missing or illegible when filedBG071308−5.7169813720.53500Mus musculus QIL1 (Qil1) mRNA, complete cdsBG072356−5.8416029160.46840Mus musculus, clone IMAGE: 3491909, mRNA, partial cdsBG071756−4.4963038750.65826Mus musculus, clone IMAGE: 4482598, mRNAAA034560−4.1502990720.31779Mus musculus, clone IMAGE: 5357662, mRNA, partial cdsAV042520−4.4085849420.60396Mus musculus, clone MGC: 11691 IMAGE: 3962417, mRNA, complete text missing or illegible when filedAV084848−5.4903161330.52085Mus musculus, clone MGC: 36369 IMAGE: 4982239, mRNA, complete text missing or illegible when filedAV094465−5.447744350.49239Mus musculus, clone MGC: 6816 IMAGE: 2648797, mRNA, complete ctext missing or illegible when filedAV014114−4.2828505340.53438Mus musculus, clone MGC: 7480 IMAGE: 3490700, mRNA, complete ctext missing or illegible when filedAV034637−5.9874568340.50215Mus musculus, clone MGC: 7530 IMAGE: 3492114, mRNA, complete ctext missing or illegible when filedAV089939−6.8333876840.58423Mus musculus, H4 histone family, member A, clone MGC: 30488 IMAGtext missing or illegible when filedAV113959−4.6224264460.45955Mus musculus, hypothetical protein MGC11287 similar to ribosomal ptext missing or illegible when filedAV031726−5.5848504450.70092Mus musculus, Similar to 3-hydroxyisobutyrate dehydrogenase, clone IAI854120−5.2498486610.50351Mus musculus, Similar to ATPase, Na+/K+ transporting, alpha 1a.1 potext missing or illegible when filedAA063844−4.7124319210.52469Mus musculus, Similar to chromosome 18 open reading frame 1, clonetext missing or illegible when filedBG070238−4.2519265110.72193Mus musculus, Similar to electron-transfer-flavoprotein, alpha polypeptext missing or illegible when filedAV088774−5.687500460.47951Mus musculus, Similar to glutamate rich WD repeat protein GRWD, ctext missing or illegible when filedBG071389−4.4641681520.69603Mus musculus, Similar to hypothetical protein BC004409, clone MGC: text missing or illegible when filedAV086576−5.2114554560.54638Mus musculus, Similar to hypothetical protein MGC4368, clone MGC: 2BG065643−4.1409090890.53064Mus musculus, Similar to hypothetical protein MGC4368, clone MGC: 2AV005807−4.4482469340.54984Mus musculus, Similar to hypothetical protein, clone MGC: 19257 IMAtext missing or illegible when filedAV055251−5.9640315650.71353Mus musculus, Similar to mannosyl (alpha-1,3-)-glycoprotein beta-1,4-text missing or illegible when filedBG063179−4.9638935640.68444Mus musculus, Similar to metallothionein 1, clone MGC: 27821 IMAGE:text missing or illegible when filedAV149953−5.0094098820.38263Mus musculus, Similar to MIPP65 protein, clone MGC: 18783 IMAGE: 4AV109599−4.7690205130.62297Mus musculus, Similar to PTD015 protein, clone MGC: 36240 IMAGE: 5AV088778−4.303127820.51111Mus musculus, Similar to secretory leukocyte protease inhibitor, cloneAV089194−5.3935530480.56725Mus musculus, Similar to transmembrane protein 5, clone MGC: 28135text missing or illegible when filedAV095048−4.7554426460.65205myeloblastosis oncogeneAV222464−5.5943730430.63770myeloid leukemia factor 1AV042698−6.2860603460.36555myosin binding protein C, cardiacAV005840−4.404790520.56183myosin light chain, alkali, cardiac atriaAV005821−7.0479644240.31699N-acetyltransferase ARD1 homolog (S. cerevisiae)AI841645−4.2308555830.72328NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 2AV016078−6.7934614750.40427NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 2AV093541−5.3802074210.51264NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 1AV140287−7.6712349890.49739NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 4AV050140−4.6417987890.43550NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 6 (14 kD, B1text missing or illegible when filedAV106199−5.5402010210.41067NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 6 (14 kD, B1text missing or illegible when filedAV087995−4.8577596920.46752NADH dehydrogenase (ubiquinone) 1 alpha subcomplex, 7 (14.5 kD, Btext missing or illegible when filedAV133797−4.4633388460.45989NADH dehydrogenase (ubiquinone) 1 beta subcomplex 5AV057902−6.333454290.40844NADH dehydrogenase (ubiquinone) 1 beta subcomplex, 9BG075174−5.5250397060.44325NADH dehydrogenase (ubiquinone) 1, subcomplex Unsequenced ESTtext missing or illegible when filedAV088122−4.473288540.43713NADH dehydrogenase (ubiquinone) Fe—S protein 3BG076060−7.8292526990.40260NADH dehydrogenase (ubiquinone) Fe—S protein 4BG066265−4.7867955980.56585nebulin-related anchoring proteinAV013274−4.7098649850.31656neurotensin receptor 2AV032954−6.3947901550.34827Niemann Pick type C1AV012796−5.8182454820.57019N-myc downstream regulated 2AV149939−4.9565489730.47960non MHC restricted killing associatedBG076189−5.9065322970.56544N-sulfotransferaseAV051308−4.5483627270.41566nuclear distribution gene C homolog (Aspergillus)BG073422−10.86265690.56353nuclear receptor coactivator 6 interacting proteinAV113681−6.1486699950.34592nuclear receptor interacting protein 1AI840578−4.6127423670.59793nuclear receptor subfamily 2, group F, member 1BG071238−4.9806255320.35648nuclear transcription factor-Y betaAV016446−6.2464442830.41297olfactomedin 1BG073096−7.2862356880.39555oxysterol binding protein-like 1ABG073162−6.8129131310.57590p53 apoptosis effector related to Pmp22BG065306−4.6789754040.40269p53 regulated PA26 nuclear proteinBG076140−5.4483061490.55541paired box gene 6AV032892−4.4886299510.61857pantophysinAV091203−4.1491007990.69535PCTAIRE-motif protein kinase 1AV157322−5.0352900360.46140pellino 1BG063809−6.1566179860.49251peptidase 4U51014−4.33230710.47568peptidylprolyl isomerase (cyclophilin)-like 1AV015645−4.8212473510.32093periplakinBG074644−4.7574372180.33818peroxiredoxin 3AA168985−10.69037420.41739peroxiredoxin 6AV052763−4.5301391450.54965peroxisomal membrane protein 2, 22 kDaBG073687−5.2661962310.36957peroxisomal membrane protein 3, 35 kDaBG075110−4.8515559620.58487peroxisome proliferative activated receptor, gamma, coactivator 1AF049330−5.7418199350.48224phosphate cytidylyltransferase 1, choline, alpha isoformBG071157−8.2145813060.56759phosphatidylinositol 3 kinase, regulatory subunit, polypeptide 4, p150BG069962−5.6346624610.72045phosphofructokinase, muscleAV012100−4.8633783380.31668phospholipase A2 group VII (platelet-activating factor acetylhydrolase,AV033702−4.1768052140.45211phospholipase A2, group IB, pancreasAV085478−7.1510344270.68461phosphoribosylglycinamide formyltransferaseAV009977−6.778433990.62257phytanoyl-CoA hydroxylaseAV084314−9.878018120.28442platelet-derived growth factor receptor-likeBG068957−5.0609995510.39457polymyositis/scleroderma autoantigen 2BG063453−5.5307265710.44618potassium voltage-gated channel, Shal-related family, member 2BG075283−4.7520894010.48273pre-B-cell colony-enhancing factorAV108470−4.1838279470.53050prefoldin 2AU020724−6.5516941730.50227pregnancy upregulated non-ubiquitously expressed CaM kinaseAI391204−4.9764554250.67410programmed cell death 5BG063248−4.3467509220.47631proteasome (prosome, macropain) 26S subunit, non-ATPase, 4AV111455−4.7862663110.70045proteasome (prosome, macropain) subunit, alpha type 7AV093698−7.2069241460.71542proteasome (prosome, macropain) subunit, beta type 6AV093807−4.1352750650.73806protein kinase inhibitor, gammaBG073627−5.4076772930.66327protein kinase, AMP-activated, gamma 1 non-catalytic subunitBG067722−5.1742841790.48660protein phospatase 3, regulatory subunit B, alpha isoform (calcineurin text missing or illegible when filedAV006032−4.2458764610.32451protein tyrosine phosphatase, non-receptor type 9AV114744−4.2378595460.58064pyruvate dehydrogenase E1 alpha 1BG068736−6.3335674910.40029quakingBG068631−4.930717260.57698Rab acceptor 1 (prenylated)BG072002−5.6080122060.48144RAN guanine nucleotide release factorAV133777−4.362796120.59926RAS-homolog enriched in brainAV095119−4.8792115650.53004RAS-related C3 botulinum substrate 1BG076502−6.0409338520.60293receptor (calcitonin) activity modifying protein 2AV085507−5.3033833780.54970receptor-associated protein of the synapse, 43 kDaAV061434−10.618621140.41436regulator of G-protein signaling 2BG068533−4.8352829560.27907reticulon 2 (Z-band associated protein)AV088718−5.6233163290.44935retinoic acid induced 1AV012729−4.2900303080.63998retinoid X receptor gammaAV089219−5.8222131610.49561ribosomal protein L27aAV013292−4.4372539140.49756ribosomal protein L30BG065356−4.2529741130.68577ribosomal protein L37aAI837822−5.1540493850.59292ribosomal protein S25AV093430−4.6583355140.58295ribosomal protein S29L31609−6.1106647660.45134RIKEN cDNA 0610006N12 geneAA110681−6.751850870.40291RIKEN cDNA 0610007H07 geneBG072309−4.1261290220.60173RIKEN cDNA 0610009D10 geneAA154397−7.084662560.34713RIKEN cDNA 0610009I16 geneAV086609−7.2361996690.35051RIKEN cDNA 0610010E03 geneAI841340−6.8022494850.47787RIKEN cDNA 0610010I17 geneAV056903−5.5387545960.46727RIKEN cDNA 0610010I23 geneAV051596−4.3288199550.61515RIKEN cDNA 0610011B04 geneBG073700−6.5559968540.38623RIKEN cDNA 0610011L04 geneBG072552−5.0544433340.37549RIKEN cDNA 0610025I19 geneAV085433−17.568099080.22127RIKEN cDNA 0610033L03 geneAV093484−7.0392847040.41225RIKEN cDNA 0610039N19 geneAV083519−5.4064483240.41668RIKEN cDNA 0610039N19 geneBG066600−5.3308824680.45065RIKEN cDNA 0610040D20 geneAV004247−4.5127573980.63567RIKEN cDNA 0710008D09 geneAW558029−4.7291466920.46971RIKEN cDNA 1010001M12 geneAV086467−7.480408130.44085RIKEN cDNA 1010001N11 geneAV133828−4.6861040190.46207RIKEN cDNA 1100001F19 geneBG070073−5.2888226970.68489RIKEN cDNA 1110001A12 geneBG070781−4.7038357150.64679RIKEN cDNA 1110001I24 geneAV140151−6.0528027970.36840RIKEN cDNA 1110001J03 geneAV065564−4.1922975910.32893RIKEN cDNA 1110001O19 geneAV056481−4.3140173960.56079RIKEN cDNA 1110003P16 geneBG075816−4.463639540.51085RIKEN cDNA 1110003P16 geneAV057754−4.9706042640.55663RIKEN cDNA 1110004A22 geneBG071279−4.4577972040.48172RIKEN cDNA 1110007A04 geneAV055217−4.9691070850.47342RIKEN cDNA 1110007C09 geneAV051158−4.1187861570.53859RIKEN cDNA 1110008L20 geneAV018091−4.6975079590.52248RIKEN cDNA 1110013H04 geneAV052337−6.7881623380.45818RIKEN cDNA 1110013H04 geneBG068276−6.068328920.56841RIKEN cDNA 1110018B13 geneAV028535−4.6150838550.43160RIKEN cDNA 1110018B13 geneAV084595−5.973221810.57666RIKEN cDNA 1110020I04 geneAV051530−14.920320870.30711RIKEN cDNA 1110020I04 geneBG063739−4.4638076890.47696RIKEN cDNA 1110020J08 geneAW550860−4.6147278870.61323RIKEN cDNA 1110021D01 geneAV071376−4.584102450.79871RIKEN cDNA 1110028A07 geneAV085772−6.1749190650.39958RIKEN cDNA 1110031C13 geneAV041472−5.0284193890.46491RIKEN cDNA 1110031I02 geneAU043030−4.4037553690.51919RIKEN cDNA 1110036H21 geneAV012479−5.1600747270.45281RIKEN cDNA 1110054G21 geneAV014368−5.0279010580.49410RIKEN cDNA 1110063J16 geneAV078407−5.9997468910.59492RIKEN cDNA 1110065A22 geneAV016366−4.925417620.51442RIKEN cDNA 1190002A23 geneAV024081−5.5357595160.60154RIKEN cDNA 1190002L16 geneBG071000−6.4905993790.52952RIKEN cDNA 1190006F07 geneAI839764−6.7665918420.28987RIKEN cDNA 1190006F07 geneBG072458−4.6153570670.47455RIKEN cDNA 1190006L01 geneBG076352−6.2382044320.38844RIKEN cDNA 1190017B19 geneAV022384−4.2860490690.61201RIKEN cDNA 1200006O19 geneBG071963−4.9044341260.49222RIKEN cDNA 1200006O19 geneAV074439−4.3599263630.57055RIKEN cDNA 1200007E24 geneBG075635−5.5476063020.54461RIKEN cDNA 1200009K13 geneBG069392−4.4973460280.66746RIKEN cDNA 1200015P04 geneAV065655−6.1522369460.15180RIKEN cDNA 1200015P04 geneAV067337−8.6369684520.18033RIKEN cDNA 1200015P04 geneAI840878−8.0896369150.18339RIKEN cDNA 1200015P04 geneAV068725−9.7964660540.22295RIKEN cDNA 1300002C13 geneBG064110−6.4287153650.48112RIKEN cDNA 1300013G12 geneBG076497−6.9398021290.53379RIKEN cDNA 1300013J15 geneAV082636−4.4316834420.42023RIKEN cDNA 1300017C12 geneBG069813−5.1588001130.47198RIKEN cDNA 1300019P08 geneAV094927−6.0364523380.46761RIKEN cDNA 1500001L03 geneBG067671−4.7405207760.33865RIKEN cDNA 1500004O06 geneAV084141−10.933314110.53732RIKEN cDNA 1500004O06 geneAV095102−4.3372758850.59115RIKEN cDNA 1500010M16 geneAV162350−4.3991182430.53491RIKEN cDNA 1500012D08 geneAV094880−5.3540926170.47779RIKEN cDNA 1500032E05 geneAI894110−5.2724454030.58956RIKEN cDNA 1500034J20 geneAV111483−8.4957555770.49446RIKEN cDNA 1500036F01 geneAV074483−4.1692902220.23080RIKEN cDNA 1600014J01 geneAV051090−6.5328507950.57481RIKEN cDNA 1600023A02 geneAV002462−4.7356997620.55362RIKEN cDNA 1700006F03 geneBG071686−6.4919081380.57462RIKEN cDNA 1700013G20 geneBG067233−5.5771437060.50168RIKEN cDNA 1700016D08 geneBG073980−4.2955786490.66457RIKEN cDNA 1700029P11 geneAV043746−4.9813580210.38488RIKEN cDNA 1700029P11 geneAV043137−8.4285404810.48877RIKEN cDNA 1810004I06 geneAV050264−5.0211839230.33763RIKEN cDNA 1810004I06 geneAV070272−4.3355004640.53518RIKEN cDNA 1810008A14 geneBG063535−8.6360213460.63781RIKEN cDNA 1810011O01 geneAV070830−5.4210785040.43645RIKEN cDNA 1810013D10 geneBG067851−4.8923798630.54634RIKEN cDNA 1810013K23 geneAW539206−4.2826266410.50783RIKEN cDNA 1810017G16 geneAV087873−7.8880583850.46376RIKEN cDNA 1810017G16 geneAV051238−4.5213249670.51059RIKEN cDNA 1810017G16 geneAV070773−4.1283556530.68677RIKEN cDNA 1810018M11 geneAV018921−9.4161929260.60647RIKEN cDNA 1810020E01 geneAV032033−5.1367987750.45741RIKEN cDNA 1810029B16 geneBG069652−6.0387297230.56189RIKEN cDNA 1810030E18 geneAV140504−5.274692450.67706RIKEN cDNA 1810030E20 geneBG064141−4.9329562160.58007RIKEN cDNA 1810030E20 geneBG063825−4.2290664610.64290RIKEN cDNA 1810033A19 geneAV054886−5.0434680740.60235RIKEN cDNA 1810035L17 geneBG072596−5.5484841270.58195RIKEN cDNA 1810036J22 geneAV113916−19.446254790.47866RIKEN cDNA 1810036J22 geneAV084361−5.9731720860.50101RIKEN cDNA 1810036J22 geneAV086261−5.2814648130.52027RIKEN cDNA 1810036J22 geneBG064173−5.1732726990.59456RIKEN cDNA 1810055D05 geneAV140588−5.312587470.39893RIKEN cDNA 1810055D05 geneAV065469−4.6765212560.43368RIKEN cDNA 1810055D05 geneAV059067−5.7064890380.56482RIKEN cDNA 2010003O02 geneBG066308−4.6368184780.52627RIKEN cDNA 2010004E11 geneAV066070−5.2936767180.58290RIKEN cDNA 2010100O12 geneBG075840−5.1843557360.56372RIKEN cDNA 2010100O12 geneAV088623−7.0436812290.61838RIKEN cDNA 2010107E04 geneBG076108−4.6767702210.48870RIKEN cDNA 2010110I09 geneBG072417−8.0470569710.50518RIKEN cDNA 2010110M21 geneAV031008−4.1522716010.62642RIKEN cDNA 2010110M21 geneAV006309−5.1743306030.63652RIKEN cDNA 2210008F15 geneAV085342−6.7609586520.43695RIKEN cDNA 2210008F15 geneAV140597−4.9767529040.50033RIKEN cDNA 2210009K14 geneAV074534−4.2442318080.58997RIKEN cDNA 2210016H18 geneAW556974−4.6952602230.48019RIKEN cDNA 2210415M14 geneAV063132−4.151385790.41701RIKEN cDNA 2210415M14 geneAV123133−6.8668913090.46633RIKEN cDNA 2210415M14 geneBG072853−5.899831160.46756RIKEN cDNA 2210418G03 geneAV081301−7.3828772160.59853RIKEN cDNA 2310001N14 geneAV083256−9.4714647780.35457RIKEN cDNA 2310002J21 geneBG063238−4.1779260760.64768RIKEN cDNA 2310005O14 geneAV104008−5.6444979120.55170RIKEN cDNA 2310015J09 geneAV085812−5.0793011580.32950RIKEN cDNA 2310016E22 geneAV085956−4.5081873610.53050RIKEN cDNA 2310016M24 geneAV109219−6.1746854790.45223RIKEN cDNA 2310020D23 geneAA087197−4.9899162770.70975RIKEN cDNA 2310020H20 geneBG063177−4.1629785420.49609RIKEN cDNA 2310021J10 geneAV086427−5.2498298960.41447RIKEN cDNA 2310026J01 geneAV087038−6.2240529950.18088RIKEN cDNA 2310034L04 geneAV088072−4.8576176070.43830RIKEN cDNA 2310039H15 geneAV103530−5.7625867810.37401RIKEN cDNA 2310039H15 geneAV088685−10.655239150.42365RIKEN cDNA 2310039H15 geneAV006258−4.7700804820.48698RIKEN cDNA 2310042M24 geneAV089703−4.9578306130.70818RIKEN cDNA 2310042N02 geneAV089174−5.2274615260.44265RIKEN cDNA 2310045A07 geneAV089574−5.7947322030.36180RIKEN cDNA 2310051E17 geneAV090635−5.3863543880.39477RIKEN cDNA 2310056B04 geneBG074855−4.9288861120.54397RIKEN cDNA 2310058J06 geneAV171032−5.5667356010.50412RIKEN cDNA 2310066N05 geneAV109445−4.1363802510.71050RIKEN cDNA 2310067L22 geneAV085162−6.0656669620.43059RIKEN cDNA 2310076O14 geneAV093026−5.2882229690.46965RIKEN cDNA 2310079P10 geneBG069582−10.794670490.31277RIKEN cDNA 2400003N08 geneBG068322−5.8318626960.57334RIKEN cDNA 2400006N03 geneAV095106−5.0229675820.63521RIKEN cDNA 2400010D15 geneBG070770−5.4256061320.50504RIKEN cDNA 2400010D15 geneAV014412−5.4226338490.58352RIKEN cDNA 2400010G15 geneAV087844−5.2410427610.59067RIKEN cDNA 2410004H02 geneAV095143−4.6612736810.52258RIKEN cDNA 2410004H02 geneBG065078−4.4259364650.60061RIKEN cDNA 2410005O16 geneAV085399−4.3040450510.66223RIKEN cDNA 2410011G03 geneBG072634−7.1025540290.34324RIKEN cDNA 2410011G03 geneAV140158−7.4122585540.53256RIKEN cDNA 2410016F19 geneBG066198−4.1538057220.67772RIKEN cDNA 2410030A14 geneAV095185−4.8825463380.56335RIKEN cDNA 2410043G19 geneAV056739−5.5797869150.39668RIKEN cDNA 2410066K11 geneBG074815−4.1894995930.65618RIKEN cDNA 2410166I05 geneBG076161−7.7465656350.56369RIKEN cDNA 2510027N19 geneBG063257−4.4240353370.64005RIKEN cDNA 2510048K03 geneAV050186−7.2148477490.39540RIKEN cDNA 2600001N01 geneBG065115−4.6228084020.65666RIKEN cDNA 2610002K22 geneAV095125−4.2222241940.65841RIKEN cDNA 2610003B19 geneAV077867−5.3924358010.50676RIKEN cDNA 2610020H15 geneBG067911−4.331849070.50925RIKEN cDNA 2610028H24 geneAU041304−8.8379084740.42891RIKEN cDNA 2610034N03 geneAV104092−4.3342791840.60381RIKEN cDNA 2610041P16 geneBG063943−9.1715423270.39169RIKEN cDNA 2610041P16 geneAV086193−4.4373905230.53171RIKEN cDNA 2610205H19 geneAV149977−5.0751804190.54297RIKEN cDNA 2610509H23 geneBG073333−4.5291887320.67762RIKEN cDNA 2610529I12 geneAV112870−4.1471331650.55866RIKEN cDNA 2700018N07 geneAI327124−4.297623640.56436RIKEN cDNA 2700033I16 geneAV060239−4.3626232190.48215RIKEN cDNA 2700049M22 geneAU022477−6.2425661560.56361RIKEN cDNA 2700055K07 geneAV086940−5.8093670540.33093RIKEN cDNA 2700094L05 geneBG070651−6.7432450250.63558RIKEN cDNA 2810403A07 geneBG064481−4.9394258610.70126RIKEN cDNA 2810403L02 geneAI838447−5.4764844950.79272RIKEN cDNA 2810417D04 geneAV141701−4.4399030750.53864RIKEN cDNA 2810422J05 geneBG064518−5.0979755310.54326RIKEN cDNA 2810432N10 geneBG070211−4.8112030490.51703RIKEN cDNA 2810468K05 geneBG071137−5.3421572380.70066RIKEN cDNA 2900010I05 geneAV056021−4.7745540890.48993RIKEN cDNA 2900055D03 geneAV140126−4.2714571430.50891RIKEN cDNA 3110004H13 geneBG071859−6.0464216310.54200RIKEN cDNA 3110005M08 geneAV108251−4.2063770490.72772RIKEN cDNA 3200001M24 geneAV093570−4.1299693770.55745RIKEN cDNA 3200001M24 geneBG074430−4.3544662690.66040RIKEN cDNA 3230402N08 geneAV089737−4.4657018640.65941RIKEN cDNA 3830417M17 geneBG076225−4.4212849480.67375RIKEN cDNA 4432406C05 geneAV085137−6.0990530610.44504RIKEN cDNA 4631426G04 geneBG068677−4.6254594940.56033RIKEN cDNA 4632432J16 geneAV060454−4.6179583690.47517RIKEN cDNA 4633402N23 geneAA408693−5.5064786860.57523RIKEN cDNA 4833415N24 geneAV086029−4.3069725420.46627RIKEN cDNA 4833417L20 geneBG070225−4.1612970630.53534RIKEN cDNA 4930422J18 geneBG074133−6.5429372110.63785RIKEN cDNA 4930438D12 geneAV114186−5.7880467410.45307RIKEN cDNA 4930564D15 geneAW539497−6.1956797980.63818RIKEN cDNA 4933411H20 geneAV094491−10.132515780.23760RIKEN cDNA 4933436C10 geneAI854103−9.221855960.25555RIKEN cDNA 4933436C10 geneAV043801−7.1452760720.26851RIKEN cDNA 5430432N15 geneAV023999−5.1688974940.42754RIKEN cDNA 5730591C18 geneAV087450−4.2920041250.52004RIKEN cDNA 5830417I10 geneBG066100−4.2646975240.71856RIKEN cDNA 5830457J20 geneAV140522−5.8732340670.57518RIKEN cDNA 5830498C14 geneAV012853−10.643074720.44318RIKEN cDNA 5830498C14 geneBG066452−4.637100170.72557RIKEN cDNA 6030457N17 geneAV094720−11.179740020.47794RIKEN cDNA 6430411K18 geneAV023331−6.5582734850.55220RIKEN cDNA 6530416A09 geneBG071475−6.138039340.53936RIKEN cDNA 6720475J19 geneBG073712−13.955636010.24131RIKEN cDNA 6720475J19 geneBG073481−7.390815530.26541RIKEN cDNA 9030421L11 geneBG075528−4.6283272460.54551RIKEN cDNA 9130012G04 geneBG073930−6.6934640960.54126RIKEN cDNA A930018B01 geneAV073463−4.816295010.73761RIKEN cDNA E130105L11 geneBG075577−5.9600517730.51388ring finger protein 11AV084728−4.2275408190.54992ring-box 1AV053017−5.3636843950.58013RNA polymerase 1-3 (16 kDa subunit)AV134053−4.4799152580.59561S100 calcium binding protein A1AV003587−4.7955633560.51956sacsinAV013617−4.7052496870.67220S-adenosylmethionine decarboxylase 1BG075459−6.5750721230.38803SEC61, gamma subunit (S. cerevisiae)AV133876−4.8854889370.76946secretory carrier membrane protein 3AV094492−4.9792513120.43904serine/threonine kinase 23AA170153−4.1856109130.46751serine/threonine kinase 25 (yeast)AA146115−6.4216996690.54596serologically defined colon cancer antigen 28BG065578−12.464094540.18651serum response factorAV014460−4.1797896290.60298signal recognition particle 14 kDa (homologous Alu RNA binding proteitext missing or illegible when filedAV005775−7.1227521780.78602small inducible cytokine A11BE137080−4.7539392590.43931small proline rich-like 7AV072477−4.1433987820.31871soggy 1AV087775−4.597256950.41376solute carrier family 1, member 7AV006313−9.0072628270.54179solute carrier family 16 (monocarboxylic acid transporters), member 2AA199215−4.2484247230.57730solute carrier family 25 (mitochondrial carrier; adenine nucleotide transtext missing or illegible when filedAV087780−4.5011009770.35837solute carrier family 25 (mitochondrial carrier; oxoglutarate carrier), metext missing or illegible when filedAV094940−7.9802025560.45584solute carrier family 27 (fatty acid transporter), member 2AA154831−6.1288824840.52385Son cell proliferation proteinBG071049−6.0364726230.57640sortilin-related receptor, LDLR class A repeats-containingAA673962−4.8412537470.44436special AT-rich sequence binding protein 1BG065579−6.0421976120.44733spermine synthaseAV113836−4.9157707220.55802sphingomyelin phosphodiesterase 2, neutralBG063429−4.5889225410.53816split hand/foot deleted gene 1AV134049−4.6467555880.56217steroid 5 alpha-reductase 2-likeAV084563−10.289266780.46589sterol carrier protein 2, liverAA146030−5.0557730430.61558succinate-Coenzyme A ligase, GDP-forming, beta subunitAV087975−4.4011537240.54934superoxide dismutase 1, solubleBG074045−4.7754997060.57536suppressor of initiator codon mutations, related sequence 1 (S. cerevistext missing or illegible when filedAV042274−5.8929462240.47109surfactant associated protein AAV024739−6.3127554630.44949synaptobrevin like 1AV113528−11.352306570.48532TAR (HIV) RNA binding protein 2BG069749−4.4795924690.60506T-box 5AA198841−5.9298929330.50092T-cell receptor beta, variable 13AV015100−5.5677299810.54115TGF-beta1-induced anti-apoptotic factor 1AV078541−5.0480082930.68665thioredoxin 2AA116866−4.641109010.58741thioredoxin-like (32 kD)AV070815−4.5719511130.54871thioredoxin-like 2AV016790−5.5616217440.50942thyroid hormone receptor interactor 13AV094724−4.6032036650.52873tight junction protein 1BG073399−7.5258776990.67799tissue inhibitor of metalloproteinase 3NM_011595−7.5571595130.56285transcription elongation factor A (Sll), 3AI322966−4.1598416460.34762transducer of ERBB2, 2BG074926−5.9870305430.45199transforming growth factor beta 1 induced transcript 4AV140519−4.6168594270.74969transforming growth factor, beta 1AA049522−8.019042040.45450tubulointerstitial nephritis antigenAV066552−4.6356665710.61805tumor differentially expressed 1, likeAV083974−4.201553290.64214tumor necrosis factor (ligand) superfamily, member 10U37522−7.1594681260.44011tumor necrosis factor receptor superfamily, member 19BG072211−4.1406576890.34852tumor necrosis factor, alpha-induced protein 3AA572306−4.1331441050.60638ubiquitin-conjugating enzyme E2B, RAD6 homology (S. cerevisiae)AV095421−4.6597077340.55089ubiquitin-like 3BG072313−4.138142740.55812Unsequenced EST413125−8.225614450.22295Unsequenced EST412659−8.8706178690.24426Unsequenced EST432064−10.136531210.26718Unsequenced EST410956−4.8183744820.26969Unsequenced EST410595−5.4307469490.29232Unsequenced EST431252−5.0303121990.29553Unsequenced EST411369−8.607776060.29715Unsequenced EST413333−4.281970170.32070Unsequenced EST413297−6.3333088670.33170Unsequenced EST411987−4.707423130.33375Unsequenced EST411660−8.2291049280.33965Unsequenced EST411054−5.2076505740.34062Unsequenced EST410682−5.2746335090.34330Unsequenced EST431081−5.5464097050.34658Unsequenced EST206294−4.1816521870.35033Unsequenced EST412975−5.6056408950.35576Unsequenced EST432689−5.972814530.35787Unsequenced EST411277−11.088977280.35956Unsequenced EST412922−10.702368420.36608Unsequenced EST431286−4.7731510930.36615Unsequenced EST410681−5.5396788260.36806Unsequenced EST410961−5.9220867560.36889Unsequenced EST412082−5.5022686590.37358Unsequenced EST411260−7.3185219130.37963Unsequenced EST413169−8.8248038660.38149Unsequenced EST431574−7.9151880190.38774Unsequenced EST201627−4.7055335760.39533Unsequenced EST411524−5.5240623070.39648Unsequenced EST207603−4.3550504070.39946Unsequenced EST411380−7.3054632360.40609Unsequenced EST412118−5.5563476550.40838Unsequenced EST412779−5.4415540430.40976Unsequenced EST413183−4.1932289010.41145Unsequenced EST412186−5.0147101770.41232Unsequenced EST412432−6.0213079480.41525Unsequenced EST202131−4.5288952910.42149Unsequenced EST411977−5.5522861220.42892Unsequenced EST411945−5.196329950.43045Unsequenced EST412392−5.2590132950.43294Unsequenced EST411789−5.9424334910.43374Unsequenced EST411605−4.3411176070.43784Unsequenced EST412744−7.3395922030.43951Unsequenced EST413539−4.9899343440.44370Unsequenced EST195728−6.1784923220.44536Unsequenced EST413134−6.2418851030.45027Unsequenced EST411383−5.4013539820.45800Unsequenced EST411085−4.1379432140.46202Unsequenced EST412790−4.9417947160.46286Unsequenced EST412128−4.1732378720.46629Unsequenced EST412515−4.3028373380.47046Unsequenced EST411160−4.399053730.47073Unsequenced EST431843−4.9158992110.47188Unsequenced EST412684−4.2412056380.47318Unsequenced EST412861−8.3411884530.47330Unsequenced EST412655−7.6545293410.47341Unsequenced EST412947−5.9874747050.47730Unsequenced EST431845−6.5890365320.47756Unsequenced EST412605−4.5454997570.47830Unsequenced EST412852−5.6662950820.48040Unsequenced EST412719−6.4362862150.48313Unsequenced EST412846−6.3796012480.48331Unsequenced EST411516−4.1862797480.48381Unsequenced EST430640−8.5437453580.48480Unsequenced EST413600−4.9013988440.48861Unsequenced EST410665−5.2445861190.48898Unsequenced EST412580−4.1210773740.49239Unsequenced EST412961−6.8838438510.49284Unsequenced EST410750−4.493364130.49891Unsequenced EST413575−8.0927139790.49917Unsequenced EST412258−4.8512816710.50038Unsequenced EST413527−5.1324684620.50202Unsequenced EST339227−5.0397958970.50472Unsequenced EST412794−4.9904106090.50493Unsequenced EST413170−4.5352806620.50708Unsequenced EST412554−5.4508415310.51085Unsequenced EST411061−4.7695423330.51494Unsequenced EST413191−4.2604931590.51664Unsequenced EST411529−4.1466715020.51863Unsequenced EST201438−5.6864983840.51877Unsequenced EST412188−5.8287688510.53010Unsequenced EST412687−4.2716650880.53249Unsequenced EST411735−4.4684624060.53596Unsequenced EST432195−4.3358452880.53607Unsequenced EST431862−6.1656606750.54297Unsequenced EST431724−4.3385536810.54756Unsequenced EST202908−5.4183946720.54969Unsequenced EST413323−4.1842456110.55110Unsequenced EST411704−5.0960462240.55200Unsequenced EST412581−5.2697374260.55208Unsequenced EST412585−4.6599181230.55273Unsequenced EST431810−4.1807488370.55450Unsequenced EST413365−4.26598710.55605Unsequenced EST433229−4.5172548930.56214Unsequenced EST411979−4.3461599530.56235Unsequenced EST413165−4.629510730.56443Unsequenced EST192693−5.0433468850.56552Unsequenced EST431411−4.2133345630.56581Unsequenced EST413343−4.8586675560.56811Unsequenced EST431024−4.5305577130.57100Unsequenced EST411004−5.5852633240.57150Unsequenced EST412778−4.9584573150.57369Unsequenced EST411679−4.3976948180.57591Unsequenced EST412092−4.6011712470.57736Unsequenced EST411187−5.4204042340.57748Unsequenced EST412049−4.1824549710.57918Unsequenced EST411739−5.2616879860.57938Unsequenced EST412792−5.8004930520.58184Unsequenced EST430792−4.2810874780.58252Unsequenced EST412248−6.655901850.58382Unsequenced EST411820−5.9406180830.58997Unsequenced EST412944−5.4702730050.59317Unsequenced EST413551−4.5822489710.59406Unsequenced EST411432−20.536978740.59957Unsequenced EST410575−5.3030846840.60532Unsequenced EST412300−4.8187065280.61404Unsequenced EST413127−4.2688796290.61420Unsequenced EST413147−4.8343869050.61435Unsequenced EST431502−4.6104707530.61626Unsequenced EST412669−6.7223695220.62754Unsequenced EST205043−4.4925341740.62848Unsequenced EST411951−4.2411511870.63106Unsequenced EST410855−7.4112669030.63325Unsequenced EST431873−4.3818285320.64516Unsequenced EST413577−4.1174831050.64824Unsequenced EST412322−5.0508006130.65809Unsequenced EST431604−4.6527212140.65891Unsequenced EST410853−5.9064985210.67231Unsequenced EST410873−5.0139766860.68258Unsequenced EST411493−5.3385238820.68321Unsequenced EST411809−4.7993645950.70861Unsequenced EST431869−5.0195253020.70973Unsequenced EST410832−4.9769673690.72665Unsequenced EST413270−4.3431677880.75177upregulated during skeletal muscle growth 5AV088589−4.4469829850.45597vesicle-associated membrane protein 2AW911135−4.740288830.67738vesicle-associated membrane protein 3AV085364−4.4336575690.34943voltage-dependent anion channel 1BG073650−4.5302369830.55543wingless-related MMTV integration site 3AAA000971−5.5455104010.58208Y box protein 2BG066570−4.5682467960.43028Yamaguchi sarcoma viral (v-yes-1) oncogene homologAA509398−4.2245961310.55530zinc finger protein 106AV013127−4.3998134910.43000zinc finger protein 216BG066068−17.411083930.55649












TABLE IA








Gene Name
Gene Description
UGRepAcc [A]
LLRepProtActext missing or illegible when filed







AA068104
transforming growth factor, beta 2
NM_009367
NP_033393


AA098349
lysyl oxidase-like
AK078512


AA498724
bone morphogenetic protein 4
NM_007554
NP_031580


AA646363
endoglin
NM_007932
NP_031958


AI323974
neuropilin
NM_008737
NP_032763


AI327133
polydomain protein
NM_022814
NP_073725


AI841353
a disintegrin and metalloproteinase domain 15 (metar
NM_009614
NP_033744


AV012617
insulin-like growth factor binding protein 5
NM_010518
NP_034648


AV015188
matrix metalloproteinase 23
NM_011985
NP_036115


AV019210
elastin
NM_007925
NP_031951


AV021712
secreted frizzled-related sequence protein 2
NM_009144
NP_033170


AV024396
reversion-inducing-cysteine-rich protein with kazal mtext missing or illegible when filed
NM_016678
NP_057887


AV029310
superoxide dismutase 3, extracellular
NM_011435
NP_035565


AV059520
peptidylprolyl isomerase C-associated protein
NM_011150
NP_035280


AV070218
amyloid beta (A4) precursor-like protein 2
NM_009691
NP_033821


AV070419
antigen identified by monoclonal antibody MRC OX-2
NM_010818
NP_034948


AV083867
retinoid-inducible serine caroboxypetidase
NM_029023
NP_083299


AV084876
osteoblast specific factor 2 (fasciclin I-like)
NM_015784
NP_056599


AV085019
extracellular matrix protein 1
NM_007899
NP_031925


AV104097
basigin
BI106083


AV104213
endothelial cell-selective adhesion molecule
NM_027102
NP_081378


AV109513
stromal cell derived factor 1
NM_013655
NP_068350


AV113097
microfibrillar associated protein 5
NM_015776
NP_056591


AV117035
manic fringe homolog (Drosophila)
NM_008595
NP_032621


AV149987
cystatin C
NM_009976
NP_034106


AV156534
matrilin 2
NM_016762
NP_058042


AV170826
biglycan
NM_007542
NP_031568


AW476537
fibroblast growth factor receptor 1
NM_010206
NP_034336


AW988741
secreted acidic cysteine rich glycoprotein


BE376968
vascular endothelial growth factor C
NM_009506
NP_033532


BF136770
Notch gene homolog 3, (Drosophila)
NM_008716
NP_032742


BG063294
follistatin-like 3
NM_031380
NP_113557


BG063616
nidogen 1
NM_010917
NP_035047


BG064180
expressed sequence AA408225
NM_009868
NP_033998


BG065640
ectonucleotide pyrophosphatase/phosphodiesterase
NM_008813
NP_032839


BG066563
N-acetylated alpha-linked acidic dipeptidase 2
NM_028279
NP_082555


BG073227
fibulin 2
NM_007992
NP_032018


BG074344
mesothelin
NM_018857
NP_061345


BG074382
sema domain, immunoglobulin domain (Ig), short bastext missing or illegible when filed
NM_011349
NP_035479


BG074663
protein tyrosine phosphatase, receptor type, S
NM_011218
NP_035348


BG075377
melanoma cell adhesion molecule
NM_023061
NP_075548


D16250
bone morphogenetic protein receptor, type 1A
BC042611
NP_033888


L26349
tumor necrosis factor receptor superfamily, member 1
NM_011609
NP_035739


U38261
superoxide dismutase 3, extracellular
NM_011435
NP_035565


X52886
cathepsin D
NM_009983
NP_034113


AI838311
matrix metalloproteinase 2
NM_008610
NP_032636


AI851067
RIKEN cDNA 2510010F10 gene
NM_175833
NP_787027


BG071948
low density lipoprotein receptor-related protein 1
NM_008512
NP_032538


BG072998
expressed sequence AU018638
NM_008524
NP_032550


AI838613
epithelial membrane protein 1


AI893233
CD34 antigen
NM_133654
NP_598415


AV001464
granulin
NM_008175
NP_032201


AV006514
interferon (alpha and beta) receptor 2
NM_010509
NP_034639


AV022379
serine (or cysteine) proteinase inhibitor, clade F (alphtext missing or illegible when filed
NM_011340
NP_035470


AV025941
aquaporin 1
NM_007472
NP_031498


AV070805
thymic stromal-derived lymphopoietin, receptor
NM_016715
NP_057924


AV223941
heat shock protein, 70 kDa 3
M12571


AW537378
EST


AA673390
fibronectin 1
AK090130


AI325851
CD97 antigen
NM_011925
NP_036055


AI325886
neuroblastoma, suppression of tumorigenicity 1
NM_008675
NP_032701


AI385650
sialyltransferase 4C (beta-galactosidase alpha-2,3-sitext missing or illegible when filed
NM_009178
NP_033204


AI838302
Cd63 antigen
NM_007653
NP_031679


AI838568
RIKEN cDNA 1300018J16 gene
NM_029092
NP_083368


AV007183
latent transforming growth factor beta binding protein
NM_023912
NP_076401


AV007276
RIKEN cDNA 1110003M08 gene
AK090329


AV009300
procollagen, type IV, alpha 1
J04694


AV010312
procollagen, type IV, alpha 2
J04695


AV011166
EST
NM_080463
NP_536711


AV013988
procollagen, type VI, alpha 1
NM_009933
NP_034063


AV015595
procollagen, type XV
NM_009928
NP_034058


AV016743
RIKEN cDNA 5730414C17 gene
NM_133680
NP_598441


AV025665
prostaglandin-endoperoxide synthase 2
NM_011198
NP_035328


AV036454_text missing or illegible when filed
lymphocyte antigen 6 complex, locus E


AV037769
expressed sequence AU022549
NM_007904
NP_031930


AV048780
stromal cell derived factor 4
NM_011341
NP_035471


AV050682
RIKEN cDNA 2700083B06 gene
NM_026531
NP_080807


AV052090
serine (or cysteine) proteinase inhibitor, clade I (neurtext missing or illegible when filed
NM_009250
NP_033276


AV053955
RIKEN cDNA 3110023E09 gene
NM_026522
NP_080798


AV057827
torsin family 3, member A
NM_023141
NP_075630


AV058250
RIKEN cDNA 1810049K24 gene
NM_030209
NP_084485


AV059445
FK506 binding protein 9
NM_012056
NP_036186


AV059924
expressed sequence AA986889
NM_134102
NP_598863


AV061081
neural proliferation, differentiation and control gene 1
NM_008721
NP_032747


AV062071
CD24a antigen
NM_009846
NP_033976


AV066211
ELAV (embryonic lethal, abnormal vision, Drosophila)
NM_010485
NP_034615


AV073997
glucose regulated protein, 58 kDa
NM_007952
NP_031978


AV083352
RIKEN cDNA 1110007F23 gene
NM_029568
NP_083844


AV084561
procollagen C-proteinase enhancer protein
NM_008788
NP_032814


AV084844
immunoglobulin superfamily containing leucine-rich rtext missing or illegible when filed
NM_012043
NP_036173


AV086002
FXYD domain-containing ion transport regulator 6
NM_022004
NP_071287


AV087039
EST
NM_008885
NP_032911


AV087220
expressed sequence AW146116
NM_133352
NP_835359


AV087499
EST, Moderately similar to A57474 extracellular matri
NM_007899
NP_031925


AV087921
benzodiazepine receptor, peripheral
NM_009775
NP_033905


AV089105
calcium binding protein, intestinal
NM_009787
NP_033917


AV093463
serine (or cysteine) proteinase inhibitor, clade H (heatext missing or illegible when filed
NM_009825
NP_033955


AV094498
milk fat globule-EGF factor 8 protein
NM_008594
NP_032620


AV103290
expressed sequence AL024047
NM_134151
NP_598912


AV104157
dolichyl-di-phosphooligosaccharide-protein glycotrans
NM_007838
NP_031864


AV109555
cellular retinoic acid binding protein I
AK090130


AV111526
RIKEN cDNA 2610002H11 gene
NM_133721
NP_598482


AV112983
platelet derived growth factor receptor, beta polypeptitext missing or illegible when filed
NM_008809
NP_032835


AV133755
RIKEN cDNA 2810002E22 gene
NM_133859
NP_598620


AV134035
granulin
NM_008175
NP_032201


AV140189
RIKEN cDNA 0610040B21 gene
NM_025334
NP_079610


AV140901
EST
NM_010368
NP_034498


AV162270
lymphocyte antigen 6 complex, locus A
NM_027015
NP_081291


AV171867
CD 81 antigen
NM_133655
NP_598416


AW548258
procollagen-proline, 2-oxoglutarate 4-dioxygenase (ptext missing or illegible when filed
BC009654


AW551778
heterogeneous nuclear ribonucleoprotein C
NM_016884
NP_058580


BF100414
integrin beta 5
NM_010580
NP_034710


BF182158
Notch gene homolog 1, (Drosophila)
NM_008714
NP_032740


BG063167
adenylate cyclase 7
NM_007406
NP_031432


BG065103
lymphocyte antigen 6 complex, locus E
NM_008529
NP_032555


BG066621

Mus musculus, Similar to pituitary tumor-transforming

NM_145925
NP_666037


BG067569
coagulation factor II (thrombin) receptor
NM_010169
NP_034299


BG069745
proline arginine-rich end leucine-rich repeat
NM_054077
NP_473418


BG070083
protein tyrosine phosphatase, receptor type, E
NM_011212
NP_035342


BG070387
interleukin 6 signal transducer
NM_010560
NP_034690


BG072624
laminin, gamma 1
BC032194
NP_034813


BG072810
Niemann Pick type 02
NM_023409
NP_075898


BG072850
sarcoglycan, epsilon
NM_011360
NP_035490


BG072908
membrane-bound transcription factor protease, site 1
NM_019709
NP_062683


BG073140
CD8 antigen, beta chain
NM_009858
NP_033988


BG073341
retinal short-chain dehydrogenase/reductase 1
NM_011303
NP_035433


BG073479
expressed sequence AW229038
NM_133918
NP_598679


BG073729
prolyl 4-hydroxylase, beta polypeptide
J05185


BG073750
prolyl 4-hydroxylase, beta polypeptide
J05185


BG074142
RIKEN cDNA 1300012G16 gene
NM_023625
NP_076114


BG074174
DNA segment, Chr 6, Wayne State University 176 etext missing or illegible when filed
NM_138587
NP_613053


BG074422
integrin beta 1 (fibronectin receptor beta)
AK088016


BG074747
alpha glucosidase 2, alpha neutral subunit
NM_008060
NP_032086


BG074915
parotid secretory protein
NM_172261
NP_758465


BG075864
procollagen, type VI, alpha 2
NM_146007
NP_666119


C79946
expressed sequence C79946
AK080023


U20156
EST


U34920
ATP-binding cassette, sub-family G (WHITE), membetext missing or illegible when filed
NM_009593
NP_033723


X00246
histocompatibility 2, D region locus 1
NM_010380
NP_034510


X01838
beta-2 microglobulin
NM_009735
NP_033865


AA087526
retinol binding protein 1, cellular
NM_011254
NP_035384


AI322274
RIKEN cDNA 2410002J21 gene
AK033091


AI851039
ESTs, Weakly similar to D2045.2.p [Caenorhabditis etext missing or illegible when filed
AK038775


AV015246
RIKEN cDNA 1110054M18 gene
NM_175132
NP_780341


AV057141
gap junction membrane channel protein beta 1
NM_008124
NP_032150


AV059438
ets variant gene 6 (TEL oncogene)
BC009120


AV077899
actin, alpha 2, smooth muscle, aorta
AK002886


AV083262
dystonin
NM_134448
NP_604443


AV083596
four and a half LIM domains 1
NM_010211
NP_034341


AV085874

Mus musculus uridindiphosphoglucosepyrophosphortext missing or illegible when filed

NM_139297
NP_647458


AV093704
small EDRK-rich factor 2
AK044479


AW547864
EST


BG065584

Mus musculus, clone IMAGE: 3589087, mRNA, partiatext missing or illegible when filed

BF124761


BG070007
expressed sequence AW494241
BC040467


BG072752
actin, gamma, cytoplasmic
NM_013798
NP_038826


BG073284
prion protein dublet
NM_023043
NP_075530


BG073319
integrin beta 4 binding protein
NM_010579
NP_034709



















TABLE IB








Gene Name
Gene Description
UGRepAcc [A]
LLRepProtAcc [A]







AA068104
transforming growth factor, beta 2
NM_009367
NP_033393


AA098349
lysyl oxidase-like
AK078512


AA498724
bone morphogenetic protein 4
NM_007554
NP_031580


AA646363
endoglin
NM_007932
NP_031958


AI323974
neuropilin
NM_008737
NP_032763


AI327133
polydomain protein
NM_022814
NP_073725


AI841353
a disintegrin and metalloproteinase domain 15 (mettext missing or illegible when filed
NM_009614
NP_033744


AV012617
insulin-like growth factor binding protein 5
NM_010518
NP_034648


AV015188
matrix metalloproteinase 23
NM_011985
NP_036115


AV019210
elastin
NM_007925
NP_031951


AV021712
secreted frizzled-related sequence protein 2
NM_009144
NP_033170


AV024396
reversion-inducing-cysteine-rich protein with kazal ntext missing or illegible when filed
NM_016678
NP_057887


AV029310
superoxide dismutase 3, extracellular
NM_011435
NP_035565


AV059520
peptidylprolyl isomerase C-associated protein
NM_011150
NP_035280


AV070218
amyloid beta (A4) precursor-like protein 2
NM_009691
NP_033821


AV070419
antigen identified by monoclonal antibody MRC OX-text missing or illegible when filed
NM_010818
NP_034948


AV083867
retinoid-inducible serine caroboxypetidase
NM_029023
NP_083299


AV084876
osteoblast specific factor 2 (fasciclin I-like)
NM_015784
NP_056599


AV085019
extracellular matrix protein 1
NM_007899
NP_031925


AV104097
basigin
BI106083


AV104213
endothelial cell-selective adhesion molecule
NM_027102
NP_081378


AV109513
stromal cell derived factor 1
NM_013655
NP_068350


AV113097
microfibrillar associated protein 5
NM_015776
NP_056591


AV117035
manic fringe homolog (Drosophila)
NM_008595
NP_032621


AV149987
cystatin C
NM_009976
NP_034106


AV156534
matrilin 2
NM_016762
NP_058042


AV170826
biglycan
NM_007542
NP_031568


AW476537
fibroblast growth factor receptor 1
NM_010206
NP_034336


AW988741——2
secreted acidic cysteine rich glycoprotein


BE376968
vascular endothelial growth factor C
NM_009506
NP_033532


BF136770
Notch gene homolog 3, (Drosophila)
NM_008716
NP_032742


BG063294
follistatin-like 3
NM_031380
NP_113557


BG063616
nidogen 1
NM_010917
NP_035047


BG064180
expressed sequence AA408225
NM_009868
NP_033998


BG065640
ectonucleotide pyrophosphatase/phosphodiesterase
NM_008813
NP_032839


BG066563
N-acetylated alpha-linked acidic dipeptidase 2
NM_028279
NP_082555


BG073227
fibulin 2
NM_007992
NP_032018


BG074344
mesothelin
NM_018857
NP_061345


BG074382
sema domain, immunoglobulin domain (Ig), short btext missing or illegible when filed
NM_011349
NP_035479


BG074663
protein tyrosine phosphatase, receptor type, S
NM_011218
NP_035348


BG075377
melanoma cell adhesion molecule
NM_023061
NP_075548


D16250
bone morphogenetic protein receptor, type 1A
BC042611
NP_033888


L26349
tumor necrosis factor receptor superfamily, membertext missing or illegible when filed
NM_011609
NP_035739


U38261
superoxide dismutase 3, extracellular
NM_011435
NP_035565


X52886
cathepsin D
NM_009983
NP_034113


AI838311
matrix metalloproteinase 2
NM_008610
NP_032636


AI851067
RIKEN cDNA 2510010F10 gene
NM_175833
NP_787027


BG071948
low density lipoprotein receptor-related protein 1
NM_008512
NP_032538


BG072998
expressed sequence AU018638
NM_008524
NP_032550


AI838613
epithelial membrane protein 1


AI893233
CD34 antigen
NM_133654
NP_598415


AV001464
granulin
NM_008175
NP_032201


AV006514
interferon (alpha and beta) receptor 2
NM_010509
NP_034639


AV022379
serine (or cysteine) proteinase inhibitor, clade F (altext missing or illegible when filed
NM_011340
NP_035470


AV025941
aquaporin 1
NM_007472
NP_031498


AV070805
thymic stromal-derived lymphopoietin, receptor
NM_016715
NP_057924
















TABLE II










Table II Genes of Use in Imaging Studies - Membrane Associated


Annotated Extracellular and Antigen genes Upregulated in TAC tissues - 149 Unique genes


One example for each gene - Passed stringent SAM criteria








Mouse Gene Information
Human Homolog Information














Gene ID
Gene Description
UGRepAcc
LLRepProtAcc
Up TAC LA
Up TAC LV
UGRepAcc
LLRepProtAcc





BG073140
**CD8 antigen, beta chain
NM_009858
NP_033988
UP TAC LA





AI841353
a disintegrin and metalloproteinase domain
NM_009614
NP_033744
UP TAC LA

AY560601
NP_997080



15 (metargidin)


AV024684
A kinase (PRKA) anchor protein 2
NM_009649
NP_033779
UP TAC LA


AA797434
adenylate cyclase 7
NM_007406
NP_031432
UP TAC LA

D25538
NP_001105


AV103043
ADP-ribosylation factor 4
NM_007479
NP_031505
UP TAC LA

BC016325
NP_001651


AV032992
ADP-ribosylation-like factor 6 interacting
NM_022992
NP_075368
UP TAC LA



protein 5


AV057752
amyloid beta (A4) precursor protein
NM_007471
NP_031497
UP TAC LA

BC018937
NP_958817


AV104479
amyloid beta (A4) precursor protein-binding,
AK004792

UP TAC LA



family B, member 2


AV070218
amyloid beta (A4) precursor-like protein 2
NM_009691
NP_033821
UP TAC LA

BX647107
NP_001633


AV043404
angiotensin converting enzyme


UP TAC LA


AV025146
angiotensin receptor-like 1
NM_011784
NP_035914
UP TAC LA

AK075252
NP_005152


AV163403
antigen identified by monoclonal antibody
NM_010818
NP_034948
UP TAC LA

BC022522
NP_005935



MRC OX-2


AV025941
aquaporin 1
NM_007472
NP_031498
UP TAC LA

NM_198098
NP_932766


AV173744
ATPase, Cu++ transporting, alpha
NM_009726
NP_033856
UP TAC LA

NM_000052
NP_000043



polypeptide


AV031502
ATPase, H+ transporting, lysosomal 70 kD,
BI100125

UP TAC LA

AK023063
NP_006326



V1 subunit A, isoform 1text missing or illegible when filed


U34920
ATP-binding cassette, sub-family G
NM_009593
NP_033723
UP TAC LA

NM_207630
NP_997513



(WHITE), member 1


BG064525
basigin
BI106083

UP TAC LA

NM_001728
NP_940993


AV104535
beclin 1 (coiled-coil, myosin-like
NM_026562
NP_080838
UP TAC LA



BCL2-interacting protein)


AV087921
benzodiazepine receptor, peripheral
NM_009775
NP_033905
UP TAC LA

BX537892
NP_009295


X01838
beta-2 microglobulin
NM_009735
NP_033865
UP TAC LA

AK022379
NP_004039


AV140458
biregional cell adhesion molecule-related/
NM_172506
NP_766094
UP TAC LA

NM_033254
NP_150279



down-regulated by oncogtext missing or illegible when filed


D16250
bone morphogenetic protein receptor,
BC042611
NP_033888
UP TAC LA

NM_004329
NP_004320



type 1A


BG065470
catenin beta
NM_177589
NP_808257
UP TAC LA


AV171867
CD 81 antigen
NM_133655
NP_598416
UP TAC LA

BM810055
NP_004347


AV062071
CD24a antigen
NM_009846
NP_033976
UP TAC LA


AI893233
CD34 antigen
NM_133654
NP_598415
UP TAC LA

BX640941
NP_001764


BG073167
Cd63 antigen
NM_007653
NP_031679
UP TAC LA

BM701371
NP_001771


AI325851
CD97 antigen
NM_011925
NP_036055
UP TAC LA

NM_078481
NP_510966


AV300841
chemokine (C—X—C) receptor 4


UP TAC LA

NM_003467
NP_003458


BG067569
coagulation factor II (thrombin) receptor
NM_010169
NP_034299
UP TAC LA

NM_001992
NP_001983


AV031224
coatomer protein complex, subunit gamma 1
NM_017477
NP_059505
UP TAC LA


AV147446
cytochrome P450, 2j6


UP TAC LA


AV037185
degenerative spermatocyte homolog
NM_007853
NP_031879
UP TAC LA

NM_003676
NP_659004



(Drosophila)


AV083741
DNA segment, Chr 8, Brigham & Women's
NM_026002
NP_080278
UP TAC LA



Genetics 1112 expresstext missing or illegible when filed


AV104157
dolichyl-di-phosphooligosaccharide-protein
NM_007838
NP_031864
UP TAC LA

NM_005216
NP_005207



glycotransferase


BG075775
downstream of tyrosine kinase 1
NM_010070
NP_034200
UP TAC LA

AK055944
NP_001372


BG065640
ectonucleotide pyrophosphatase/
NM_008813
NP_032839
UP TAC LA

NM_006208
NP_006199



phosphodiesterase 1


AV050518
elongation of very long chain fatty acids
NM_019422
NP_062295
UP TAC LA

NM_022821
NP_073732



(FEN1/Elo2, SUR4/Elo3, ytext missing or illegible when filed


AV140302
embigin
NM_010330
NP_034460
UP TAC LA


AV086531
endoglin
NM_007932
NP_031958
UP TAC LA

NM_000118
NP_000109


AV104213
endothelial cell-selective adhesion molecule
NM_027102
NP_081378
UP TAC LA


AI838613
epithelial membrane protein 1


UP TAC LA
UP TAC LV
NM_001423
NP_001414


AV087039
EST
NM_008885
NP_032911
UP TAC LA

NM_000304
NP_696997


AV087918
EST AA087124
AA896198

UP TAC LA

NM_001759
NP_001750


AV021942
ESTs, Weakly similar to ATPase, class 1,
AF156546

UP TAC LA

AB032963
NP_065185



member a; ATPase 8A2text missing or illegible when filed


AV016534
ESTs, Weakly similar to Y43F4B.7.p
NM_153170
NP_694810
UP TAC LA



[Caenorhabditis elegans] [C.etext missing or illegible when filed


AV113175
ETL1
NM_133222
NP_573485
UP TAC LA

AY358360


BG064180
expressed sequence AA408225
NM_009868
NP_033998
UP TAC LA

NM_001795
NP_001786


BG072659
expressed sequence AI316797
NM_080563
NP_542130
UP TAC LA

NM_014746
NP_055561


AV033704
expressed sequence AI504145
NM_028990
NP_083266
UP TAC LA


AV037769
expressed sequence AU022549
NM_007904
NP_031930
UP TAC LA

NM_000115
NP_003982


AV087220
expressed sequence AW146116
NM_133352
NP_835359
UP TAC LA


BG066820
expressed sequence C80501
NM_009320
NP_033346
UP TAC LA

NM_003043
NP_003034


AW476537
fibroblast growth factor receptor 1
NM_010206
NP_034336
UP TAC LA

BC018128
NP_075599


BG072676
FXYD domain-containing ion transport
NM_022004
NP_071287
UP TAC LA

AK092198
NP_071286



regulator 6


AI838468
gamma-aminobutyric acid (GABA-B)
NM_019439
NP_062312
UP TAC LA

AJ225028
NP_068705



receptor, 1


AV057141
gap junction membrane channel protein
NM_008124
NP_032150

UP TAC LV
BF570961
NP_000157



beta 1


BG067028
glycoprotein galactosyltransferase alpha 1, 3
NM_010283
NP_034413
UP TAC LA


AV033394
glycoprotein m6b
NM_023122
NP_075611
UP TAC LA

AK095657
NP_005269


AV085916
GPI-anchored membrane protein 1
BU611749

UP TAC LA


BG063447
guanine nucleotide binding protein, beta 1
NM_008142
NP_032168
UP TAC LA

AK123609
NP_002065


X00246
histocompatibility 2, D region locus 1
NM_010380
NP_034510
UP TAC LA


BG064733
HLS7-interacting protein kinase
NM_147201
NP_671734
UP TAC LA

AK122664
NP_037524


AV010401
integral membrane protein 2B
NM_008410
NP_032436
UP TAC LA

BX537657
NP_068839


AV078295
integrin alpha 6
NM_008397
NP_032423
UP TAC LA

X53586
NP_000201


BG074422
integrin beta 1 (fibronectin receptor beta)
AK088016

UP TAC LA

NM_002211
NP_596867


BF100414
integrin beta 5
NM_010580
NP_034710
UP TAC LA

AK091595
NP_002204


AV006514
interferon (alpha and beta) receptor 2
NM_010509
NP_034639
UP TAC LA

L41944
NP_997468


AV074586
interleukin 17 receptor
BC037587

UP TAC LA


BG070387
interleukin 6 signal transducer
NM_010560
NP_034690
UP TAC LA

BC071555
NP_786943


BG072624
laminin, gamma 1
BC032194
NP_034813
UP TAC LA

NM_002293
NP_002284


AV054666
leptin receptor
NM_175036
NP_778201
UP TAC LA


BG075361
low density lipoprotein receptor-related
NM_008512
NP_032538
UP TAC LA

NM_002332
NP_002323



protein 1


AV162270
lymphocyte antigen 6 complex, locus A
NM_027015
NP_081291
UP TAC LA


BG065103
lymphocyte antigen 6 complex, locus E
NM_008529
NP_032555
UP TAC LA

BF969813
NP_002337


AV117035
manic fringe homolog (Drosophila)
NM_008595
NP_032621
UP TAC LA

U94352
NP_002396


AV026219
mannosidase 1, alpha
NM_008548
NP_032574
UP TAC LA


BG075377
melanoma cell adhesion molecule
NM_023061
NP_075548
UP TAC LA

NM_006500
NP_006491


BG072908
membrane-bound transcription factor
NM_019709
NP_062683
UP TAC LA

NM_003791
NP_957720



protease, site 1


AV025927

Mus musculus, clone IMAGE: 5066061,

BC046959

UP TAC LA



mRNA, partial cds


AV057440

Mus musculus, clone MGC: 27672 IMAGE:

NM_144852
NP_659101
UP TAC LA

BC062565
NP_004164



4911158, mRNA, comptext missing or illegible when filed


BG066621

Mus musculus, Similar to pituitary

NM_145925
NP_666037
UP TAC LA



tumor-transforming 1 interactingtext missing or illegible when filed


BG064673

Mus musculus, Similar to xylosylprotein

NM_146045
NP_666157
UP TAC LA

AK022566
NP_009186



beta1,4-galactosyltransfertext missing or illegible when filed


BG072632
myeloid-associated differentiation marker
NM_016969
NP_058665
UP TAC LA

AF087882
NP_612382


BG072584
myristoylated alanine rich protein kinase
NM_008538
NP_032564
UP TAC LA

NM_002356
NP_002347



C substrate


BG066563
N-acetylated alpha-linked acidic
NM_028279
NP_082555
UP TAC LA
UP TAC LV
AK075390
NP_005458



dipeptidase 2


AV061081
neural proliferation, differentiation
NM_008721
NP_032747
UP TAC LA

AK054950
NP_056207



and control gene 1


BG074219
neuroblastoma ras oncogene
NM_010937
NP_035067
UP TAC LA

X02751
NP_002515


AI323974
neuropilin
NM_008737
NP_032763
UP TAC LA


BG063616
nidogen 1
NM_010917
NP_035047
UP TAC LA


BF182158
Notch gene homolog 1, (Drosophila)
NM_008714
NP_032740
UP TAC LA

NM_017617
NP_060087


BF136770
Notch gene homolog 3, (Drosophila)
NM_008716
NP_032742
UP TAC LA

NM_000435
NP_000426


AV145718
parathyroid hormone receptor
NM_011199
NP_035329
UP TAC LA

AF495723
NP_000307


AV059520
peptidylprolyl isomerase C-associated
NM_011150
NP_035280
UP TAC LA



protein


AV006019
phosphatidylinositol glycan, class Q
NM_011822
NP_035952
UP TAC LA

NM_004204
NP_683721


BG064035
phosphoprotein enriched in astrocytes 15
NM_008556
NP_035193
UP TAC LA

NM_003768
NP_003759


AV112983
platelet derived growth factor receptor,
NM_008809
NP_032835
UP TAC LA

BC032224
NP_002600



beta polypeptide


AV234882
polycystic kidney disease 1 homolog
NM_013630
NP_038658
UP TAC LA

L33243
NP_000287


AV009300
procollagen, type IV, alpha 1
J04694

UP TAC LA

NM_001845
NP_001836


BG074718
procollagen, type IV, alpha 2
J04695

UP TAC LA

NM_001846
NP_001837


AV025665
prostaglandin-endoperoxide synthase 2
NM_011198
NP_035328
UP TAC LA

NM_000963
NP_000954


BG067870
protein kinase C, delta
NM_011103
NP_035233
UP TAC LA

NM_006254
NP_997704


BG070083
protein tyrosine phosphatase, receptor
NM_011212
NP_035342
UP TAC LA

BX648180
NP_569119



type, E


BG074663
protein tyrosine phosphatase, receptor
NM_011218
NP_035348
UP TAC LA

NM_002850
NP_570925



type, S


AI893212
proteolipid protein 2
NM_019755
NP_062729
UP TAC LA

BF214130
NP_002659


BG073000
protocadherin 13
NM_033576
NP_291054
UP TAC LA


AV086128
regulator of G-protein signaling 19
NM_018771
NP_061241
UP TAC LA

NM_005716
NP_974223



interacting protein 1


AU040596
regulator of G-protein signaling 3
NM_019492
NP_062365
UP TAC LA

AK128127
NP_652760


AV084219
reticulon 4
NM_024226
NP_077188
UP TAC LA

NM_020532
NP_997404


BG073341
retinal short-chain dehydrogenase/
NM_011303
NP_035433
UP TAC LA

BX648476
NP_004744



reductase 1


AV024396
reversion-inducing-cysteine-rich
NM_016678
NP_057887
UP TAC LA

BX648668
NP_066934



protein with kazal motifs


BG063638
ribosome binding protein 1
AK019964
NP_598329
UP TAC LA

AB037819
NP_004578


AW538766
RIKEN cDNA 0610013I17 gene
NM_029789
NP_084065
UP TAC LA

NM_012432
NP_036564


AV133782
RIKEN cDNA 0610039A15 gene
NM_175101
NP_780310
UP TAC LA


AV007276
RIKEN cDNA 1110003M08 gene
AK090329

UP TAC LA

AK124975
NP_005818


AV058524
RIKEN cDNA 1110007A14 gene
NM_025841
NP_080117
UP TAC LA

AK093917
NP_006845


AV133706
RIKEN cDNA 1110059L23 gene
NM_134255
NP_599016
UP TAC LA

AL833001
NP_068586


AV086520
RIKEN cDNA 1200003O06 gene
NM_025813
NP_080089
UP TAC LA


BG064285
RIKEN cDNA 1200013F24 gene
NM_025822
NP_080098
UP TAC LA


AV088097
RIKEN cDNA 1200015A22 gene
NM_028766
NP_083042
UP TAC LA


BG074142
RIKEN cDNA 1300012G16 gene
NM_023625
NP_076114
UP TAC LA


AV086327
RIKEN cDNA 2310008D10 gene
NM_025858
NP_080657
UP TAC LA


AV087181
RIKEN cDNA 2310028N02 gene
NM_025864
NP_080140
UP TAC LA


AV085104
RIKEN cDNA 2410001H17 gene
NM_025889
NP_080165
UP TAC LA


BG067332
RIKEN cDNA 2610002H11 gene
NM_133721
NP_598482
UP TAC LA

BX647350
NP_002198


BG073064
RIKEN cDNA 2610027H02 gene
BC027791

UP TAC LA


AV061276
RIKEN cDNA 5031406P05 gene
NM_026669
NP_080945
UP TAC LA

AK130050
NP_003208


AV020551
RIKEN cDNA 5730403E06 gene
NM_027439
NP_081715
UP TAC LA


AV016743
RIKEN cDNA 5730414C17 gene
NM_133680
NP_598441
UP TAC LA


AV085966
RIKEN cDNA 6720474K14 gene
NM_175414
NP_780623
UP TAC LA


BG072850
sarcoglycan, epsilon
NM_011360
NP_035490
UP TAC LA

NM_003919
NP_003910


AV087531
scavenger receptor class B1
NM_016741
NP_058021
UP TAC LA

AK023485
NP_005496


AV021712
secreted frizzled-related sequence protein 2
NM_009144
NP_033170
UP TAC LA

NM_003013
NP_003004


AV062462
serine palmitoyltransferase, long chain
NM_009269
NP_033295
UP TAC LA

NM_006415
NP_847894



base subunit 1


D16106
sialyltransferase 1 (beta-galactoside
NM_145933
NP_666045
UP TAC LA



alpha-2,6-sialyltransferase)


AI385650
sialyltransferase 4C (beta-galactosidase
NM_009178
NP_033204
UP TAC LA

AK128605
NP_006269



alpha-2,3-sialytransferasetext missing or illegible when filed


AV093704
small EDRK-rich factor 2
AK044479


UP TAC LV


BG075739
solute carrier family 29 (nucleoside
NM_022880
NP_075018
UP TAC LA

AK090615
NP_004946



transporters), member 1


AA499432
sprouty homolog 4 (Drosophila)
NM_011898
NP_036028
UP TAC LA

AF227516
NP_112226


AV074505
surfeit gene 4
NM_011512
NP_035642
UP TAC LA

NM_033161
NP_149351


AV111434
transient receptor protein 2
BF583628

UP TAC LA

BM701565
NP_852667


AV083947
transmembrane domain protein regulated
NM_011906
NP_036036
UP TAC LA



in adipocytes 40 kDa


AA023493
transmembrane protein with EGF-like and
AK079633

UP TAC LA

NM_003692
NP_003683



two follistatin-like domaitext missing or illegible when filed


L26349
tumor necrosis factor receptor superfamily,
NM_011609
NP_035739
UP TAC LA

NM_001065
NP_001056



member 1a


AV024570
tumor necrosis factor, alpha-induced
NM_009395
NP_033421
UP TAC LA

BC003694
NP_066960



protein 1 (endothelial)


BG062994
UDP-GlcNAc: betaGal
NM_016888
NP_058584
UP TAC LA

BC047933
NP_150274



beta-1,3-N-acetylglucosaminyltransferase 1text missing or illegible when filed


BG073697
UDP-glucuronate decarboxylase 1
NM_026430
NP_080706
UP TAC LA

BC035177
NP_079352


BG064510
vanilloid receptor-like protein 1
NM_011706
NP_035836
UP TAC LA

AK126996
NP_057197


BE376968
vascular endothelial growth factor C
NM_009506
NP_033532
UP TAC LA

NM_005429
NP_005420


AV103195
zinc finger protein 36
NM_133786
NP_598547
UP TAC LA

NM_005496
NP_005487
















TABLE III










Table III Genes of Use in Serologic Assays and/or Imaging Studies


Annotated Extracellular and Antigen genes Upregulated in TAC tissues - 169 Unique genes


One example for each gene - Passed stringent SAM criteria









Human Homolog Information









Mouse Gene Information
Human
Human














Gene ID
Gene Description
UGRepAcc
LLReProtA
Up TAC LA
Up TAC LV
UGRepAtext missing or illegible when filed
LLReptext missing or illegible when filed





AI841353
a disintegrin and metalloproteinase
NM_009614
NP_033744
UP TAC LA

AY560601
NP_997080



domain 15 (metargidin)


AV077899
actin, alpha 2, smooth muscle, aorta
AK002886


UP TAC LV


BG072752
actin, gamma, cytoplasmic
NM_013798
NP_038826

UP TAC LV


BG063167
adenylate cyclase 7
NM_007406
NP_031432
UP TAC LA
UP TAC LV
D25538
NP_001105


BG074747
alpha glucosidase 2, alpha neutral
NM_008060
NP_032086
UP TAC LA



subunit


AV070218
amyloid beta (A4) precursor-like
NM_009691
NP_033821
UP TAC LA

BX647107
NP_001633



protein 2


AV070419
antigen identified by monoclonal
NM_010818
NP_034948
UP TAC LA

BC022522
NP_005935



antibody MRC OX-2


AV025941
aquaporin 1
NM_007472
NP_031498
UP TAC LA

NM_198098
NP_932766


U34920
ATP-binding cassette, sub-family G
NM_009593
NP_033723
UP TAC LA

NM_207630
NP_997513



(WHITE), member 1


AV104097
basigin
BI106083

UP TAC LA

NM_001728
NP_940993


AV087921
benzodiazepine receptor, peripheral
NM_009775
NP_033905
UP TAC LA

BX537892
NP_009295


X01838
beta-2 microglobulin
NM_009735
NP_033865
UP TAC LA

AK022379
NP_004039


AV170826
biglycan
NM_007542
NP_031568
UP TAC LA

BC004244
NP_001702


AA498724
bone morphogenetic protein 4
NM_007554
NP_031580
UP TAC LA

NM_001202
NP_570912


D16250
bone morphogenetic protein receptor,
BC042611
NP_033888
UP TAC LA

NM_004329
NP_004320



type 1A


AV089105
calcium binding protein, intestinal
NM_009787
NP_033917
UP TAC LA


X52886
cathepsin D
NM_009983
NP_034113
UP TAC LA

NM_001909
NP_001900


AV171867
CD 81 antigen
NM_133655
NP_598416
UP TAC LA

BM810055
NP_004347


AV062071
CD24a antigen
NM_009846
NP_033976
UP TAC LA


AI893233
CD34 antigen
NM_133654
NP_598415
UP TAC LA

BX640941
NP_001764


AI838302
Cd63 antigen
NM_007653
NP_031679
UP TAC LA

BM701371
NP_001771


BG073140
CD8 antigen, beta chain
NM_009858
NP_033988
UP TAC LA


AI325851
CD97 antigen
NM_011925
NP_036055
UP TAC LA

NM_078481
NP_510966


AV109555
cellular retinoic acid binding protein I
AK090130

UP TAC LA

NM_212482
NP_997647


BG067569
coagulation factor II (thrombin) receptor
NM_010169
NP_03429
UP TAC LA

NM_001992
NP_001983


AV149987
cystatin C
NM_009976
NP_034106
UP TAC LA

BX647523
NP_000090


BG074174
DNA segment, Chr 6, Wayne State
NM_138587
NP_613053
UP TAC LA



University 176, expressed


AV104157
dolichyl-di-phosphooligosaccharide-
NM_007838
NP_031864
UP TAC LA

NM_005216
NP_005207



protein glycotransferase


AV083262
dystonin
NM_134448
NP_604443

UP TAC LV
NM_183380
NP_899236


BG065640
ectonucleotide pyrophosphatase/
NM_008813
NP_032839
UP TAC LA

NM_006208
NP_006199



phosphodiesterase 1


AV019210
elastin
NM_007925
NP_031951
UP TAC LA

BX537939
NP_000492


AV066211
ELAV (embryonic lethal, abnormal
NM_010485
NP_034615
UP TAC LA

NM_001419
NP_001410



vision, Drosophila)-like 1 (Htext missing or illegible when filed


AA646363
endoglin
NM_007932
NP_031958
UP TAC LA

NM_000118
NP_000109


AV104213
endothelial cell-selective adhesion
NM_027102
NP_081378
UP TAC LA



molecule


AI838613
epithelial membrane protein 1


UP TAC LA
UP TAC LV
NM_001423
NP_001414


AV011166
EST
NM_080463
NP_536711
UP TAC LA

AF375884
NP_758436


AV087039
EST
NM_008885
NP_032911
UP TAC LA

NM_000304
NP_696997


AV140901
EST
NM_010368
NP_034498
UP TAC LA


AW537378
EST


SAM
UP TAC LV






DOWN


AW547864
EST



UP TAC LV


U20156
EST


UP TAC LA
UP TAC LV
BQ056329
NP_002406


AV087499
EST, Moderately similar to A57474
NM_007899
NP_031925
UP TAC LA

AK097205
NP_073155



extracellular matrix proteintext missing or illegible when filed


AI851039
ESTs, Weakly similar to D2045.2.p
AK038775


UP TAC LV



[Caenorhabditis elegans] [text missing or illegible when filed


AV059438
ets variant gene 6 (TEL oncogene)
BC009120


UP TAC LV


BG064180
expressed sequence AA408225
NM_009868
NP_033998
UP TAC LA

NM_001795
NP_001786


AV059924
expressed sequence AA986889
NM_134102
NP_598863
UP TAC LA

BX647516
NP_056984


AV103290
expressed sequence AL024047
NM_134151
NP_598912
UP TAC LA

AK125213
NP_003671


BG072998
expressed sequence AU018638
NM_008524
NP_032550

UP TAC LV
BG114678
NP_002336


AV037769
expressed sequence AU022549
NM_007904
NP_031930
UP TAC LA

NM_000115
NP_003982


AV087220
expressed sequence AW146116
NM_133352
NP_835359
UP TAC LA


BG073479
expressed sequence AW229038
NM_133918
NP_598679
UP TAC LA

AL050138
NP_008977


BG070007
expressed sequence AW494241
BC040467


UP TAC LV


C79946
expressed sequence C79946
AK080023

UP TAC LA
UP TAC LV


AV085019
extracellular matrix protein 1
NM_007899
NP_031925
UP TAC LA

AK097205
NP_073155


AW476537
fibroblast growth factor receptor 1
NM_010206
NP_034336
UP TAC LA

BC018128
NP_075599


AA673390
fibronectin 1
AK090130

UP TAC LA

NM_212482
NP_997647


BG073227
fibulin 2
NM_007992
NP_032018
UP TAC LA

AY130459
NP









001004019


AV059445
FK506 binding protein 9
NM_012056
NP_036186
UP TAC LA

AK075331
NP_009201


BG063294
follistatin-like 3
NM_031380
NP_113557
UP TAC LA

BC005839
NP_005851


AV083596
four and a half LIM domains 1
NM_010211
NP_034341

UP TAC LV
AK122708
NP_001440


AV086002
FXYD domain-containing ion
NM_022004
NP_071287
UP TAC LA

AK092198
NP_071286



transport regulator 6


AV057141
gap junction membrane channel
NM_008124
NP_032150

UP TAC LV
BF570961
NP_000157



protein beta 1


AV073997
glucose regulated protein, 58 kDa
NM_007952
NP_031978
UP TAC LA

AK075455
NP_005304


AV001464
granulin
NM_008175
NP_032201
UP TAC LA

NM_002087
NP_002078


AV134035
granulin
NM_008175
NP_032201
UP TAC LA

NM_002087
NP_002078


AV223941
heat shock protein, 70 kDa 3
M12571

SAM
UP TAC LV
NM_005345
NP_005336






DOWN


AW551778
heterogeneous nuclear
NM_016884
NP_058580
UP TAC LA
UP TAC LV
AK126950
NP_112604



ribonucleoprotein C


X00246
histocompatibility 2, D region locus 1
NM_010380
NP_034510
UP TAC LA


AV084844
immunoglobulin superfamily containing
NM_012043
NP_036173
UP TAC LA

NM_005545.3
NP_005536.1



leucine-rich repeat


AV012617
insulin-like growth factor binding
NM_010518
NP_034648
UP TAC LA

NM_000599
NP_000590



protein 5


BG074422
integrin beta 1 (fibronectin receptor
AK088016

UP TAC LA

NM_002211
NP_596867



beta)


BG073319
integrin beta 4 binding protein
NM_010579
NP_034709

UP TAC LV
BQ278496
NP_852134


BF100414
integrin beta 5
NM_010580
NP_034710
UP TAC LA

AK091595
NP_002204


AV006514
interferon (alpha and beta) receptor 2
NM_010509
NP_034639
UP TAC LA

L41944
NP_997468


BG070387
interleukin 6 signal transducer
NM_010560
NP_034690
UP TAC LA

BC071555
NP_786943


BG072624
laminin, gamma 1
BC032194
NP_034813
UP TAC LA

NM_002293
NP_002284


AV007183
latent transforming growth factor
NM_023912
NP_076401
UP TAC LA

AK024477
NP_066548



beta binding protein 3


BG071948
low density lipoprotein receptor-related
NM_008512
NP_032538

UP TAC LV
NM_002332
NP_002323



protein 1


AV162270
lymphocyte antigen 6 complex, locus A
NM_027015
NP_081291
UP TAC LA

NM_001030
NP_001021


BG065103
lymphocyte antigen 6 complex, locus E
NM_008529
NP_032555
UP TAC LA

BF969813
NP_002337


AA098349
lysyl oxidase-like
AK078512

UP TAC LA

BC068542
NP_005567


AV117035
manic fringe homolog (Drosophila)
NM_008595
NP_032621
UP TAC LA

U94352
NP_002396


AV156534
matrilin 2
NM_016762
NP_058042
UP TAC LA

BX648291
NP_085072


AI838311
matrix metalloproteinase 2
NM_008610
NP_032636

UP TAC LV
AL832088
NP_004521


AV015188
matrix metalloproteinase 23
NM_011985
NP_036115
UP TAC LA


BG075377
melanoma cell adhesion molecule
NM_023061
NP_075548
UP TAC LA

NM_006500
NP_006491


BG072908
membrane-bound transcription
NM_019709
NP_062683
UP TAC LA

NM_003791
NP_957720



factor protease, site 1


BG074344
mesothelin
NM_018857
NP_061345
UP TAC LA

BC003512
NP_037536


AV113097
microfibrillar associated
NM_015776
NP_056591
UP TAC LA

NM_003480
NP_003471



protein 5


AV094498
milk fat globule-EGF factor 8 protein
NM_008594
NP_032620
UP TAC LA

AK092157
NP_005919


AV085874

Mus musculus

NM_139297
NP_647458

UP TAC LV
BX537559
NP_006750



uridindiphosphoglucosepyrophosphorylase



2 (Utext missing or illegible when filed


BG065584

Mus musculus, clone IMAGE: 3589087,

BF124761


UP TAC LV



mRNA, partial cds


BG066621

Mus musculus, Similar to pituitary

NM_145925
NP_666037
UP TAC LA



tumor-transforming 1 interactext missing or illegible when filed


BG066563
N-acetylated alpha-linked acidic
NM_028279
NP_082555
UP TAC LA
UP TAC LV
AK075390
NP_005458



dipeptidase 2


AV061081
neural proliferation, differentiation
NM_008721
NP_032747
UP TAC LA

AK054950
NP_056207



and control gene 1


AI325886
neuroblastoma, suppression of
NM_008675
NP_032701
UP TAC LA

NM_182744
NP_877421



tumorigenicity 1


AI323974
neuropilin
NM_008737
NP_032763
UP TAC LA


BG063616
nidogen 1
NM_010917
NP_035047
UP TAC LA


BG072810
Niemann Pick type C2
NM_023409
NP_075898
UP TAC LA

BQ896617
NP_006423


BF182158
Notch gene homolog 1, (Drosophila)
NM_008714
NP_032740
UP TAC LA

NM_017617
NP_060087


BF136770
Notch gene homolog 3, (Drosophila)
NM_008716
NP_032742
UP TAC LA

NM_000435
NP_000426


AV084876
osteoblast specific factor 2
NM_015784
NP_056599
UP TAC LA



(fasciclin I-like)


BG074915
parotid secretory protein
NM_172261
NP_758465
UP TAC LA

AL713642
NP_115984


AV059520
peptidylprolyl isomerase C-associated
NM_011150
NP_035280
UP TAC LA



protein


AV112983
platelet derived growth factor
NM_008809
NP_032835
UP TAC LA

BC032224
NP_002600



receptor, beta polypeptide


AI327133
polydomain protein
NM_022814
NP_073725
UP TAC LA


BG073284
prion protein dublet
NM_023043
NP_075530

UP TAC LV
NM_012409
NP_036541


AV084561
procollagen C-proteinase enhancer protein
NM_008788
NP_032814
UP TAC LA
UP TAC LV
BM994449
NP_002584


AV009300
procollagen, type IV, alpha 1
J04694

UP TAC LA

NM_001845
NP_001836


AV010312
procollagen, type IV, alpha 2
J04695

UP TAC LA

NM_001846
NP_001837


AV013988
procollagen, type VI, alpha 1
NM_009933
NP_034063
UP TAC LA

NM_001848
NP_001839


BG075864
procollagen, type VI, alpha 2
NM_146007
NP_666119
UP TAC LA

AK128695
NP_478055


AV015595
procollagen, type XV
NM_009928
NP_034058
UP TAC LA

NM_001855
NP_001846


AW548258
procollagen-proline, 2-oxoglutarate
BC009654

UP TAC LA

BX648829
NP_000908



4-dioxygenase (proline 4-htext missing or illegible when filed


BG069745
proline arginine-rich end leucine-rich
NM_054077
NP_473418
UP TAC LA

NM_002725
NP_958505



repeat


BG073729
prolyl 4-hydroxylase, beta polypeptide
J05185

UP TAC LA

J02783
NP_000909


BG073750
prolyl 4-hydroxylase, beta polypeptide
J05185

UP TAC LA

J02783
NP_000909


AV025665
prostaglandin-endoperoxide synthase 2
NM_011198
NP_035328
UP TAC LA

NM_000963
NP_000954


BG070083
protein tyrosine phosphatase, receptor
NM_011212
NP_035342
UP TAC LA

BX648180
NP_569119



type, E


BG074663
protein tyrosine phosphatase, receptor
NM_011218
NP_035348
UP TAC LA

NM_002850
NP_570925



type, S


BG073341
retinal short-chain dehydrogenase/
NM_011303
NP_035433
UP TAC LA

BX648476
NP_004744



reductase 1


AV083867
retinoid-inducible serine caroboxypetidase
NM_029023
NP_083299
UP TAC LA


AA087526
retinol binding protein 1, cellular
NM_011254
NP_035384

UP TAC LV
BF508021
NP_002890


AV024396
reversion-inducing-cysteine-rich
NM_016678
NP_057887
UP TAC LA

BX648668
NP_066934



protein with kazal motifs


AV140189
RIKEN cDNA 0610040B21 gene
NM_025334
NP_079610
UP TAC LA


AV007276
RIKEN cDNA 1110003M08 gene
AK090329

UP TAC LA

AK124975
NP_005818


AV083352
RIKEN cDNA 1110007F23 gene
NM_029568
NP_083844
UP TAC LA


AV015246
RIKEN cDNA 1110054M18 gene
NM_175132
NP_780341

UP TAC LV


BG074142
RIKEN cDNA 1300012G16 gene
NM_023625
NP_076114
UP TAC LA


AI838568
RIKEN cDNA 1300018J16 gene
NM_029092
NP_083368
UP TAC LA
UP TAC LV


AV058250
RIKEN cDNA 1810049K24 gene
NM_030209
NP_084485
UP TAC LA


AI322274
RIKEN cDNA 2410002J21 gene
AK033091


UP TAC LV


AI851067
RIKEN cDNA 2510010F10 gene
NM_175833
NP_787027

UP TAC LV


AV111526
RIKEN cDNA 2610002H11 gene
NM_133721
NP_598482
UP TAC LA

BX647350
NP_002198


AV050682
RIKEN cDNA 2700083B06 gene
NM_026531
NP_080807
UP TAC LA
UP TAC LV


AV133755
RIKEN cDNA 2810002E22 gene
NM_133859
NP_598620
UP TAC LA


AV053955
RIKEN cDNA 3110023E09 gene
NM_026522
NP_080798
UP TAC LA


AV016743
RIKEN cDNA 5730414C17 gene
NM_133680
NP_598441
UP TAC LA


BG072850
sarcoglycan, epsilon
NM_011360
NP_035490
UP TAC LA

NM_003919
NP_003910


AW988741_2
secreted acidic cysteine rich glycoprotein


UP TAC LA

AK126525
NP_003109


AV021712
secreted frizzled-related sequence
NM_009144
NP_033170
UP TAC LA

NM_003013
NP_003004



protein 2


BG074382
sema domain, immunoglobulin domain
NM_011349
NP_035479
UP TAC LA

U38276
NP_004177



(Ig), short basic domaintext missing or illegible when filed


AV022379
serine (or cysteine) proteinase inhibitor,
NM_011340
NP_035470
UP TAC LA

BM918904
NP_002606



clade F (alpha-2 antipltext missing or illegible when filed


AV093463
serine (or cysteine) proteinase inhibitor,
NM_009825
NP_033955
UP TAC LA

AK122936
NP_001226



clade H (heat shock prtext missing or illegible when filed


AV052090
serine (or cysteine) proteinase inhibitor,
NM_009250
NP_033276
UP TAC LA

BC018043
NP_005016



clade I (neuroserpin), text missing or illegible when filed


AI385650
sialyltransferase 4C (beta-galactosidase
NM_009178
NP_033204
UP TAC LA

AK128605
NP_006269



alpha-2,3-sialytransfetext missing or illegible when filed


AV093704
small EDRK-rich factor 2
AK044479


UP TAC LV


AV109513
stromal cell derived factor 1
NM_013655
NP_068350
UP TAC LA

BX647204
NP_954637


AV048780
stromal cell derived factor 4
NM_011341
NP_035471
UP TAC LA


U38261
superoxide dismutase 3, extracellular
NM_011435
NP_035565
UP TAC LA

NM_003102
NP_003093


AV070805
thymic stromal-derived lymphopoietin,
NM_016715
NP_057924
UP TAC LA



receptor


AV057827
torsin family 3, member A
NM_023141
NP_075630
UP TAC LA

NM_022371
NP_071766


AA068104
transforming growth factor, beta 2
NM_009367
NP_033393
UP TAC LA

M19154
NP_003229


L26349
tumor necrosis factor receptor
NM_011609
NP_035739
UP TAC LA

NM_001065
NP_001056



superfamily, member 1a


BE376968
vascular endothelial growth factor C
NM_009506
NP_033532
UP TAC LA

NM_005429
NP_005420
















TABLE IV










Table IV Genes of Use in Metabolic Assays


Annotated Metabolism Genes Downregulated in TAC tissues - 109 Unique genes


One example for each gene - Passed stringent SAM criteria








Mouse Gene Information















Gene Name
Gene Description
UGRepAcc
LLRepProtA
Down TAC LA
Down TAC LV
UGRepAcc
LLRepProtAcc





BG066890
**DNA segment, Chr 13, ERATO
NM_007749
NP_031775
DOWN TAC LA

BI118114
NP_001858



Doi 332, expressed


BG062980
**DNA segment, Chr 2, Wayne State
U37501

DOWN TAC LA

NM_005560
NP_005551



University 85, expressed


AV025301
2,4-dienoyl CoA reductase 1,
NM_026172
NP_080448

DOWN TAC LV
BM920635
NP_001350



mitochondrial


AV029241
acetyl-Coenzyme A dehydrogenase,
NM_007381
NP_031407
DOWN TAC LA
DOWN TAC LV
BC039063
NP_001599



long-chain


AI840666
acetyl-Coenzyme A dehydrogenase,
NM_007382
NP_031408
DOWN TAC LA
DOWN TAC LV
NM_000016
NP_000007



medium chain


AV004604
acetyl-Coenzyme A dehydrogenase,
NM_007383
NP_031409

DOWN TAC LV
AK057021
NP_000008



short chain


AI839605
acyl-Coenzyme A dehydrogenase,
NM_017366
NP_059062
DOWN TAC LA

AK097243
NP_000009



very long chain


AF006688
acyl-Coenzyme A oxidase 1,
NM_015729
NP_056544

DOWN TAC LV
BC008767
NP_009223



palmitoyl


U07235
aldehyde dehydrogenase 2,
NM_009656
NP_033786

DOWN TAC LV
AL832043
NP_000681



mitochondrial


AV006235
ATPase, Ca++ transporting, cardiac
NM_009722
NP_033852

DOWN TAC LV
BX648282
NP_733765



muscle, slow twitch 2


BG074044
ATPase, Ca++ transporting, cardiac
NM_009722
NP_033852
DOWN TAC LA
DOWN TAC LV
BX648282
NP_733765



muscle, slow twitch 2


AI837797
ATPase, Ca++ transporting, cardiac
NM_009722
NP_033852
DOWN TAC LA

BX648282
NP_733765



muscle, slow twitch 2


AV095181
AU RNA binding protein/
NM_016709
NP_057918
DOWN TAC LA

AK124142
NP_001689



enoyl-coenzyme A hydratase


AI323918
branched chain ketoacid
NM_007533
NP_031559

DOWN TAC LV
BF206112
NP_000700



dehydrogenase E1, alpha polypeptidetext missing or illegible when filed


AV014385
carbonic anhydrase 14
NM_146104
NP_666216
DOWN TAC LA
DOWN TAC LV


AV170903
carbonic anhydrase 14
NM_146104
NP_666216

DOWN TAC LV


AI323923
carbonyl reductase 1
NM_007620
NP_031646
DOWN TAC LA

BM810059
NP_001748


AV006197
carnitine palmitoyltransferase 2
NM_009949
NP_034079
DOWN TAC LA
DOWN TAC LV
NM_000098
NP_000089


AV093569
copper chaperone for superoxide
NM_016892
NP_058588
DOWN TAC LA

BM543741
NP_005116



dismutase


AV085004
creatine kinase, mitochondrial 2
AK009042

DOWN TAC LA

NM_001825
NP_001816


AV005997
cytochrome c oxidase, subunit IVa
NM_009941
NP_034071
DOWN TAC LA

AK027136
NP_001852


AV095075
cytochrome c oxidase, subunit Va
NM_007747
NP_031773

DOWN TAC LV
BM911641
NP_004246


AV088644
cytochrome c oxidase, subunit Vb
NM_009942
NP_034072
DOWN TAC LA

BM912880
NP_001853


AV001082
cytochrome c oxidase, subunit
NM_009943
NP_034073
DOWN TAC LA
DOWN TAC LV
BM712970
NP_005196



VI a, polypeptide 2


AV149855
cytochrome c oxidase, subunit VIc
NM_053071
NP_444301
DOWN TAC LA
DOWN TAC LV
AK128382
NP_004365


AV086493
cytochrome c oxidase, subunit VIIa 1
NM_009944
NP_034074
DOWN TAC LA

BM726594
NP_001855


AV133935
cytochrome c oxidase, subunit VIIa 3
NM_009945
NP_034075
DOWN TAC LA
DOWN TAC LV
BF210089
NP_001856


BG063960
cytochrome c oxidase, subunit VIIc
NM_007749
NP_031775
DOWN TAC LA

BI118114
NP_001858


AV086888
cytochrome c, somatic
NM_007808
NP_031834
DOWN TAC LA

NM_018947
NP_061820


AV093672
cytochrome c-1
NM_025567
NP_079843
DOWN TAC LA

BF569085
NP_001907


AV095067
DNA segment, Chr 18, Wayne
NM_138600
NP_613066

DOWN TAC LV
AK092507
NP_001173



State University 181, expressedtext missing or illegible when filed


AV083353
dodecenoyl-Coenzyme A delta
NM_010023
NP_034153
DOWN TAC LA
DOWN TAC LV
BQ277959
NP_001910



isomerase (3,2 trans-enoyl-Coetext missing or illegible when filed


BG074113
enoyl coenzyme A hydratase 1,
NM_016772
NP_058052
DOWN TAC LA

AK126566
NP_001389



peroxisomal


AU022217
epoxide hydrolase 2, cytoplasmic
NM_007940
NP_031966

DOWN TAC LV
AK094393
NP_001970


BG067242
ESTs
BE988802

DOWN TAC LA

NM_002660
NP_877963


AV006522
ESTs
NM_028545
NP_082821
DOWN TAC LA


AV095205
eukaryotic translation initiation
NM_010121
NP_034251
DOWN TAC LA

NM_004836
NP_004827



factor 2 alpha kinase 3


AV109470
expressed sequence AA959857
BC048412

DOWN TAC LA

NM_005463
NP_112740


AV006061
fatty acid Coenzyme A ligase,
NM_007981
NP_032007
DOWN TAC LA



long chain 2


AV140552
fumarate hydratase 1
BC006048


DOWN TAC LV


BG072359
fumarylacetoacetate hydrolase
NM_010176
NP_034306

DOWN TAC LV
BX537608
NP_000128


AI841654
G protein-coupled receptor 56
NM_018882
NP_061370

DOWN TAC LV
NM_201524
NP_958933


AV108357
galactokinase
NM_016905
NP_058601
DOWN TAC LA

BM471434
NP_000145


AA162908
gamma-glutamyl transpeptidase
NM_008116
NP_032142
DOWN TAC LA

BC035341
NP_038347


BG068200
GATA binding protein 6
AF179425


DOWN TAC LV
X95701
NP_005248


BG066689
glutamate oxaloacetate transaminase
NM_010324
NP_034454
DOWN TAC LA

BM994502
NP_002070



1, soluble


AV009064
glutamine synthetase
NM_008131
NP_032157
DOWN TAC LA

AL161952
NP_002056


AV134367
glutaryl-Coenzyme A dehydrogenase
NM_008097
NP_032123

DOWN TAC LV
BC002579
NP_039663


AV087315
guanosine monophosphate reductase
NM_025508
NP_079784

DOWN TAC LV
BM994423
NP_006868


AV022721
histidine ammonia lyase
NM_010401
NP_034531
DOWN TAC LA

NM_002108
NP_002099


BG073539
hydroxysteroid (17-beta)
NM_016763
NP_058043
DOWN TAC LA

BQ940058
NP_004484



dehydrogenase 10


BG068774
isocitrate dehydrogenase 3
NM_029573
NP_083849
DOWN TAC LA
DOWN TAC LV
AK123316
NP_005521



(NAD+) alpha


AA036340
isocitrate dehydrogenase 3
NM_130884
NP_570954
DOWN TAC LA

BQ051868
NP_777281



(NAD+) beta


AV005828
L-3-hydroxyacyl-Coenzyme A
NM_008212
NP_032238

DOWN TAC LV
AK096018
NP_005318



dehydrogenase, short chain


AV022047
lipin 1
NM_015763
NP_766538
DOWN TAC LA

AK127039
NP_663731


AV006290
lipoprotein lipase
NM_008509
NP_032535
DOWN TAC LA

NM_000237
NP_000228


BG064854
low density lipoprotein
AK084165

DOWN TAC LA

NM_004525
NP_004516



receptor-related protein 2


AV088662
malic enzyme, supernatant
NM_008615
NP_032641

DOWN TAC LV


AV057294
methylcrotonoyl-Coenzyme A
NM_023644
NP_076133

DOWN TAC LV
BC042453
NP_064551



carboxylase 1 (alpha)


AA108913
methylmalonyl-Coenzyme A mutase
NM_008650
NP_032676

DOWN TAC LV
BX647789
NP_000246


AV006153

Mus musculus, clone MGC: 7898

BF180657


DOWN TAC LV



IMAGE: 3582717, mRNA, comtext missing or illegible when filed


AI854120

Mus musculus, Similar to

NM_145567
NP_663542
DOWN TAC LA



3-hydroxyisobutyrate dehydrogenase,text missing or illegible when filed


AV088774

Mus musculus, Similar to

NM_145615
NP_663590
DOWN TAC LA

BM907902
NP_000117



electron-transfer-flavoprotein,



alpha ptext missing or illegible when filed


AV103083
NAD(P)H menadione oxidoreductase
NM_020282
NP_064678

DOWN TAC LV



2, dioxin inducible


AA162428
NADH dehydrogenase (ubiquinone) 1
NM_010885
NP_035015
DOWN TAC LA



alpha subcomplex 2


AV016078
NADH dehydrogenase (ubiquinone) 1
NM_010885
NP_035015
DOWN TAC LA



alpha subcomplex 2


AV140287
NADH dehydrogenase (ubiquinone) 1
NM_019443
NP_062316
DOWN TAC LA



alpha subcomplex, 1


AV050140
NADH dehydrogenase (ubiquinone) 1
BQ044115

DOWN TAC LA

BX538277
NP_002480



alpha subcomplex, 4


AV106199
NADH dehydrogenase (ubiquinone) 1
NM_025987
NP_080263
DOWN TAC LA
DOWN TAC LV
BM709562
NP_002481



alpha subcomplex, 6 (14text missing or illegible when filed


AW555047
NADH dehydrogenase (ubiquinone) 1
NM_023202
NP_075691
DOWN TAC LA
DOWN TAC LV
BM545518
NP_004992



alpha subcomplex, 7 (14text missing or illegible when filed


AI836747
NADH dehydrogenase (ubiquinone) 1
NM_023172
NP_075661
DOWN TAC LA

BM994434
NP_004996



beta subcomplex, 9


BG076060
NADH dehydrogenase (ubiquinone)
BU756147

DOWN TAC LA
DOWN TAC LV



Fe—S protein 3


AV084172
ornithine aminotransferase
NM_016978
NP_058674

DOWN TAC LV
BC016928
NP_000265


BG073162
oxysterol binding protein-like 1A
NM_020573
NP_065598
DOWN TAC LA

BX647893
NP_579802


BG071157
phosphate cytidylyltransferase 1,
AK083965

DOWN TAC LA

BC046355
NP_005008



choline, alpha isoform


AV033702
phospholipase A2 group VII
NM_013737
NP_038765
DOWN TAC LA

BC025674
NP_005075



(platelet-activating factor acetylhydtext missing or illegible when filed


BG068736
pyruvate dehydrogenase E1 alpha 1
NM_008810
NP_032836
DOWN TAC LA

AK092210
NP_000275


AV012729
retinoic acid induced 1
NM_011480
NP_035610
DOWN TAC LA

NM_030665
NP_109590


AA403731
RIKEN cDNA 0610009I16 gene
NM_026695
NP_080971
DOWN TAC LA

AL833205
NP_001976


AI841340
RIKEN cDNA 0610010E03 gene
NM_025321
NP_079597
DOWN TAC LA

BQ899032
NP_002992


BG072552
RIKEN cDNA 0610011L04 gene
NM_177470
NP_803421
DOWN TAC LA


AV093484
RIKEN cDNA 0610033L03 gene
NM_026703
NP_080979
DOWN TAC LA
DOWN TAC LV
BM704035
NP_055037


AW558029
RIKEN cDNA 0710008D09 gene
NM_025650
NP_079926
DOWN TAC LA


AV086467
RIKEN cDNA 1010001M12 gene
NM_025348
NP_079624
DOWN TAC LA

BM805609
NP_004533


AV133828
RIKEN cDNA 1010001N11 gene
NM_025358
NP_079634
DOWN TAC LA
DOWN TAC LV
BM546373
NP_004993


AV012912
RIKEN cDNA 1110038I05 gene
NM_134042
NP_598803

DOWN TAC LV
NM_005589
NP_005580


AV022384
RIKEN cDNA 1190017B19 gene
NM_023175
NP_075664
DOWN TAC LA


AV114239
RIKEN cDNA 1200006L06 gene
NM_024181
NP_077143

DOWN TAC LV


AV095102
RIKEN cDNA 1500004O06 gene
NM_025899
NP_080175
DOWN TAC LA

AK094006
NP_003357


AV052491
RIKEN cDNA 1810022C23 gene
NM_026947
NP_081223

DOWN TAC LV


AV063132
RIKEN cDNA 2210415M14 gene
NM_026219
NP_080495
DOWN TAC LA

BC041005
NP_006285


AV081301
RIKEN cDNA 2210418G03 gene
AK008974

DOWN TAC LA


AV085923
RIKEN cDNA 2310016C19 gene
NM_025862
NP_080138

DOWN TAC LV
AK125373
NP_055199


AV086427
RIKEN cDNA 2310021J10 gene
NM_025641
NP_079917
DOWN TAC LA


AV103530
RIKEN cDNA 2310039H15 gene
NM_028177
NP_082453
DOWN TAC LA
DOWN TAC LV
BE547177
NP_004994


AV095143
RIKEN cDNA 2410004H02 gene
NM_145954
NP_666066
DOWN TAC LA


BG063257
RIKEN cDNA 2510027N19 gene
NM_026330
NP_080606
DOWN TAC LA


AV077867
RIKEN cDNA 2610003B19 gene
NM_028177
NP_082453
DOWN TAC LA

BE547177
NP_004994


BG067911
RIKEN cDNA 2610020H15 gene
NM_025638
NP_079914
DOWN TAC LA
DOWN TAC LV


AV104092
RIKEN cDNA 2610034N03 gene
NM_025478
NP_079754
DOWN TAC LA


BG063943
RIKEN cDNA 2610041P16 gene
NM_025641
NP_079917
DOWN TAC LA


BG072165
RIKEN cDNA 2610205J15 gene
NM_152813
NP_690026

DOWN TAC LV


AV030438
RIKEN cDNA 2610207I16 gene
NM_024255
NP_077217

DOWN TAC LV


AV089737
RIKEN cDNA 3230402N08 gene
NM_021509
NP_067484
DOWN TAC LA

AY007239
NP_056344


AA154831
solute carrier family 27
NM_011978
NP_036108
DOWN TAC LA

D88308
NP_003636



(fatty acid transporter), member 2


AA673962
sortilin-related receptor, LDLR
AF031816

DOWN TAC LA

NM_003105
NP_003096



class A repeats-containing


AA146030
sterol carrier protein 2, liver
BC018384

DOWN TAC LA
DOWN TAC LV
BX537619
NP_002970


AV088223
succinate-CoA ligase, GDP-forming,
NM_019879
NP_063932

DOWN TAC LV
AK125502
NP_003840



alpha subunit


AV016790
thioredoxin-like 2
NM_023140
NP_075629
DOWN TAC LA

AJ010841
NP_006532








Claims
  • 1. A method for the diagnosis of pressure overload in the heart, the method comprising: determining the differential expression in one or more of the sequences set forth in Table I.
  • 2. The method according to claim 1, wherein said pressure overload is associated with atrial enlargement and/or ventricular hypertrophy.
  • 3. The method according to claim 1, wherein said determining comprises: contacting a biological sample comprising protein with an antibody that specifically binds to one or more of the proteins having amino acid sequences encoded by said pressure overload associated genes; detecting the presence of a complex formed between said antibody and said protein; wherein an alteration in the presence of said complex, compared to a control sample, is indicative of pressure overload in the heart.
  • 4. The method according to claim 3, wherein said biological sample is blood or serum.
  • 5. The method according to claim 4, wherein said biological sample is contacted with a panel of antibodies specific for pressure overload associated polypeptides.
  • 6. The method according to claim 3, wherein said pressure overload associated genes are set forth in Table II.
  • 7. The method according to claim 5, wherein said biological sample is cardiac cells.
  • 8. The method according to claim 7, wherein said contacting is performed in vivo.
  • 9. The method according to claim 8, the steps comprising: a) administering to a patient an effective amount of an imaging composition comprising: an antibody that specifically binds to a pressure overload associated polypeptide, and increases contrast between an overloaded cardiac tissue and surrounding tissue in a visualization method; and b) visualizing said imaging composition.
  • 10. The method according to claim 7, wherein said pressure overload associated genes are set forth in Table III.
  • 11. The method according to claim 1, wherein said determining comprises: contacting a biological sample comprising protein with a labeled substrate for a metabolic reaction catalyzed by said pressure overload associated genes; detecting the presence of the product of said metabolic reaction; wherein an increase in the presence of said complex, compared to a control sample, is indicative of pressure overload in the heart.
  • 12. The method according to claim 11, wherein said pressure overload associated gene is set forth in Table IV.
  • 13. The method according to claim 1, wherein said determining step comprises: contacting a biological sample comprising nucleic acids from a patient suspected of suffering from pressure overload with a probe that specifically binds to one or more of said sequences; detecting the presence of a complex formed between said probe and said nucleic acid; wherein an increase in the presence of said complex, compared to a control sample, is indicative of pressure overload of the heart.
  • 14. The method according to claim 13, wherein said biological sample comprises nucleic acids specifically amplified with said sequences.
  • 15. The method according to claim 13, wherein said biological sample is blood.
  • 16. The method according to claim 13, wherein said biological sample is contacted with a panel of pressure overload associated gene sequences.
  • 17. An array comprising two or more pressure overload associated genes as set forth in Table I, gene products, or antibodies specific for said gene products.
  • 18. A method for identifying an agent that modulates activity of a pressure overload associated gene or gene product, the method comprising: combining a candidate biologically active agent with any one of: (a) a polypeptide encoded by any one of the sequences set forth in Table I; (b) a cell comprising a nucleic acid encoding and expressing a polypeptide encoded by any one of the sequences set forth in Table I; or (c) a non-human transgenic animal model for pressure overload associated gene function comprising one of: (i) a knockout of a gene corresponding to any one of the sequences set forth in Table I; (ii) an exogenous and stably transmitted mammalian gene sequence comprising any one of the sequences set forth in Table I; and determining the effect of said agent on pressure overload induced molecular and cellular changes.
  • 19. The method according to claim 18, wherein said biologically active agent upregulates activity.
  • 20. The method according to claim 18, wherein said biologically active agent downregulates activity.
  • 21. The method according to claim 20, wherein said biologically active agent binds to said polypeptide.
  • 22. The method according to claim 1, wherein said sequence is set forth in Table IA.
  • 23. The method according to claim 1, wherein said sequence is set forth in Table IB.
Provisional Applications (1)
Number Date Country
60611674 Sep 2004 US