Methods and compositions for diagnosis, monitoring and development of therapeutics for treatment of atherosclerotic disease

Information

  • Patent Application
  • 20070092886
  • Publication Number
    20070092886
  • Date Filed
    March 22, 2006
    18 years ago
  • Date Published
    April 26, 2007
    17 years ago
Abstract
Polynucleotide sequences are provided that correspond to genes that are differentially expressed in atherosclerotic disease conditions. Methods for using these sequences to detect gene expression and/or for transcriptional profiling in mammals are also provided. The polynucleotide sequences of the invention may be used, for example, to diagnose atherosclerotic disease, to monitor extent of progression or efficacy of treatment or to assess prognosis of atherosclerotic disease, and/or to identify compounds effective to treat an atherosclerotic disease condition.
Description
FIELD OF THE INVENTION

This application is in the field of atherosclerotic disease. In particular, this invention relates to methods and compositions for diagnosing, monitoring, and development of therapeutics for atherosclerotic disease.


BACKGROUND OF THE INVENTION

Atherosclerosis is the primary cause of heart disease and stroke (Kannel and Belanger (1991) Am. Heart J 121:951-57), and is the most common cause of morbidity and mortality in the United States (NHLBI Morbidity and Mortality Chartbook, National Heart, Lung, and Blood Institute, Bethesda, MD, May, 2002; NHLBI Fact Book, Fiscal Year 2003, pp. 35-53, National Heart, Lung, and Blood Institute, Bethesda, MD, February, 2004). Atherosclerosis is currently conceptualized as a chronic inflammatory disease of the arterial vessel wall that develops due to complex interactions between the environment and the genetic makeup of an individual (Ross (1999) N Engl J Med 340:115-26). Development of an atherosclerotic plaque occurs in stages, beginning with simple fatty streak formation and culminating in complex calcified lesions containing abnormal accumulation of smooth muscle cells, inflammatory cells, lipids, and necrotic debris. It is likely that the various stages of atherosclerotic disease are governed by a set of genes that are expressed by a variety of cell types present in the vessel wall.


The propensity for developing atherosclerosis is dependent on underlying genetic risk, and varies as a function of age and exposure to environmental risk factors. However, despite the chronic nature of atherosclerotic disease, knowledge regarding temporal gene expression during the course of disease progression is very limited. The prolonged, chronic, and unpredictable nature of the disease in humans, by virtue of heterogeneous genetic and environment factors, has limited systematic temporal gene expression studies in humans.


The roles of a limited number of genes that are differentially expressed in vascular disease have been identified, and a few of these genes linked through mechanistic studies to disease processes (Glass and Witztum (2001) Cell 104:503-16; Breslow (1996) Science 272:685-88; Lusis (2000) Nature 407:233-41). Recent efforts to identify disease related gene expression patterns have employed transcriptional profiling with DNA microarrays. However, these studies have included relatively small arrays (Wuttge et al. (2001) Mol Med 7:383-392) as well as limited time points, with the primary comparison between normal and late stage diseased tissue (Archacki et al. (2003) Physiol Genomics 15:65-74; Faber et al. (2002) Curr Opin Lipidol 13:545-552; McCaffrey et al. (2000) J Clin Invest 105:653-662; Randi et al. (2003) J Throm Haemost 1:829-835; Seo et al. (2004) Arterioscler Thromb Vasc Biol 24:1922-1927; Zohlnhofer et al. (2001) Mol Cell 7:1059-1069. Utilizing microarrays in animal models, where a disease process can be studied over time, the impact of individual risk factors and perturbations on the expression of individual genes during disease development can be studied systematically without a priori knowledge of gene identity. The temporal expression patterns of the genes can then be correlated with the well-described disease stages.


There is a need for a comprehensive list of atherosclerosis-related genes that are predictive of atherosclerotic disease conditions, for use as diagnostic markers and for discovery of biochemical pathways involved in development of atherosclerotic disease and discovery and/or testing of new therapeutics.


BRIEF SUMMARY OF THE INVENTION

This invention provides compositions, methods, and kits for detection of gene expression, diagnosis, monitoring, and development of therapeutics with respect to atherosclerotic disease.


In one aspect, the invention provides a system for detecting gene expression, comprising at least two isolated polynucleotide molecules, wherein each isolated polynucleotide molecule detects an expressed gene product from a gene that is differentially expressed in atherosclerotic disease in a mammal. In one embodiment, the differentially expressed gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the differentially expressed gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927. In various embodiments, a system for detecting gene expression comprises any of at least 3, 5, 10, 15, 20, 25, 30, 35, 40, 50, 60, 70, 80, 90, or 100 of the isolated polynucleotide molecules described herein or their polynucleotide complements, or human homologs or orthologs thereof. In one embodiment, the gene expression system comprises at least two isolated polynucleotide molecules, wherein each isolated polynucleotide molecule detects an expressed gene product, wherein the gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927, wherein the gene is differentially expressed in atherosclerotic disease in a mammal, and wherein the gene expression system comprises at least 1, 3, 5, 10, 15, 20, 25, or 30 isolated polynucleotide molecules that detect genes corresponding to the polynucleotide sequences selected from the group consisting of SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.


In some embodiments, the isolated polynucleotide molecules are immobilized on an array, which may be selected from the group consisting of a chip array, a plate array, a bead array, a pin array, a membrane array, a solid surface array, a liquid array, an oligonucleotide array, a polynucleotide array, a cDNA array, a microtiter plate, a membrane, and a chip. The isolated polynucleotide molecules may be selected from the group consisting of synthetic DNA, genomic DNA, cDNA, RNA, or PNA. A gene corresponding to an isolated polynucleotide molecules described herein may be differentially expressed in any blood vessel or portion thereof which has developed an atherosclerotic or inflammatory disease, for example, the aorta, a coronary artery, the carotid artery, or a blood vessel of the peripheral vasculature.


In another aspect, the invention provides a kit comprising a system for detecting gene expression as described above. In one embodiment, the kit comprises an array comprising a system for detecting gene expression as described above.


In another aspect, the invention provides a method of detecting gene expression, comprising contacting products of gene expression with the system for detecting gene expression as described above. In one embodiment, the method comprises isolating mRNA, for example from a sample from individual who has or who is suspected of having an atherosclerotic disease, and hybridizing the RNA to the polynucleotide molecules from the system for detecting gene expression. In another embodiment, the method comprises isolating mRNA, converting the RNA to nucleic acid derived from the RNA, e.g., cDNA, and hybridizing the nucleic acid derived from the RNA to the polynucleotide molecules of the system for detecting gene expression. Optionally, the RNA may be amplified prior to hybridization to the system for gene expression. Optionally, the RNA is detectably labeled, and determination of presence, absence, or amount of an RNA molecule corresponding to a gene detected by a polynucleotide molecule of the system for detecting gene expression comprises detection of the label.


In another embodiment, the method for detecting gene expression comprises isolating proteins from an individual who has or who is suspected of having an atherosclerotic disease, and detecting the presence, absence, or amount of one or more proteins corresponding to the gene expression product of a gene that is differentially expressed in atherosclerotic disease and corresponds to a polynucleotide molecule of the system for detecting gene expression as described above. Detection may be via an antibody that recognizes the protein, for example, by contacting the isolated proteins with an antibody array.


In another aspect, the invention provides a method for diagnosing an atherosclerotic disease in an individual, comprising contacting polynucleotides derived from a sample from the individual with a system for detecting gene expression as described above. In one embodiment, the method comprises detecting hybridization complexes formed, if any, wherein presence, absence or amount of hybridization complexes formed from at least one of the polynucleotides from the individual is indicative of presence or absence of the atherosclerotic disease. In another embodiment, the method comprises comparing levels of expression of the genes with a molecular signature indicative of the presence or absence of the atherosclerotic disease.


In another aspect, the invention provides a method for assessing extent of progression of atherosclerotic disease in an individual, comprising contacting polynucleotides derived from a sample from the individual with a system for detecting gene expression as described above. In one embodiment, the method comprises detecting hybridization complexes formed, if any, wherein presence, absence or amount of hybridization complexes formed from at least one of the polynucleotides from the individual is indicative of extent of progression of the atherosclerotic disease. In another embodiment, the method comprises detecting hybridization complexes formed, if any, and comparing levels of expression of the genes with a molecular signature indicative of extent of progression of the atherosclerotic disease.


In another aspect, the invention provides a method of assessing efficacy of treatment of atherosclerotic disease in an individual, comprising contacting polynucleotides derived from a sample from the individual with a system for detecting gene expression as described above. In one embodiment, the method comprises detecting hybridization complexes formed, if any, wherein presence, absence or amount of hybridization complexes formed from at least one of the polynucleotides from the individual is indicative of extent of progression of the atherosclerotic disease. In another embodiment, the method comprises comparing levels of expression of the genes with a molecular signature indicative of extent of progression of the atherosclerotic disease.


In another aspect, the invention provides a method for determining prognosis of atherosclerotic disease in an individual, comprising contacting polynucleotides derived from a sample from the individual with a system for detecting gene expression as described above. In one embodiment, the method comprises detecting hybridization complexes formed, if any, wherein presence, absence or amount of hybridization complexes formed from at least one of the polynucleotides from the individual is indicative of prognosis of the atherosclerotic disease. In another embodiment, the method comprises comparing levels of expression of the genes with a molecular signature indicative of prognosis of the atherosclerotic disease.


In another aspect, the invention provides a method for identifying a compound effective to treat an atherosclerotic disease, comprising administering a test compound to a mammal with an atherosclerotic disease condition and contacting polynucleotides derived from a sample from the mammal with a system for detecting gene expression as described above. In one embodiment, the method comprises detecting hybridization complexes formed, if any, wherein presence, absence or amount of hybridization complexes formed from at least one of the polynucleotides from the individual is indicative of treatment of the disease. In another embodiment, the invention comprises detecting hybridization complexes formed, if any, and comparing levels of expression of the genes with a molecular signature indicative of treatment of the disease.


In another aspect, the invention provides a method of monitoring atherosclerotic disease in a mammal, comprising detecting the expression level of at least one, at least two, at least ten, at least one hundred, or more genes selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927. In some embodiments, at least one of the genes for which expression level is detected is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In one embodiment, the atherosclerotic disease comprises coronary artery disease. In one embodiment, the atherosclerotic disease comprises carotid atherosclerosis. In one embodiment, the atherosclerotic disease comprises peripheral vascular disease. In some embodiments, the expression level of said gene(s) is detected by measuring the RNA expression level. In one embodiment, RNA is isolated from the individual prior to detection of the RNA expression level. Measurement of RNA expression level may comprise amplifying RNA from an individual, for example, by polymerase chain reaction (PCR), using a primer that is complementary to a polynucleotide sequence corresponding to a gene to be detected, wherein the gene corresponds to a polynucleotide sequence selected from the group of genes depicted in SEQ ID NOs: 1-927. In some embodiments, a primer is used that is complementary to a polynucleotide sequence corresponding to a gene to be detected, wherein the gene corresponds to a polynucleotide sequence selected from the group of genes depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. Measurement of RNA expression level may comprise hybridization of RNA from the individual to a polynucleotide corresponding to a gene to be detected, wherein the gene corresponds to a polynucleotide sequence selected from the group of genes depicted in SEQ ID NOs: 1-927. In some embodiments, RNA from the individual is hybridized to a polynucleotide corresponding to a gene to be detected, wherein the gene to be detected is selected from the group of genes depicted in 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In some embodiments, gene expression level is detected by measuring the expressed protein level. In some embodiments, the method further comprises selecting an appropriate therapy for treatment or prevention of the atherosclerotic disease. In some embodiments, gene expression level, for example, RNA or protein level, is detected in serum from an individual.


In another aspect, the invention provides a method of monitoring atherosclerotic disease in an individual, comprising detecting RNA expressed from at least one gene selected from the group of genes corresponding to at least one polynucleotide sequence depicted in SEQ ID NOs: 1-927. In one embodiment, the at least one gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In one embodiment, the method comprises measuring the expressed RNA in serum from the individual.


In another aspect, the invention provides a method of monitoring atherosclerotic disease in an individual, comprising detecting protein expressed from at least one gene selected from the group of genes corresponding to at least one polynucleotide sequence depicted in SEQ ID NOs:1-927. In one embodiment, the at least one gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In one embodiment, the method comprises measuring the expressed protein in serum from the individual.




BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 depicts the experimental design of the experiments described in Example 1. ApoE deficient mice (C57BL/6J-Apoe5mlUnc), were fed non-cholate-containing high-fat diet from 4 weeks of age for a maximum period of 40 weeks. Aortas were obtained for transcriptional profiling at pre-determined time intervals corresponding to various stages of atherosclerotic plaque formation. For each time point, aortas from 15 mice were combined into 3 pools for microarray replicate studies. To eliminate gene expression differences due to aging, diet, and genetic differences, a number of control groups were also used at each time point, including apoE deficient mice on normal chow, aw well as C57Bl/6 and C3H/HeJ wild type mice on both normal and atherogenic diets.



FIG. 2 depicts quantification of atherosclerotic disease in the experiments described in Example 1. Percent lesion area was determined by calculating the ratio of atherosclerotic area versus total surface area of the aorta. ApoE-deficient mice (n=7) on high-fat diet were compared to other control mice (n=5-7 for each mouse/diet combination). Representative time intervals were used for analysis, including baseline (TOO) measurements in mice prior to initiation of diet at 4 weeks of age and end point measurements corresponding to 40 weeks (T40) on either high-fat or normal diet. At T00, three were no statistically significant differences in lesion area among the various conditions. At 40 weeks on high-fat diet, the controls did not develop any lesions. In contrast to the control mice, the ApoE-deficient mice on normal chow and on high-fat diet had significantly larger atherosclerotic area (14.00% +/−3.92%, p<0.0001, and 37.98% +/−6.3%, p<0.0001, respectively.)



FIG. 3 depicts atherosclerosis genes identified in the experiments described in Example 1. Employing a newly-developed statistical algorithm which relies on permutation analysis and generalized regression, atherosclerosis-related genes were identified. Selecting the genes on the basis of their false detection rate (FDR<0.05) and depicting their expression with a heatmap (ordered by hierarchical clustering), demonstrates profiles which closely correlate with disease progression. The heatmap is a graphic representation of expression patterns of 6 parallel time course studies with time progressing from left to right for each of the 6 sets of strain-diet combination. Each set of the strain-diet combination therefore contains 15 columns (3 for each of 5 time points). Each row represents the row normalized expression pattern of a single gene. The dominant temporal pattern of expression is one that increases linearly with time (667 genes). Fewer genes (64) reveal an opposite pattern. HF: high-fat diet; NC: normal chow.



FIG. 4 depicts time-related patterns of gene expression in atherosclerosis observed in the experiments described in Example 1. Using AUC analysis, a number of distinct time-related patterns of gene expression in ApoE-deficient mice on high-fat diet were observed. Eight different time-related patterns are depicted, with the y-axis representing normalized gene expression values and the x-axis representing 6 different time points from time 0 to 40 weeks. The genes in each pattern were clustered based on positive correlation values. The mean distance of genes from the center of each cluster is noted in parentheses for each pattern. Using enrichment analysis for each cluster of genes, specific pathways were found to be associated with these patterns that reflect particular biological processes.



FIG. 5 depicts the identification and validation of mouse atherosclerotic disease classifier genes as determined in the experiments described in Example 1. FIG. 5A depicts identification of the classification gene set. The SVM algorithm described in Example 1 was employed to rank genes based on their abilities to accurately discriminate between 5 time points in ApoE-deficient mice on high-fat diet. An optimal set of 38 genes was identified to classify the experiments at a minimal error rate of 15%. The optimal 15% error rate was determined with a 1000 step cross-validation method with 25% of the experiments employed as the test group and the rest as the training group. FIG. 5B depicts classification of an independent mouse atherosclerosis data set. Aortas of ApoE-deficient mice aged 16 weeks were used for gene expression profiling utilizing a different microarray and labeling protocol than in the experiment depicted in FIG. 5A. Using the SVM algorithm, where known experiments were the five time points in the original experimental design and the independent set of experiments was the test set, these mice most closely classified with the 24 week time point. SVM scores for each experiment based on one-versus-all comparisons are represented graphically in a heatmap.



FIG. 6 depicts expression of atherosclerosis-related genes in human coronary artery disease, as described in Example 1. To investigate the expression profile of differently regulated mouse genes in human coronary artery atherosclerosis, 40 coronary artery samples with and without atherosclerotic lesions were used for transcriptional profiling. Atherosclerosis-associated mouse genes were matched to human orthologs/homologs by gene symbol and by known homology, and their expression was compared in human atherosclerotic plaques classified as lesion versus no lesion (SAM FDR<0.025). The expression of the top genes is represented graphically as a heatmap, where rows represent row normalized expression of each gene and the columns represent coronary artery samples. Calculated SAM FDR<0.009 for d-score 4.25-2.45, FDR<0.015 for d-score 2.41-2.357, FDR<0.025 for d-score 2.33-2.05.



FIG. 7 depicts the experimental design of the experiments described in Example 2. FIG. 7A: Four-week-old female C3H/HeJ (C3H) and C57B16 (C57) mice were fed normal chow vs. high-fat diet for the maximum period of 40 weeks. Triplicate microarray experiments were performed for each time point using 3 pools of 5 aortas at 0, 4, 10, 24, and 40 weeks on either diet (total of 15 mice per time point). FIG. 7B: Data analysis overview. Of the 20,283 genes present on the array, 311 genes were found to be significantly differentially expressed between C3H and C57 mice at baseline (SAM FDR 10% and >1.5-fold change). Differential gene expression during aging was determined by comparing C57 vs. C3H time-course differences on normal and atherogenic high-fat diets using AUC analysis.



FIG. 8 depicts differential gene expression between C3H and C57 mice at baseline. The SAM analysis shown was associated with an FDR of 10%, and a total of 311 probes were identified as differentially regulated at this level of confidence. Lists represent a select group of genes (expressed sequence tags excluded) with higher expression in C3H (top 20 ranking genes) and C57 (top 45 ranking genes). The heatmap reflects normalized gene expression ratios and is organized with individual hybridizations for each of the 3 replicates for each mouse strain arranged along the x axis.



FIG. 9 depicts differential gene expression between C3H and C57 mice in response to normal aging. FIG. 9A: Response to aging was determined by comparing C57 vs. C3H time-course differences on normal diet (AUC analysis F statistic>10). FIG. 9B: Functional annotation of the 413 differentially expressed genes reveals differences in various biological processes, including growth and differentiation. The probability rates provided area based on Fisher exact test (P<0.02). FIG. 9C: K-means clustering of the 413 genes reveals several profiles of gene expression. Clusters 1, 4, and 9 reveal increased gene expression in C3H vs. C57 mice, whereas clusters 2, 6, and 14 reveal the opposite pattern.



FIG. 10 depicts differential gene expression between C3H and C57 mice in response to high-fat diet. FIG. 10A: Response to atherogenic stimulus was determined by comparing C57 vs. C3H time-course differences on high -fat diet (AUC analysis F statistic>10). FIG. 1OB: Functional annotation of the 509 differentially expressed genes reveals differences in various biological processes and cellular components. The probability rates provided are based on Fisher exact test (P<0.02). FIG. 1OC: K-means clustering of the 509 differentially expressed genes revealed several patterns of gene expression with clusters 3 and 9 exhibiting increased gene expression in C3H vs. C57 mice and clusters 8 and 10 with the opposite pattern.



FIG. 11 shows the results of evaluation in the apoe knockout model of genes identified as differentially expressed between C3H and C57 strains. FIG. 11A: ApoE knockout mice (C57BL/6J-Apoe™lUnc) were fed normal chow versus high-fat diet for the maximum period of 40 weeks. Triplicate microarray experiments were preformed for each time point using 3 pools of 5 aortas at 0, 4, 10, 24, and 40 weeks for regular and high-fat diet groups (total of 15 mice per time point). SOMs were used to visualize patterns of expression of genes of interest. Genes which were differentially regulated by aging (FIG. 9, K-means clusters 1, 4, and 9 with higher expression in C3H and clusters 4, 6, and 14 with higher expression in C57) and genes identified with atherogenic stimuli (FIG. 10, K-means clusters 3 and 9 with higher expression in C3H and clusters 8 and 10 with opposite pattern) as well as genes which were differentially expressed at the baseline time point (FIG. 8), were grouped and their expression was studied using SOM analysis. SOM analysis reveals diverse patterns of expression of these genes throughout the development of atherosclerosis in apoe knockout mice. Cluster 8 contains genes that are consistently increasing in expression with progression of atherosclerosis. Pie charts reflect the analysis group from which the genes populating each cluster were derived. The relative size of sectors of the pie chart indicates the relative number of genes that are derived from the various staging groups. FIG. 11B lists genes with higher expression in C57 mice at baseline and in C3H mice at baseline or on a high fat diet.




DETAILED DESCRIPTION OF THE INVENTION

The invention provides polynucleotide sequences that correspond to genes that are differentially expressed in atherosclerotic disease conditions, and methods for using these sequences to detect gene expression and/or for transcriptional profiling in mammals. The polynucleotide sequences provided herein may be used, for example, to diagnose, assess extent of progression, assess efficacy of treatment of, to determine prognosis of, and/or to identify compounds effective to treat an atherosclerotic disease condition. The polynucleotide sequences herein may also be used in methods for elucidation of biochemical pathways that are involved in development and/or maintenance of atherosclerotic disease conditions.


General Techniques


The practice of the present invention will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, and biochemistry, which are within the skill of the art. Such techniques are explained fully in the literature, such as: Molecular Cloning: A Laboratory Manual, vol. 1-3, third edition (Sambrook et al., 2001); Oligonucleotide Synthesis (M.J. Gait, ed., 1984); Methods in Enzymology (Academic Press, Inc.); Current Protocols in Molecular Biology (F.M. Ausubel et al., eds., 1987); PCR Cloning Protocols, (Yuan and Janes, eds., 2002, Humana Press).


In addition to the above references, protocols for in vitro amplification techniques, such as the polymerase chain reaction (PCR), the ligase chain reaction (LCR), Qβ-replicase amplification, and other RNA polymerase mediated techniques (e.g., NASBA), useful, e.g., for amplifying oligonucleotide probes of the invention, are found in Mullis et al., U.S. Pat. No. (1987) 4,683,202; PCR Protocols: A Guide to Methods and Applications (Innis et al., eds.) Academic Press, Inc., San Diego, CA (1990); Amnheim and Levinson (1990) C&EN 36; The Journal of NIH Research (1991) 3:81; 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; Van Brunt (1990) Biotechnology 8:291; Wu and Wallace (1989) Gene 4:560; Barringer et al. (1990) Gene 89:117; Sooknanan and Malek (1995) Biotechnology 13:563. Additional methods, useful for cloning nucleic acids, include Wallace et al., U.S. Patent No. 5,426,039. Improved methods of amplifying large nucleic acids by PCR are summarized in Cheng et al. (1994) Nature 369:684, and the references therein.


Definitions


Unless defined otherwise, all scientific and technical terms are understood to have the same meaning as commonly used in the art to which they pertain. For the purpose of the present invention, the following terms are defined below.


As used herein, the term “gene expression system” or “system for detecting gene expression” refers to any system, device or means to detect gene expression and includes candidate libraries, oligonucleotide sets or probe sets.


The term “diagnostic oligonucleotide set” generally refers to a set of two or more oligonucleotides that, when evaluated for differential expression of their products, collectively yields predictive data. Such predictive data typically relates to diagnosis, prognosis, monitoring of therapeutic outcomes, and the like. In general, the components of a diagnostic oligonucleotide set are distinguished from nucleotide sequences that are evaluated by analysis of the DNA to directly determine the genotype of an individual as it correlates with a specified trait or phenotype, such as a disease, in that it is the pattern of expression of the components of the diagnostic nucleotide set, rather than mutation or polymorphism of the DNA sequence that provides predictive value. It will be understood that a particular component (or member) of a diagnostic nucleotide set can, in some cases, also present one or more mutations, or polymorphisms that are amenable to direct genotyping by any of a variety of well known analysis methods, e.g., Southern blotting, RFLP, AFLP, SSCP, SNP, and the like.


A “disease specific target oligonucleotide sequence” is a gene or other oligonucleotide that encodes a polypeptide, most typically a protein, or a subunit of a multi-subunit protein, that is a therapeutic target for a disease, or group of diseases.


A “candidate library” or a “candidate oligonucleotide library” refers to a collection of oligonucleotide sequences (or gene sequences) that by one or more criteria have an increased probability of being associated with a particular disease or group of diseases. The criteria can be, for example, a differential expression pattern in a disease state, tissue specific expression as reported in a sequence database, differential expression in a tissue or cell type of interest, or the like. Typically, a candidate library has at least 2 members or components; more typically, the library has in excess of about 10, or about 100, or about 500, or even more, members or components.


The term “disease criterion” is used herein to designate an indicator of a disease, such as a diagnostic factor, a prognostic factor, a factor indicated by a medical or family history, a genetic factor, or a symptom, as well as an overt or confirmed diagnosis of a disease associated with several indicators. A disease criterion includes data describing a patient's health status, including retrospective or prospective health data, e.g., in the form of the patient's medical history, laboratory test results, diagnostic test results, clinical events, medications, lists, response(s) to treatment and risk factors, etc.


The terms “molecular signature” or “expression profile” refers to the collection of expression values for a plurality (e.g., at least 2, but frequently at least about 10, about 30, about 100, about 500, or more) of members of a candidate library. In many cases, the molecular signature represents the expression pattern for all of the nucleotide sequences in a library or array of candidate or diagnostic nucleotide sequences or genes. Alternatively, the molecular signature represents the expression pattern for one or more subsets of the candidate library.


The terms “oligonucleotide” and “polynucleotide” and “nucleic acid,” used interchangeably herein, refer to a polymeric form of two or more nucleotides of any length and any three-dimensional structure (e.g., single-stranded, double-stranded, triple-helical, etc.), which contain deoxyribonucleotides, ribonucleotides, and/or analogs or modified forms of deoxyribonucleotides or ribonucleotides. Nucleotides may be DNA or RNA, and may be naturally occurring, or synthetic, or non-naturally occurring. A nucleic acid of the present invention may contain phosphodiester bonds or an alternate backbone, comprising, for example, phosphoramide, phosphorothioate, phosphorodithioate, O-methylphosphoroamidite linkages, and peptide nucleic acid backbones and linkages. The term polynucleotide includes peptide nucleic acids (PNA).


The terms “polypeptide,”“peptide,” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. The terms apply to amino acid polymers in which one or more amino acid residue is an analogue of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers. The term also includes variants on the traditional peptide linkage joining the amino acids making up the polypeptide.


An “isolated” or “purified” polynucleotide or polypeptide is one that is substantially free of the materials with which it is associated in nature. By substantially free is meant at least 50%, preferably at least 70%, more preferably at least 80%, and even more preferably at least 90% free of the materials with which it is associated in nature.


As used herein, “individual” refers to a vertebrate, typically a mammal, such as a human, a nonhuman primate, an experimental animal, such as a mouse or rat, a pet animal, such as a cat or dog, or a farm animal, such as a horse, sheep, cow, or pig.


The term “healthy individual,” as used herein, is relative to a specified disease or disease criterion, e.g., the individual does not exhibit the specified disease criterion or is not diagnosed with the specified disease. It will be understood that the individual in question can exhibit symptoms, or possess various indicator factors, for another disease.


Similarly, an “individual diagnosed with a disease” refers to an individual diagnosed with a specified disease (or disease criterion). Such an individual may, or may not, also exhibit a disease criterion associated with, or be diagnosed with another (related or unrelated) disease.


An “array” is a spatially or logically organized collection, e.g., of oligonucleotide sequences or nucleotide sequence products such as RNA or proteins encoded by an oligonucleotide sequence. In some embodiments, an array includes antibodies or other binding reagents specific for products of a candidate library.


When referring to a pattern of expression, a “qualitative” difference in gene expression refers to a difference that is not assigned a relative value. That is, such a difference is designated by an “all or nothing” valuation. Such an all or nothing variation can be, for example, expression above or below a threshold of detection (an on/off pattern of expression). Alternatively, a qualitative difference can refer to expression of different types of expression products, e.g., different alleles (e.g., a mutant or polymorphic allele), variants (including sequence variants as well as post-translationally modified variants), etc.


In contrast, a “quantitative” difference, when referring to a pattern of gene expression, refers to a difference in expression that can be assigned a numerical value, such as a value on a graduated scale, (e.g., a 0-5 or 1-10 scale, a +-+++ scale, a grade 1-grade 5 scale, or the like; it will be understood that the numbers selected for illustration are entirely arbitrary and in no-way are meant to be interpreted to limit the invention).


The term “monitoring” is used herein to describe the use of gene sets to provide useful information about an individual or an individual's health or disease status. “Monitoring” can include, for example, determination of prognosis, risk-stratification, selection of drug therapy, assessment of ongoing drug therapy, determination of effectiveness of treatment, prediction of outcomes, determination of response to therapy, diagnosis of a disease or disease complication, following of progression of a disease or providing any information relating to a patient's health status over time, selecting patients most likely to benefit from experimental therapies with known molecular mechanisms of action, selecting patients most likely to benefit from approved drugs with known molecular mechanisms where that mechanism may be important in a small subset of a disease for which the medication may not have a label, screening a patient population to help decide on a more invasive/expensive test, for example, a cascade of tests from a non-invasive blood test to a more invasive option such as biopsy, or testing to assess side effects of drugs used to treat another indication.


System for Detecting Gene Expression


The invention provides a system for detecting expression of genes that are differentially expressed in atherosclerotic disease. In one embodiment, the system for detecting gene expression detects at least two expressed gene products of genes selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the system for detecting gene expression detects at least two expressed gene products of genes selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927. The term “corresponding” as used herein in the context of a gene corresponding to a polynucleotide sequence depicted in the Sequence Listing refers to a gene that is detectable by interaction of a product of expression of the gene (e.g., mRNA, protein) or a product derived from a product of expression of the gene (e.g., cDNA) with the system for detecting gene expression. The polynucleotide sequences represented by Sequence Identification Nos. 1-927 and accompanying identifying information are depicted in Table 1 below. These sequences have been shown to be differentially expressed in atherosclerosis in mice (see Example 1). The 60 mer sequences represented in Table I are encompassed within the genes indicated therein. The gene sequences are obtainable from publicly available databases such as GenBank, and at http://www.ncbi.nlm.nih.gov or http://source.stanford.edu/cgi-bin/source/sourceSearch, using the identifying information provided in Table 1.


In one embodiment, the system for detecting gene expression includes at least two isolated polynucleotide molecules, each of which detects an expressed gene product of a gene that is differentially expressed in atherosclerotic disease in a mammal. The gene expression system includes at least two isolated polynucleotides that each comprise at least a portion of a sequence depicted in the Sequence Listing or its complement (i.e., a polynucleotide sequence capable of hybridizing to a sequence depicted in the sequence listing). A system for detecting gene expression in accordance with the invention may include any of at least 2, 3, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, or 100 polynucleotides each comprising at least a portion of a polynucleotide depicted in the Sequence Listing or a polynucleotide complement thereof.


It is understood that the polynucleotides of the invention may have slightly different sequences than those identified herein. Such sequence variations are understood to those of ordinary skill in the art to be variations in the sequence that do not significantly affect the ability of the sequences to detect gene expression. For example, homologs and variants of the polynucleotides disclosed herein may be used in the present invention. Homologs and variants of these polynucleotide molecules possess a relatively high degree of sequence identity when aligned using standard methods. Polynucleotide sequences encompassed by the invention have at least 40-50, 50-60, 70-80, 80-85, 85-90, 90-95 or 95-100% sequence identity to the sequences disclosed herein.


It is understood that for expression profiling, variations in the disclosed polynucleotide sequences will still permit detection of gene expression. The degree of sequence identity required to detect gene expression varies depending on the length of an oligonucleotide. For example, for a 60mer (i.e., an oligonucleotide with 60 nucleotides), 6-8 random mutations or 6-8 random deletions do not affect gene expression detection. Hughes, T. R., et al. (2001) Nature Biotechnology 19:343-347. As the length of the polynucleotide sequence is increased, the number of mutations or deletions permitted while still allowing gene expression detection is increased.


As will be appreciated by those skilled in the art, the sequences of the present invention may contain sequencing errors. For example, there may be incorrect nucleotides, frameshifts, unknown nucleotides, or other types of sequencing errors in any of the sequences; however, the correct sequences will fall within the homology and stringency definitions herein.


In some embodiments, polynucleotide molecules are less than about any of the following lengths (in bases or base pairs): 10,000; 5000; 2500; 2000; 1500; 1250; 1000; 750; 500; 300; 250; 200; 175; 150; 125; 100; 75; 50; 25; 10. In some embodiments, polynucleotide molecules are greater than about any of the following lengths (in bases or base pairs): 10; 15; 20; 25; 30; 40; 50; 60; 75; 100; 125; 150; 175; 200; 250; 300; 350; 400; 500; 750; 1000; 2000; 5000; 7500; 10,000; 20,000; 50,000. Alternately, a polynucleotide molecule can be any of a range of sizes having an upper limit of 10,000; 5000; 2500; 2000; 1500; 1250; 1000; 750; 500; 300; 250; 200; 175; 150; 125; 100; 75; 50; 25; or 10 and an independently selected lower limit of 10; 15; 20; 25; 30; 40; 50; 60; 75; 100; 125; 150; 175; 200; 250; 300; 350; 400; 500; 750; 1000; 2000; 5000; or 7500, wherein the lower limit is less than the upper limit.


The isolated polynucleotides of the system for detecting gene expression may include DNA or RNA or a combination thereof, and/or modified forms thereof, and/or may also include a modified polynucleotide backbone. In some embodiments, the isolated polynucleotides are selected from the group consisting of synthetic oligonucleotides, genomic DNA, cDNA, RNA, or PNA.


In one embodiment, the system for detecting gene expression comprises two antibody molecules or antigen binding fragments thereof, each of which detects an expressed gene product (e.g., a polypeptide) of a gene that is differentially expressed in atherosclerotic disease in a mammal.


As used herein, “atherosclerotic disease” refers to a vascular inflammatory disease characterized by the deposition of atheromatous plaques containing cholesterol, lipids, and inflammatory cells within the walls of large and medium-sized blood vessels, which can lead to hardening of blood vessels, stenosis, and thrombotic and embolic events. Atherosclerosis includes coronary vascular disease, cerebral vascular disease, and peripheral vascular disease. The term “atherosclerotic disease” as used herein includes any condition associated with atherosclerosis in a mammal in which differential gene expression may be detected by a system for detecting gene expression as described herein. Examples of such atherosclerotic disease conditions include, but are not limited to, coronary artery disease (e.g., stable angina, unstable angina, exertional angina, myocardial infarction, congestive heart failure, sudden cardiac death, atrial fibrillation), cerebral vascular disease (e.g., stroke, cerebrovascular accident (CVA), transient ischemic attack (TIA), cerebral infarction, cerebral intermittent claudication), peripheral vascular disease (e.g., claudications), extracranial carotid disease, carotid plaque, and carotid bruit.


Arrays


In some embodiments, a system for detecting gene expression in accordance with the invention is in the form of an array. “Microarray” and “array,” as used interchangeably herein, comprise a surface with an array, preferably ordered array, of putative binding (e.g., by hybridization) sites for a biochemical sample (target) which often has undetermined characteristics. In one embodiment, a microarray refers to an assembly of distinct polynucleotide or oligonucleotide probes immobilized at defined positions on a substrate. Arrays may be formed on substrates fabricated with materials such as paper, glass, plastic (e.g., polypropylene, nylon, polystyrene), polyacrylamide, nitrocellulose, silicon, optical fiber or any other suitable solid or semi-solid support, and configured in a planar (e.g., glass plates, silicon chips) or three-dimensional (e.g., pins, fibers, beads, particles, microtiter wells, capillaries) configuration. Probes forming the arrays may be attached to the substrate by any number of ways including (i) in situ synthesis (e.g., high-density oligonucleotide arrays) using photolithographic techniques (see, Fodor et al., Science (1991), 251:767-773; Pease et al., Proc. Natl. Acad. Sci. U.S.A. (1994), 91:5022-5026; Lockhart et al., Nature Biotechnology (1996), 14:1675; U.S. Pat. Nos. 5,578,832; 5,556,752; and 5,510,270); (ii) spotting/printing at medium to low-density (e.g., cDNA probes) on glass, nylon or nitrocellulose (Schena et al, Science (1995), 270:467-470, DeRisi et al, Nature Genetics (1996), 14:457-460; Shalon et al., Genome Res. (1996), 6:639-645; and Schena et al., Proc. Natl. Acad Sci. U.S.A. (1995), 93:10539-11286); (iii) by masking (Maskos and Southern, Nuc. Acids. Res. (1992), 20:1679-1684) and (iv) by dot-blotting on a nylon or nitrocellulose hybridization membrane (see, e.g., Sambrook et al., Eds., 1989, Molecular Cloning: A Laboratory Manual, 2nd ed., Vol. 1-3, Cold Spring Harbor Laboratory (Cold Spring Harbor, N.Y.)). Probes may also be noncovalently immobilized on the substrate by hybridization to anchors, by means of magnetic beads, or in a fluid phase such as in microtiter wells or capillaries. The probe molecules are generally nucleic acids such as DNA, RNA, PNA, and cDNA but may also include proteins, polypeptides, oligosaccharides, cells, tissues and any permutations thereof which can specifically bind the target molecules.


For example, microarrays, in which either defined cDNAs or oligonucleotides are immobilized at discrete locations on, for example, solid or semi-solid substrates, or on defined particles, enable the detection and/or quantification of the expression of a multitude of genes in a given specimen.


Several techniques are well-known in the art for attaching nucleic acids to a solid substrate such as a glass slide. One method is to incorporate modified bases or analogs that contain a moiety that is capable of attachment to a solid substrate, such as an amine group, a derivative of an amine group or another group with a positive charge, into the amplified nucleic acids. The amplified product is then contacted with a solid substrate, such as a glass slide, which is coated with an aldehyde or another reactive group which will form a covalent link with the reactive group that is on the amplified product and become covalently attached to the glass slide. Microarrays comprising the amplified products can be fabricated using a Biodot (BioDot, Inc. Irvine, Calif.) spotting apparatus and aldehyde-coated glass slides (CEL Associates, Houston, Tex.). Amplification products can be spotted onto the aldehyde-coated slides, and processed according to published procedures (Schena et al., Proc. Natl. Acad. Sci. U.S.A. (1995) 93:10614-10619). Arrays can also be printed by robotics onto glass, nylon (Ramsay, G., Nature Biotechnol. (1998), 16:40-44), polypropylene (Matson, et al., Anal Biochem. (1995), 224(l):110-6), and silicone slides (Marshall, A. and Hodgson, J., Nature Biotechnol. (1998), 16:27-31). Other approaches to array assembly include fine micropipetting within electric fields (Marshall and Hodgson, supra), and spotting the polynucleotides directly onto positively coated plates. Methods such as those using amino propyl silicon surface chemistry are also known in the art, as disclosed at www.cmt.corning.com and http://cmgm.stanford.edu/pbrown/.


One method for making microarrays is by making high-density polynucleotide arrays. Techniques are known for rapid deposition of polynucleotides (Blanchard et al., Biosensors & Bioelectronics, 11:687-690). Other methods for making microarrays, e.g., by masking (Maskos and Southern, Nuc. Acids. Res. (1992), 20:1679-1684), may also be used. In principle, and as noted above, any type of array, for example, dot blots on a nylon hybridization membrane, could be used. However, as will be recognized by those skilled in the art, very small arrays will frequently be preferred because hybridization volumes will be smaller.


In one embodiment, the invention provides an array comprising at least two isolated polynucleotide molecules, wherein each isolated polynucleotide molecule detects an expressed gene product of a gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927, and wherein the gene is differentially expressed in atherosclerotic disease in a mammal. In one embodiment, the invention provides an array comprising at least two isolated polynucleotide molecules, wherein each isolated polynucleotide molecule detects an expressed gene product of a gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs:1-927, and wherein the gene is differentially expressed in atherosclerotic disease in a mammal. In various embodiments, an array in accordance with the invention comprises any of at least 2, 3, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, or 100 polynucleotides each comprising at least a portion of a polynucleotide depicted in the Sequence Listing or a polynucleotide complement thereof.


In another embodiment, the invention provides an array comprising at least two antibody molecules or antigen binding fragments thereof, wherein each antibody molecule or antigen binding fragment thereof detects an expressed gene product of a gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927, and wherein the gene is differentially expressed in atherosclerotic disease in a mammal. In another embodiment, the invention provides an array comprising at least two antibody molecules or antigen binding fragments thereof, wherein each antibody molecule or antigen binding fragment thereof detects an expressed gene product of a gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs:1-927, and wherein the gene is differentially expressed in atherosclerotic disease in a mammal. In various embodiments, an antibody array in accordance with the invention comprises any of at least 2, 3, 5, 10, 15, 20, 25, 30, 40, 50, 60, 70, 80, 90, or 100 antibodies or antigen binding fragments thereof each recognizing an expression product (e.g., a polypeptide) of a gene corresponding to a polynucleotide sequence depicted in the Sequence Listing.


Methods of the Invention


Methods for Detecting Gene Expression


The invention provides methods for detecting gene expression, comprising contacting products of gene expression (e.g., mRNA, protein) in a sample with a system for detecting gene expression as described above, and detecting interaction between the products of gene expression in the sample and the system for detecting gene expression. The methods for detecting gene expression described herein may be used to detect or quantify differential expression and/or for expression profiling of a sample. As used herein, “differential expression” refers to increased (upregulated) or decreased (downregulated) production of an expressed product of a gene (e.g., mRNA, protein). Differential expression may be assessed qualitatively (presence or absence of a gene product) and/or quantitatively (change in relative amount, i.e., increase or decrease, of a gene product).


In one embodiment, MRNA from a sample is contacted with a system for detecting gene expression comprising isolated polynucleotide molecules as described above, and hybridization complexes formed, if any, between the mRNA in the sample and the polynucleotide sequences of the system for detecting gene expression, are detected. In other embodiments, the mRNA is converted to nucleic acid derived from the mRNA, for example, cDNA, and/or amplified, prior to contact with the system for detecting gene expression.


In another embodiment, polypeptides from a sample are contacted with a system for detecting gene expression comprising antibodies or antigen fragments thereof that bind to polypeptide expression products of genes corresponding to the polynucleotide sequences described herein, and binding between the antibodies and polypeptides in the sample, if any, is detected.


Methods for Expression Profiling


An “expression profile” or “molecular signature” is a representation of gene expression in a sample, for example, evaluation of presence, absence, or amount of a plurality of gene expression products, such as mRNA transcripts, or polypeptide translation products of mRNA transcripts. Expression patterns constitute a set of relative or absolute expression values for a number of RNA or protein products corresponding to the plurality of genes evaluated, referred to as the subject's “expression profile” for those nucleotide sequences. In various embodiments, expression patterns corresponding to at least about 2, 5, 10, 20, 30, 50, 100, 200, or 500, or more nucleotide sequences are obtained. The expression pattern for each differentially expressed component member of the expression profile may provide a specificity and sensitivity with respect to predictive value, e.g., for diagnosis, prognosis, monitoring treatment, etc. In some embodiments, a molecular signature is determined by a statistical algorithm that determines the optimal relation between patterns of expression for various genes.


In some embodiments, an expression profile from an individual is compared with a reference expression profile to determine, for example, presence or absence of a disease condition, symptom, or criterion, extent of progression of disease, effectiveness of treatment of disease, or prognosis for prophylaxis, therapy, or cure of disease.


As used herein, the term “subject” refers to an individual regardless of health and/or disease status. For example, a subject may be a patient, a study participant, a control subject, a screening subject, or any other class of individual from whom a sample is obtained and assessed in the context of the invention. Accordingly, a subject may be diagnosed with a disease, can present with one or more symptom of a disease, or may have a predisposing factor, such as a genetic or medical history factor, for a disease. Alternatively, a subject may be healthy with respect to any of the aforementioned disease factors or criteria. It will be appreciated that the term “healthy” as used herein, is relative to a specified disease condition, factor, or criterion. Thus, an individual described as healthy with reference to any specified disease or disease criterion, can be diagnosed with any other one or more disease, or may exhibit any other one or more disease criterion.


Methods for Obtaining Expression Data


Numerous methods for obtaining expression data are known, and any one or more of these techniques, singly or in combination, are suitable for determining expression profiles in the context of the present invention. For example, expression patterns can be evaluated by northern analysis, PCR, RT-PCR, Taq Man analysis, FRET detection, monitoring one or more molecular beacon, hybridization to an oligonucleotide array, hybridization to a CDNA array, hybridization to a polynucleotide array, hybridization to a liquid microarray, hybridization to a microelectric array, molecular beacons, cDNA sequencing, clone hybridization, cDNA fragment fingerprinting, serial analysis of gene expression (SAGE), subtractive hybridization, differential display and/or differential screening (see, e.g., Lockhart and Winzeler (2000) Nature 405:827-836, and references cited therein).


For example, specific PCR primers are designed to a member(s) of a candidate nucleotide library (e.g., a polynucleotide member of a system for detecting gene expression). cDNA is prepared from subject sample RNA by reverse transcription from a poly-dT oligonucleotide primer, and subjected to PCR. Double stranded cDNA may be prepared using primers suitable for reverse transcription of the PCR product, followed by amplification of the cDNA using in vitro transcription. The product of in vitro transcription is a sense-RNA corresponding to the original member(s) of the candidate library. PCR product may be also be evaluated in a number of ways known in the art, including real-time assessment using detection of labeled primers, e.g. TaqMan or molecular beacon probes. Technology platforms suitable for analysis of PCR products include the ABI 7700, 5700, or 7000 Sequence Detection Systems (Applied Biosystems, Foster City, Calif.), the MJ Research Opticon (MJ Research, Waltham, Mass.), the Roche Light Cycler (Roche Diagnostics, Indianapolis, Ind.), the Stratagene MX4000 (Stratagene, La Jolla, Calif.), and the Bio-Rad iCycler (Bio-Rad Laboratories, Hercules, Calif.). Alternatively, molecular beacons are used to detect presence of a nucleic acid sequence in an unamplified RNA or CDNA sample, or following amplification of the sequence using any method, e.g., IVT (in vitro transcription) or NASBA (nucleic acid sequence based amplification). Molecular beacons are designed with sequences complementary to member(s) of a candidate nucleotide library, and are linked to fluorescent labels. Each probe has a different fluorescent label with non-overlapping emission wavelengths. For example, expression of ten genes may be assessed using ten different sequence-specific molecular beacons.


Alternatively, or in addition, molecular beacons are used to assess expression of multiple nucleotide sequences simultaneously. Molecular beacons with sequences complimentary to the members of a diagnostic nucleotide set are designed and linked to fluorescent labels. Each fluorescent label used must have a non-overlapping emission wavelength. For example, 10 nucleotide sequences can be assessed by hybridizing 10 sequence specific molecular beacons (each labeled with a different fluorescent molecule) to an amplified or non-amplified RNA or cDNA sample. Such an assay bypasses the need for sample labeling procedures.


Alternatively, or in addition, bead arrays can be used to assess expression of multiple sequences simultaneously (see, e.g., LabMAP 100, Luminex Corp, Austin, Tex.). Alternatively, or in addition, electric arrays can be used to assess expression of multiple sequences, as exemplified by the e-Sensor technology of Motorola (Chicago, Ill.) or Nanochip technology of Nanogen (San Diego, Calif.).


Of course, the particular method elected will be dependent on such factors as quantity of RNA recovered, practitioner preference, available reagents and equipment, detectors, and the like. Typically, however, the elected method(s) will be appropriate for processing the number of samples and probes of interest. Methods for high-throughput expression analysis are discussed below.


Alternatively, expression at the level of protein products of gene expression is performed. For example, protein expression in a sample can be evaluated by one or more method selected from among: western analysis, two-dimensional gel analysis, chromatographic separation, mass spectrometric detection, protein-fusion reporter constructs, calorimetric assays, binding to a protein array (e.g., antibody array), and characterization of polysomal mRNA. One particularly favorable approach involves binding of labeled protein expression products to an array of antibodies specific for members of the candidate library. Methods for producing and evaluating antibodies are well known in the art, see, e.g., Coligan, supra; and Harlow and Lane (1989) Antibodies: A Laboratory Manual, Cold Spring Harbor Press, NY (“Harlow and Lane”). Additional details regarding a variety of immunological and immunoassay procedures adaptable to the present invention by selection of antibody reagents specific for the products of candidate nucleotide sequences can be found in, e.g., Stites and Terr (eds.) (1991) Basic and Clinical Immunology, 7th ed. Another approach uses systems for performing desorption spectrometry. Commercially available systems, e.g., from Ciphergen Biosystems, Inc. (Fremont, Calif.) are particularly well suited to quantitative analysis of protein expression. Protein Chip.RTM. arrays (see, e.g., the website, ciphergen.com) used in desorption spectrometry approaches provide arrays for detection of protein expression. Alternatively, affinity reagents, (e.g., antibodies, small molecules, etc.) may be developed that recognize epitopes of one or more protein products. Affinity assays are used in protein array assays, e.g., to detect the presence or absence of particular proteins. Alternatively, affinity reagents are used to detect expression using the methods described above. In the case of a protein that is expressed on a cell surface, labeled affinity reagents are bound to a sample, and cells expressing the protein are identified and counted using fluorescent activated cell sorting (FACS).


High Throughput Expression Assays


A number of suitable high throughput formats exist for evaluating gene expression. Typically, the term high throughput refers to a format that performs at least about 100 assays, or at least about 500 assays, or at least about 1000 assays, or at least about 5000 assays, or at least about 10,000 assays, or more per day. When enumerating assays, either the number of samples or the number of candidate nucleotide sequences evaluated can be considered. For example, a northern analysis of, e.g., about 100 samples performed in a gridded array, e.g., a dot blot, using a single probe corresponding to a polynucleotide sequence as described herein can be considered a high throughput assay. More typically, however, such an assay is performed as a series of duplicate blots, each evaluated with a distinct probe corresponding to a different polynucleotide sequence of a system for detecting gene expression. Alternatively, methods that simultaneously evaluate expression of about 100 or more polynucleotide sequences in one or more samples, or in multiple samples, are considered high throughput.


Numerous technological platforms for performing high throughput expression analysis are known. Generally, such methods involve a logical or physical array of either the subject samples, or the candidate library, or both. Common array formats include both liquid and solid phase arrays. For example, assays employing liquid phase arrays, e.g., for hybridization of nucleic acids, binding of antibodies or other receptors to ligand, etc., can be performed in multiwell, or microtiter, plates. Microtiter plates with 96, 384 or 1536 wells are widely available, and even higher numbers of wells, e.g., 3456 and 9600 can be used. In general, the choice of microtiter plates is determined by the methods and equipment, e.g., robotic handling and loading systems, used for sample preparation and analysis. Exemplary systems include, e.g., the ORCA.TM. system from Beckman-Coulter, Inc. (Fullerton, Calif.) and the Zymate systems from Zymark Corporation (Hopkinton, Mass.).


Alternatively, a variety of solid phase arrays can favorably be employed to determine expression patterns in the context of the invention. Exemplary formats include membrane or filter arrays (e.g., nitrocellulose, nylon), pin arrays, and bead arrays (e.g., in a liquid “slurry”). Typically, probes corresponding to nucleic acid or protein reagents that specifically interact with (e.g., hybridize to or bind to) an expression product corresponding to a member of the candidate library, are immobilized, for example by direct or indirect cross-linking, to the solid support. Essentially any solid support capable of withstanding the reagents and conditions necessary for performing the particular expression assay can be utilized. For example, functionalized glass, silicon, silicon dioxide, modified silicon, any of a variety of polymers, such as (poly)tetrafluoroethylene, (poly)vinylidenedifluoride, polystyrene, polycarbonate, or combinations thereof can all serve as the substrate for a solid phase array.


In one embodiment, the array is a “chip” composed, e.g., of one of the above-specified materials. Polynucleotide probes, e.g., RNA or DNA, such as cDNA, synthetic oligonucleotides, and the like, or binding proteins such as antibodies or antigen-binding fragments or derivatives thereof, that specifically interact with expression products of individual components of the candidate library are affixed to the chip in a logically ordered manner, i.e., in an array. In addition, any molecule with a specific affinity for either the sense or anti-sense sequence of the marker nucleotide sequence (depending on the design of the sample labeling), can be fixed to the array surface without loss of specific affinity for the marker and can be obtained and produced for array production, for example, proteins that specifically recognize the specific nucleic acid sequence of the marker, ribozymes, peptide nucleic acids (PNA), or other chemicals or molecules with specific affinity.


Detailed discussion of methods for linking nucleic acids and proteins to a chip substrate, are found in, e.g., U.S. Pat. No. 5,143,854, “Large Scale Photolithographic Solid Phase Synthesis Of Polypeptides And Receptor Binding Screening Thereof,” to Pirrung et al., issued, Sep. 1, 1992; U.S. Pat. No. 5,837,832, “Arrays Of Nucleic Acid Probes On Biological Chips,” to Chee et al., issued Nov. 17, 1998; U.S. Pat. No. 6,087,112, “Arrays With Modified Oligonucleotide And Polynucleotide Compositions,” to Dale, issued Jul. 11, 2000; U.S. Pat. No. 5,215,882, “Method Of Immobilizing Nucleic Acid On A Solid Substrate For Use In Nucleic Acid Hybridization Assays,” to Bahl. et al., issued Jun. 1, 1993; U.S. Pat. No. 5,707,807, “Molecular Indexing For Expressed Gene Analysis,” to Kato, issued Jan. 13, 1998; U.S. Pat. No. 5,807,522, “Methods For Fabricating Microarrays Of Biological Samples,” to Brown et al., issued Sep. 15, 1998; U.S. Pat. No. 5,958,342, “Jet Droplet Device,” to Gamble et al., issued Sep. 28, 1999; U.S. Pat. No. 5,994,076, “Methods Of Assaying Differential Expression,” to Chenchik et al., issued Nov. 30, 1999; U.S. Pat. No. 6,004,755, “Quantitative Microarray Hybridization Assays,” to Wang, issued Dec. 21, 1999; U.S. Pat. No. 6,048,695, “Chemically Modified Nucleic Acids And Method For Coupling Nucleic Acids To Solid Support,” to Bradley et al., issued Apr. 11, 2000; U.S. Pat. No. 6,060,240, “Methods For Measuring Relative Amounts Of Nucleic Acids In A Complex Mixture And Retrieval Of Specific Sequences Therefrom,” to Kamb et al., issued May 9, 2000; U.S. Pat. No. 6,090,556, “Method For Quantitatively Determining The Expression Of A Gene,” to Kato, issued Jul. 18, 2000; and U.S. Pat. No. 6,040,138, “Expression Monitoring By Hybridization To High Density Oligonucleotide Arrays,” to Lockhart et al., issued Mar. 21, 2000.


For example, cDNA inserts corresponding to candidate nucleotide sequences, in a standard TA cloning vector, are amplified by a polymerase chain reaction for approximately 30-40 cycles. The amplified PCR products are then arrayed onto a glass support by any of a variety of well-known techniques, e.g., the VSLIPS.TM. technology described in U.S. Pat. No. 5,143,854. RNA, or cDNA corresponding to RNA, isolated from a subject sample, is labeled, e.g., with a fluorescent tag, and a solution containing the RNA (or cDNA) is incubated under conditions favorable for hybridization, with the “probe” chip. Following incubation, and washing to eliminate non-specific hybridization, the labeled nucleic acid bound to the chip is detected qualitatively or quantitatively, and the resulting expression profile for the corresponding candidate nucleotide sequences is recorded. Multiple cDNAs from a nucleotide sequence that are non-overlapping or partially overlapping may also be used.


In another approach, oligonucleotides corresponding to members of a candidate nucleotide library are synthesized and spotted onto an array. Alternatively, oligonucleotides are synthesized onto the array using methods known in the art, e.g. Hughes, et al. supra. The oligonucleotide is designed to be complementary to any portion of the candidate nucleotide sequence. In addition, in the context of expression analysis for, e.g. diagnostic use of diagnostic nucleotide sets, an oligonucleotide can be designed to exhibit particular hybridization characteristics, or to exhibit a particular specificity and/or sensitivity, as further described below.


Oligonucleotide probes may be designed on a contract basis by various companies (for example, Compugen, Mergen, Affymetrix, Telechem), or designed from the candidate sequences using a variety of parameters and algorithms as indicated at the website genome.wi.mit.edu/cgi-bin/prtm-er/primer3.cgi. Briefly, the length of the oligonucleotide to be synthesized is determined, preferably at least 16 nucleotides, generally 18-24 nucleotides, 24-70 nucleotides and, in some circumstances, more than 70 nucleotides. The sequence analysis algorithms and tools described above are applied to the sequences to mask repetitive elements, vector sequences and low complexity sequences. Oligonucleotides are selected that are specific to the candidate nucleotide sequence (based on a Blast n search of the oligonucleotide sequence in question against gene sequences databases, such as the Human Genome Sequence, UniGene, dbEST or the non-redundant database at NCBI), and have<50% G content and 25-70% G+C content. Desired oligonucleotides are synthesized using well-known methods and apparatus, or ordered from a commercial supplier.


A hybridization signal may be amplified using methods known in the art, and as described herein, for example use of the Clontech kit (Glass Fluorescent Labeling Kit), Stratagene kit (Fairplay Microarray Labeling Kit), the Micromax kit (New England Nuclear, Inc.), the Genisphere kit (3DNA Submicro), linear amplification, e.g., as described in U.S. Pat. No. 6,132,997 or described in Hughes, T R, et al. (2001) Nature Biotechnology 19:343-347 (2001) and/or Westin et al. (2000) Nat Biotech. 18:199-204. In some cases, amplification techniques do not increase signal intensity, but allow assays to be done with small amounts of RNA.


Alternatively, fluorescently labeled cDNA are hybridized directly to the microarray using methods known in the art. For example, labeled cDNA are generated by reverse transcription using Cy3-and Cy5-conjugated deoxynucleotides, and the reaction products purified using standard methods. It is appreciated that the methods for signal amplification of expression data useful for identifying diagnostic nucleotide sets are also useful for amplification of expression data for diagnostic purposes.


Microarray expression may be detected by scanning the microarray with a variety of laser or CCD-based scanners, and extracting features with numerous software packages, for example, Imagene (Biodiscovery), Feature Extraction Software (Agilent), Scanalyze (Eisen, M. 1999. SCANALYZE User Manual; Stanford Univ., Stanford, Calif. Ver 2.32.), GenePix (Axon Instruments).


In another approach, hybridization to microelectric arrays is performed, e.g., as described in Umek et al (2001) J Mol Diagn. 3:74-84. An affinity probe, e.g., DNA, is deposited on a metal surface. The metal surface underlying each probe is connected to a metal wire and electrical signal detection system. Unlabelled RNA or cDNA is hybridized to the array, or alternatively, RNA or cDNA sample is amplified before hybridization, e.g., by PCR. Specific hybridization of sample RNA or cDNA results in generation of an electrical signal, which is transmitted to a detector. See Westin (2000) Nat Biotech. 18:199-204 (describing anchored multiplex amplification of a microelectronic chip array); Edman (1997) NAR 25:4907-14; Vignali (2000) J Immunol Methods 243:243-55.


Evaluation of Expression Patterns


Expression patterns can be evaluated by qualitative and/or quantitative measures. Certain of the above described techniques for evaluating gene expression (e.g., as RNA or protein products) yield data that are predominantly qualitative in nature, i.e., the methods detect differences in expression that classify expression into distinct modes without providing significant information regarding quantitative aspects of expression. For example, a technique can be described as a qualitative technique if it detects the presence or absence of expression of a candidate nucleotide sequence, i.e., an on/off pattern of expression. Alternatively, a qualitative technique measures the presence (and/or absence) of different alleles, or variants, of a gene product.


In contrast, some methods provide data that characterize expression in a quantitative manner. That is, the methods relate expression on a numerical scale, e.g., a scale of 0-5, a scale of 1-10, a scale of +-+++, from grade 1 to grade 5, a grade from a to z, or the like. It will be understood that the numerical, and symbolic examples provided are arbitrary, and that any graduated scale (or any symbolic representation of a graduated scale) can be employed in the context of the present invention to describe quantitative differences in nucleotide sequence expression. Typically, such methods yield information corresponding to a relative increase or decrease in expression.


Any method that yields either quantitative or qualitative expression data is suitable for evaluating expression of candidate nucleotide sequences in a subject sample. In some cases, e.g., when multiple methods are employed to determine expression patterns for a plurality of candidate nucleotide sequences, the recovered data, e.g., the expression profile, for the nucleotide sequences is a combination of quantitative and qualitative data.


In some embodiments, qualitative and/or quantitative expression data from a sample is compared with a reference molecular signature that is indicative of, for example, presence or absence of a disease condition, symptom, or criterion, extent of progression of disease, effectiveness of treatment of disease, or prognosis for prophylaxis, therapy, or cure of disease. The reference molecular signature may be from a reference healthy individual (e.g., an individual who does not exhibit symptoms of the disease condition to be evaluated) or an individual with a disease condition for comparison with the sample (e.g., an individual with the same or different stage of disease for comparison with the individual being evaluated, or with a genotype or phenotype that indicates, for example, prognosis for successful treatment), or the reference molecular signature may be established from a compilation of data from multiple individuals


In some applications, expression of a plurality of candidate polynucleotide sequences is evaluated sequentially. This is typically the case for methods that can be characterized as low-to moderate throughput. In contrast, as the throughput of the elected assay increases, expression for the plurality of candidate polynucleotide sequences in a sample or multiple samples is typically assayed simultaneously. Again, the methods (and throughput) are largely determined by the individual practitioner, although, typically, it is preferable to employ methods that permit rapid, e.g. automated or partially automated, preparation and detection, on a scale that is time-efficient and cost-effective.


Genotyping


In addition to, or in conjunction with, the correlation of expression profiles and clinical data, it is often desirable to correlate expression patterns with a subject's genotype at one or more genetic loci or to correlate both expression profiles and genetic loci data with clinical data. The selected loci can be, for example, chromosomal loci corresponding to one or more member of the candidate library, polymorphic alleles for marker loci, or alternative disease related loci (not contributing to the candidate library) known to be, or putatively associated with, a disease (or disease criterion). Indeed, it will be appreciated that where a (polymorphic) allele at a locus is linked to a disease (or to a predisposition to a disease), the presence of the allele can itself be a disease criterion.


Numerous well known methods exist for evaluating the genotype of an individual, including southern analysis, restriction fragment length polymorphism (RFLP) analysis, polymerase chain reaction (PCR), amplification length polymorphism (AFLP) analysis, single stranded conformation polymorphism (SSCP) analysis, single nucleotide polymorphism (SNP) analysis (e.g., via PCR, Taqman or molecular beacons), among many other useful methods. Many such procedures are readily adaptable to high throughput and/or automated (or semi-automated) sample preparation and analysis methods. Often, these methods can be performed on nucleic acid samples recovered via simple procedures from the same sample as yielded the material for expression profiling. Exemplary techniques are described in, e.g., Sambrook, and Ausubel, supra.


Samples


Samples which may be evaluated for differential expression of the polynucleotide sequences described herein include any blood vessel or portion thereof with atherosclerotic and/or inflammatory disease. Such blood vessels include, but are not limited to, the aorta, a coronary artery, the carotid artery, and peripheral blood vessels such as, for example, iliac or femoral arteries. In one embodiment, the sample is derived from an arterial biopsy. In another embodiment, the sample is derived from an atherectomy. Samples may also be derived from peripheral blood cells or serum.


Samples may be stabilized for storage by addition of reagents such as Trizol. Total RNA and/or protein may be isolated using standard techniques known in the art for expression profiling experiments.


Methods for RNA isolation include those described in standard molecular biology textbooks. Commercially available kits such as those provided by Qiagen (RNeasy Kits) may also be used for RNA isolation.


Methods for Diagnosing Atherosclerotic Disease


The invention provides methods for diagnosing an atherosclerotic disease condition in an individual. Diagnosis includes, for example, determining presence or absence of a disease condition or a symptom of a disease condition in an individual who has, who is suspected of having, or who may be suspected of being predisposed to an atherosclerotic disease. In accordance with methods of the invention for diagnosing atherosclerotic disease, gene expression products (e.g., RNA or proteins) from a sample from an individual are contacted with a system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a test sample are compared with levels of expression in a molecular signature that is indicative of presence or absence of an atherosclerotic disease condition for which diagnosis is desired. To obtain a diagnosis, the levels of gene expression in a sample may be compared to one or more than one molecular signature, each of which may be indicative of presence or absence one or more than one atherosclerotic disease condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g., mRNA or polynucleotides derived from mRNA, for example cDNA) are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of presence or absence of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of presence or absence of a disease condition, criterion, or symptom for which diagnosis is desired.


In some embodiments, polypeptides derived from a sample from an individual are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of presence or absence of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of presence or absence of a disease condition, criterion, or symptom for which diagnosis is desired.


Methods for Assessing Extent of Progression of Atherosclerotic Disease


The invention provides methods for assessing extent of progression of an atherosclerotic disease condition in an individual. For example, a stage to which a disease condition or particular symptom has progressed may be assessed. In accordance with methods of the invention for assessing extent of progression of atherosclerotic disease, gene expression products (e.g., RNA or proteins) from a sample from an individual are contacted with a system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a test sample are compared with levels of expression in a molecular signature that is indicative of extent of progression of an atherosclerotic disease condition for which assessment is desired. The levels of gene expression may be compared to one or more than one molecular signature, each of which may be indicative of extent of progression of one or more than one atherosclerotic disease condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g., mRNA or polynucleotides derived from mRNA, for example CDNA) are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of extent of progression of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of extent of progression of a disease condition for which diagnosis is desired.


In some embodiments, polypeptides derived from a sample from an individual are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of extent of progression of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of extent of progression of a disease condition for which diagnosis is desired.


Methods for Assessing Efficacy of Treatment of Atherosclerotic Disease


The invention provides methods for assessing extent of progression of an atherosclerotic disease condition in an individual. For example, a stage to which a disease condition or particular symptom has progressed may be assessed by the methods of the invention. In accordance with methods of the invention for assessing extent of progression of atherosclerotic disease, gene expression products (e.g., RNA or proteins) from a sample from an individual are contacted with the system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a test sample are compared with levels of expression in a molecular signature that is indicative of extent of progression of an atherosclerotic disease condition for which assessment is desired. The levels of gene expression may be compared to one or more than one molecular signature, each of which may be indicative of extent of progression of one or more than one atherosclerotic disease condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g, mRNA or polynucleotides derived from mRNA, for example cDNA) are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of extent of progression of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of extent of progression of a disease condition for which assessment is desired.


In some embodiments, polypeptides derived from a sample from an individual are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of extent of progression of an atherosclerotic disease in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of extent of progression of a disease condition for which assessment is desired.


Methods for Assessing Efficacy of Treatment


The invention provides methods for assessing efficacy of treatment of an atherosclerotic disease symptom or condition in an individual. As used herein, “efficacy of treatment” refers to achievement of a desired therapeutic outcome (e.g., reduction or elimination of one or more symptoms of atherosclerotic disease). “Treatment” as used herein may refer to prophylaxis, therapy, or cure with respect to one or more symptoms of an atherosclerotic disease or condition. Treatment includes administration of one or more compounds or biological substances with potential therapeutic benefit and/or alterations in environmental factors, such as, for example, diet and/or exercise. In one embodiment, administration of the one or more compounds or biological substances comprises administration via a medical device such as, for example, a drug eluting stent. In other embodiments, treatment may include gene therapy or any other method that alters expression of the polynucleotide sequences described herein. In accordance with methods of the invention for assessing efficacy of treatment of atherosclerotic disease, gene expression products (e.g., RNA or proteins) from a sample from an individual are contacted with a system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a test sample are compared with levels of expression in a molecular signature that is indicative of efficacy of treatment of an atherosclerotic disease symptom or condition for which assessment is desired. The levels of gene expression may be compared to one or more than one molecular signature, each of which may be indicative of extent of effectiveness of treatment of one or more than one atherosclerotic disease symptom or condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g., mRNA or polynucleotides derived from mRNA, for example cDNA) are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of efficacy of treatment of an atherosclerotic disease symptom or condition in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of efficacy of treatment of a disease symptom or condition for which assessment is desired.


In some embodiments, polypeptides derived from a sample from an individual are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of efficacy of treatment of an atherosclerotic disease condition in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of efficacy of treatment of a disease condition for which assessment is desired.


Methods for Identifying Compounds Effective for Treatment of Atherosclerotic Disease


The invention provides methods for identifying compounds effective for treatment of an atherosclerotic disease symptom or condition in an individual. In accordance with methods of the invention for identifying compounds effective for treatment of atherosclerotic disease, at least one test compound (i.e., one or more than one test compound) is administered, for example as a pharmaceutical composition comprising the at least one test compound and a pharmaceutically acceptable excipient, to an individual with an atherosclerotic disease symptom or condition or suspected of having an atherosclerotic disease symptom or condition, or to an individual who is predisposed to or suspected of being predisposed to development of an atherosclerotic disease symptom or condition. Gene expression products (e.g., RNA or proteins) from a sample from the individual are contacted with a system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a test sample from the individual to whom the at least one test compound has been administered are compared with levels of expression in a molecular signature that is indicative of efficacy of treatment of the atherosclerotic disease symptom or condition for which assessment is desired. The levels of gene expression may be compared to one or more than one molecular signature, each of which may be indicative of extent of effectiveness of treatment of one or more than one atherosclerotic disease symptom or condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g., mRNA or polynucleotides derived from mRNA, for example cDNA) to whom at least one test compound has been administered are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of efficacy of treatment of an atherosclerotic disease symptom or condition in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of efficacy of treatment of a disease symptom or condition for which assessment is desired.


In some embodiments, polypeptides derived from a sample from an individual to whom at least one test compound has been administered are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of efficacy of treatment of an atherosclerotic disease condition in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of efficacy of treatment of a disease condition for which assessment is desired.


Methods for Determining prognosis of Atherosclerotic Disease


The invention provides methods for determining prognosis of atherosclerotic disease in an individual, comprising contacting polynucleotides derived from a sample from the individual with a system for detecting gene expression as described above. “Prognosis” as used herein refers to the probability that an individual will develop an atherosclerotic disease symptom or condition, or that atherosclerotic disease will progress in an individual who has an atherosclerotic disease. Prognosis is a determination or prediction of probable course and/or outcome of a disease condition, i.e., whether an individual will exhibit or develop symptoms of the disease, i.e., a clinical event. In cardiovascular medicine, a common measure of prognosis is (but is not limited to) MACE (major adverse cardiac event). MACE includes mortality as well as morbidity measures, such as myocardial infarction, angina, stroke, rate of revascularization, hospitalization, etc.


For determination of prognosis of atherosclerotic disease, gene expression products (e.g., RNA or proteins) from a sample from an individual are contacted with the system for detecting gene expression as described above. In one embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 133, 745, 806, 824, 886, 882, 901, 905, 913, and 927. In another embodiment, the genes for which expression is detected are selected from the group of genes corresponding to SEQ ID NOs: 1-927.


In some embodiments, qualitative and/or quantitative levels of gene expression in a sample from the individual are compared with levels of expression in a molecular signature that is indicative of prognosis of the atherosclerotic disease symptom or condition for which assessment is desired. The levels of gene expression may be compared to one or more than one molecular signature, each of which may be indicative of prognosis for one or more than one atherosclerotic disease symptom or condition.


In some embodiments, polynucleotides derived from a sample from an individual (e.g., mRNA or polynucleotides derived from mRNA, for example cDNA) are contacted with isolated polynucleotide molecules in a system for detecting gene expression as described above, wherein each isolated polynucleotide molecule detects an expressed product of a gene that is differentially expressed in atherosclerotic disease in a mammal, and hybridization complexes formed, if any, are detected, wherein presence, absence, or amount of hybridization complexes formed from at least one of the isolated polynucleotides is indicative of prognosis for development or progression an atherosclerotic disease symptom or condition in the individual. In some embodiments, presence, absence, or amount of the polynucleotides derived from the sample is compared with presence, absence, or amount of polynucleotides in a molecular signature indicative of prognosis for development or progression of a disease symptom or condition for which assessment is desired.


In some embodiments, polypeptides derived from a sample from an individual are contacted with a system for detecting gene expression as described above which comprises molecules capable of detectably binding to polypeptides that are differentially expressed in atherosclerotic disease, for example, antibodies or antigen binding fragments thereof, that detect expressed polypeptide products of genes corresponding to polynucleotide sequences depicted in the Sequence Listing, wherein presence, absence, or amount of bound polypeptide is indicative of prognosis for development or progression of an atherosclerotic disease symptom or condition in the individual. In some embodiments, presence, absence, or amount of the polypeptides derived from the sample is compared with presence, absence, or amount of polypeptides in a molecular signature indicative of prognosis for development or progression of an atherosclerotic disease symptom or condition for which assessment is desired.


Novel Polynucleotide Sequences


The invention provides novel polynucleotide sequences that are differentially expressed in atherosclerotic disease. We have identified unnamed (not previously described as corresponding to a gene or an expressed gene, and/or for which no function has previously been assigned) polynucleotide sequences herein. The novel differentially expressed nucleotide sequences of the invention are useful in a system for detecting gene expression, such as a diagnostic oligonucleotide set, and are also useful as probes in a diagnostic oligonucleotide set immobilized on an array. The novel polynucleotide sequences may be useful as disease target polynucleotide sequences and/or as imaging reagents as described herein.


As used herein, “novel polynucleotide sequence” refers to (a) a polynucleotide sequence containing at least one of the polynucleotide sequences disclosed herein (as depicted in the Sequence Listing); (b) a polynucleotide sequence that encodes the amino acid sequence encoded by a polynucleotide sequence disclosed herein; (c) a polynucleotide sequence that hybridizes to the complement of a coding sequence disclosed herein under highly stringent conditions, e.g., hybridization to filter-bound DNA in 0.5 M NaHPO4, 7% sodium dodecyl sulfate (SDS), 1 mM EDTA at 65° C., and washing in 0.×SSC/0.1% SDS at 68° C. (Ausubel, F.M. et al., eds. (1989) Current Protocols in Molecular Biology, Vol. 1, Green Publishing Associates, Inc., and John Wiley & Sons, Inc., New York, at p. 2.01.3); (d) a polynucleotide sequence that hybridizes to the complement of a coding sequence disclosed herein under less stringent conditions, such as moderately stringent conditions, e.g., washing in 0.2×SSC/0.1% SDS at 42° C. (Ausubel et al. (1989), supra), yet which still encodes a functionally equivalent gene product; and/or (e) a polynucleotide sequence that is at least 90% identical, at least 80% identical, or at least 70% identical to the coding sequences disclosed herein, wherein % identity is determined using standard algorithms known in the art.


The invention also includes polynucleotide molecules that hybridize to, and are therefore the complements of, novel polynucleotide molecules as described in (a) through (c) in the preceding paragraph. Such hybridization conditions may be highly stringent or less highly stringent, as described above. In instances wherein the polynucleotide molecules are deoxyoligonucleotides, highly stringent conditions may refer to, e.g., washing in 6×SSC/0.05% sodium pyrophosphate at 37° C. (for 14-base oligonucleotides), 48° C. (for 17-base oligonucleotides), 55° C. (for 20-base oligonucleotides, and 60° C. (for 23-base oligonucleotides). These polynucleotide molecules may act as target nucleotide sequence antisense molecules, useful, for example, in target nucleotide sequence regulation and/or as antisense primers in amplification reactions of target nucleic acid sequences. Further, such sequences may be used as part of ribozyme and/or triple helix sequences, also useful for target nucleotide sequence regulation. Such molecules may also be used as components of diagnostic methods whereby the presence of a disease-causing allele may be detected.


The invention also encompasses nucleic acid molecules contained in full-length gene sequences that are related to or derived from novel polynucleotide sequences as described above and as depicted in the Sequence Listing. One sequence may map to more than one full-length gene.


The invention also encompasses (a) polynucleotide vectors that contain any of the foregoing novel polynucleotide sequences and/or their complements; (b) polynucleotide expression vectors that contain any of the foregoing novel polynucleotide sequences and/or their complements; and (c) genetically engineered host cells that contain any of the foregoing novel polynucleotide sequences operatively associated with a regulatory element that directs expression of the polynucleotide in the host cell. As used herein, regulatory elements include, but are not limited to, inducible and non-inducible promoters, enhancers, operators, and other elements known to those skilled in the art that drive and regulate gene expression.


The invention includes fragments of the novel polynucleotide sequences described above. Fragments may be any of at least 5, 10, 15, 20, 25, 50, 100, 200, or 500 nucleotides, or larger.


Novel Polypeptide Products


The invention includes novel polypeptide products, encoded by genes corresponding to the novel polynucleotide sequences described above, or functionally equivalent polypeptide gene products thereof. “Functionally equivalent,” as used herein, refers to a protein capable of exhibiting a substantially similar in vivo function, e.g., activity, as a novel polypeptide gene product encoded by a novel polynucleotide of the invention.


Equivalent novel polypeptide products may include deletions, additions, and/or substitutions of amino acid residues within the amino acid sequence encoded by a gene corresponding to a novel polynucleotide sequence of the invention as described above, but which results in a “silent” change (i.e., a change which does not substantially change the functional properties of the polypeptide). 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.


Novel polypeptide products of genes corresponding to novel polynucleotide sequences described herein may be produced by recombinant nucleic acid technology using techniques that are well known in the art. For example, methods that are well known to those skilled in the art may be used to construct expression vectors containing novel polynucleotide 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. See, for example, the techniques described in Sambrook et al., 1989, supra, and Ausubel et al., 1989, supra. Alternatively, PNA capable of encoding novel nucleotide sequence protein sequences may be chemically synthesized using, for example, synthesizers. See, for example, the techniques described in “Oligonucleotide Synthesis” (1984) Gait, M. J. ed., IRL Press, Oxford. A variety of host-expression vector systems may be utilized to express the novel nucleotide sequence coding sequences of the invention. Ruther et al. (1983) EMBO J 2:1791; Inouye & Inouye (1985) Nucleic Acids Res. 13:3101-3109; Van Heeke & Schuster (1989) J Biol. Chem. 264:5503; Smith et al. (1983) J Virol. 46: 584; Smith, U.S. Pat. No. 4,215,051; Logan & Shenk (1984) Proc. Natl. Acad Sci. USA 81:3655-3659; Bittner et al. (1987) Methods in Enzymol. 153:516-544; Wigler, et al. (1977) Cell 11:223; Szybalska & Szybalski (1962) Proc. Natl. Acad. Sci. USA 48:2026; Lowy, et al. (1980) Cell 22:817; Wigler, et al. (1980) Proc. Natl. Acad. Sci. USA 77:3567; O'Hare, et al. (1981) Proc. Natl. Acad. Sci. USA 78:1527; Mulligan & Berg (1981) Proc. Natl. Acad. Sci. USA 78:2072; Colberre-Garapin, etal. (1981) J Mol. Biol. 150:1; Santerre, etal. (1984) Gene 30:147; Janknecht, etal. (1991) Proc. Natl. Acad. Sci. USA 88: 8972-8976. When recombinant DNA technology is used to produce the protein encoded by a gene corresponding to the novel polynucleotide sequence, it may be advantageous to engineer fusion proteins that can facilitate labeling, immobilization and/or detection.


Antibodies


The invention also provides antibodies or antigen binding fragments thereof that specifically bind to novel polypeptide products encoded by genes that correspond to novel polynucleotide sequences as described above. Antibodies capable of specifically recognizing one or more novel nucleotide sequence epitopes may be prepared by methods that are well known in the art. Such antibodies include, but are not limited to, polyclonal antibodies, monoclonal antibodies (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab')2 fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above. Such antibodies may be used, for example, in the detection of a novel polynucleotide sequence in a biological sample, or, alternatively, as a method for the inhibition of abnormal gene activity, for example, the inhibition of a disease target nucleotide sequence, as further described below. Thus, such antibodies may be utilized as part of a disease treatment method, and/or may be used as part of diagnostic techniques whereby patients may be tested for abnormal levels of novel nucleotide sequence encoded proteins, or for the presence of abnormal forms of the such proteins.


For the production of antibodies that bind to a polypeptide encoded by a novel nucleotide sequence, various host animals may be immunized by injection with a novel protein encoded by the novel nucleotide sequence, or a portion thereof. Such host animals may include, but are not limited to rabbits, mice, and rats. Various adjuvants may be used to increase the immunological response, depending on the host species, including but not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and Corynebacterium parvum.


Polyclonal antibodies are heterogeneous populations of antibody molecules derived from the sera of animals immunized with an antigen, such as novel polypeptide gene product, or an antigenic functional derivative thereof. For the production of polyclonal antibodies, host animals such as those described above, may be immunized by injection with novel polypeptide gene product supplemented with adjuvants as also described above.


Monoclonal antibodies, which are homogeneous populations of antibodies to a particular antigen, may be obtained by any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique of Kohler and Milstein (1975) Nature 256:495-497; and U.S. Pat. No. 4,376,110, the human B-cell hybridoma technique (Kosbor et al. (1983) Immunology Today 4:72; and Cole et al. (1983) Proc. Natl. Acad. Sci. USA 80:2026-2030), and the EBV-hybridoma technique (Cole et al. (1985) Monoclonal Antibodies And Cancer Therapy, Alan R. Liss, Inc., pp. 77-96). Such antibodies may be of any immunoglobulin class including IgG, IgM, IgE, IgA, IgD and any subclass thereof. A hybridoma producing a mAb may be cultivated in vitro or in vivo.


In addition, techniques developed for the production of “chimeric antibodies” by splicing the genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. Morrison et al. (1984) Proc. Natl. Acad. Sci. 81:6851-6855; Neuberger et al. (1984) Nature 312:604-608; Takeda et al. (1985) Nature 314:452-454. A chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region.


Alternatively, techniques described for the production of single chain antibodies can be adapted to produce novel nucleotide sequence-single chain antibodies. (U.S. Pat. No. 4,946,778; Bird (1988) Science 242:423-426; Huston et al. (1988) Proc. NatL. Acad. Sci. USA 85:5879-5883; and Ward et al. (1989) Nature 334:544-546) Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.


Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, such fragments include but are not limited to: the F(ab')2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab')2 fragments. Alternatively, Fab expression libraries may be constructed (Huse et al. (1989) Science 246:1275-1281) to allow rapid and easy identification of monoclonal Fab fragments with a desired specificity.


Disease Specific Target Polynucleotide Sequences


The invention also provides disease specific target polynucleotide sequences, and sets of disease specific target polynucleotide sequences. The diagnostic oligonucleotide sets, individual members of the diagnostic oligonucleotide sets and subsets thereof, and novel polynucleotide sequences, as described above, may also serve as disease specific target polynucleotide sequences. In particular, individual polynucleotide sequences that are differentially regulated or have predictive value that is strongly correlated with an atherosclerotic disease or disease criterion are especially favorable as atherosclerotic disease specific target polynucleotide sequences. Sets of genes that are co-regulated may also be identified as disease specific target polynucleotide sets. Such polynucleotide sequences and/or their complements and/or the expression products of genes corresponding to such polynucleotide sequences (e.g., mRNA, proteins) are targets for modulation by a variety of agents and techniques. For example, disease specific target polynucleotide sequences (or the expression products of genes corresponding to such polynucleotide sequences, or sets of disease specific target polynucleotide sequences) can be inhibited or activated by, e.g., target specific monoclonal antibodies or small molecule inhibitors, or delivery of the polynucleotide sequence or an expression product of a gene corresponding to the polynucleotide sequence to patients. Also, sets of genes can be inhibited or activated by a variety of agents and techniques. The specific usefulness of the target polynucleotide sequence(s) depends on the subject groups from which they were discovered, and the disease or disease criterion with which they correlate.


Kits


The invention provides kits containing a system for detecting gene expression, a diagnostic nucleotide set, candidate nucleotide library, one or novel polynucleotide sequence, one or more polypeptide products of the novel polynucleotide sequences, and/or one or more antibodies that recognize polypeptide expression products of the differentially regulated polynucleotide sequences described herein. A kit may contain a diagnostic nucleotide probe set, or other subset of a candidate library (e.g., as a cDNA, oligonucleotide or antibody microarray or reagents for performing an assay on a diagnostic gene set using any expression profiling technology), packaged in a suitable container. The kit may further comprise one or more additional reagents, e.g., substrates, labels, primers, reagents for labeling expression products, tubes and/or other accessories, reagents for collecting tissue or blood samples, buffers, hybridization chambers, cover slips, etc., and may also contain a software package, e.g., for analyzing differential expression using statistical methods as described herein, and optionally a password and/or account number for accessing the compiled database. The kit optionally further comprises an instruction set or user manual detailing preferred methods of performing the methods of the invention, and/or a reference to a site on the Internet where such instructions may be obtained.

TABLE 1Polynucleotide sequences which detect differentially expressedgenes in atherosclerotic diseaseSEQIDGENEGENECLONEUGCHR_LOCATION6O merNO:CLONE IDSYMBOLNAMENAMECLUSTERPENG [A]SEQUENCE1.C0267B04-3C0267B04-5NC0267B04No chromosomeATGAGCCTAGANIA MouselocationACTCACATGCA7.5 dpc Wholeinfo availableTTTTCCTGACTEmbryo cDNATCTATCATTAGLibrary (Long)AATAAGTTCATMus musculusCAAGAcDNA cloneNIA:C0267B04IMAGE:30017007 5′, MRNAsequence2.M29697.1I17rinterleukin 7M29697Mm.389Chromosome 15CCTATTGTTGAreceptorGTGTCAAACATCACCACTAAGTGGATGGTTATGTAGTCCATTATCCAAA3.L0304D03-3Wnt4wingless-L0304D03Mm.103301Chromosome 4TACCTGAACCArelated MMTVCTCTCTACTGTintegration siteTGTTGTCACAA4GGCAAAAGTGGCATTCCTTCCTCCAAG4.L0237D12-3Cstdcathepsin DL0237D12Mm.231395Chromosome 7CCCTTTGCTGTGTGGGCAGTACTCTGAAGCAGGCAAATGGGTCTTAGGATCCCTCCCAGA5.C0266b08-3BM204200ESTsC0266B08Mm.222000Chromosome 6TCCAAAGATAABM204200AATGAGCAACCGCACTGGCTTAGCCATAGATGACTGACAGTGATTGGAA6.J0537C05-3Pfdn2prefoldin 2J0537C05Mm.10756Chromosome 1TGCCTTGGAGGGCAACAAGGAGCAGATACAGAAGATCATTGAGACACTGTTCACAGCAGC7.L0216F02-3C430008C19RikRIKEN cDNAL0216F02Mm.268474Chromosome 10CATGAATTCCAC430008C19AACCAGTTATTgeneATTAACATGAACCTGAACCTGAACAATTATGACTGTGC8.NM_017372.1LyzslysozymeNM_017372Mm.45436Chromosome 10TTTCTGTCACTGCTCAGGCCAAGGTCTATGAACGTTGTGAGTTTGCCAGAACTCTGAAAA9.C0271B02-34732437J24RikRIKEN cDNAC0271B02Mm.39102Chromosome 4TTCATACCAAG4732437J24GAACCTGACCTgeneCTCTGACAATTGCATTTTGAACATTGTTGTCCCCAAAG10.H3022C10-3AA408868expreexpressedH3022C10Mm.247272Chromosome 16CATTGGAAACAsequenceGACACGTTTGTAA408868AGGCATTTGCGTATTCTTGAAGAGACTGTTTTATGAAT11.L0806E05-3Gtl2GTL2,L0806E05Mm.200506Chromosome 12GTAATGGAGAimprintedATGTATCTGAAmaternallyCCCATATCAAGexpressedCCATCTCTCTTuntranslatedCCTTAACATGTmRNATAAGCA12.H3111E06-5Acas21acetyl-H3111E06Mm.7044Chromosome 2ACACCTCTAACCoenzyme ATCCCAAGAAGsynthetase 2ACGGAGTGAA(AMPTGTCCTCTCCTforming)-likeATCATTT13.H3091H05-3Hras1Harvey ratH3091H05Mm.6793Chromosome 7GTGAGATTCGGsarcoma virusCAGCATAAATToncogene 1GCGGAAACTGAACCCACCCGATGAGAGTGGTCCTGGCT14.K0324B10-3Timp1tissue inhibitorK0324B10Mm.8245Chromosome XTCATAAGGGCTofAAATTCATGGGmetalloproteinaTTCCCCAGAAAse 1TCAACGAGACCACCTTATACCAGCGTT15.K0508B06-3transcribedK0508B06Mm.217234Chromosome 5AAAGACTGAGsequence withAGGAGTCATGmoderateAACCAGGGTAsimilarity toAAACTTATTGGproteinTGCTTTGAGACref:NP_077285.1TTCCAGCA(H. spaiens)A20-bindinginhibitor of NF-kappaBactivation 2;LKB1-interactingprotein [Homosapiens]16.C0176A01-3Syngr1synaptogyrin 1C0176A01Mm.230301Chromosome 15GCAGCATCGCTTCCTTGGTTTATTCTTTGTGTTTGTTCCTTCAGTAAACATTTATTGAGC17.J0748G02-3AU018093J0748G02Chromosome 2TTTTAACGGAGMouse two-cellCCTGAATATAGstage embryoCAGGTTTAAAAcDNA MusTTTAAACAGGTmusculusATAAAATGAAcDNA cloneAAATAAJ0748G02 3′,MRNAsequence18.J0035G10-3C77672ESTs C77672J0035G10Mm.36571Chromosome 4TAGCATGAACCACCATGTTTGGCAATACTGTATTTTAGAAAGAATTAATGGACTGGAGAG19.C0630C02-3Cxcl16chemokine (C-C0630C02Mm.46424Chromosome 11CCTGAGCTCACX-C motif)TGTTTCTCATGligand 16CTGTCTTGAGACAAAGTATCCATATGGAACCTAGGTTA20.K0313A10-35430435G22RikRIKEN cDNAK0313A10Mm.44508Chromosome 1GCTGGTGTTTG5340435G22TGTCAAGAAAgeneATGGCTGAAGCTTGTTTCCAGGCTGTAGGAATGTTGAAC21.L0070E11-3Cbfa2t1HCBFA2T1L0070E11Mm.4909Chromosome 4ACTTAAGTTATidentified geneCTGCATAGAGGhomologCAATCCTCCTG(human)GGTTTGCTTTATGTCTCGAAAATCTAA22.H3072E02-3BG069076ESTsH3072E02Mm.26437Chromosome 12GGGCAAAGGTBG069076ACTTTCTGACAAACTGAGTACCTGAGATCAACCCCCAAGAAGGGAAAAAA23.H3079B06-3Mus musculusH3079B06Mm.295683Chromosome 5ACTATGCAATTunkknownGGACAGATGGmRNAATTACCAAGGAGACTAAAAATATATTCTTTGACTTTGGG24.H3002D08-34833412N02RikRIKEN cDNAH3002D08Mm.195099Chromosome 5TCACTGACCTC4833412N02AACCCCTCCTGgeneCAGAGAAGCCTGAAGACCCCAAAAGCTGCCAGTCCAAA25.H3159A08-3Gp49bglycoprotein 49H3159A08Mm.196617Chromosome 10GATATAATGTGBATAAAGTTCCAAAAGGATCTCTCTGGCTGAAGGAGATACTGGATGGAAC26.C0612F12-3BM207436ESTsC0612F12Mm.260421No ChromosomeCTGAACCCCAABM207436locationTTAATAGCAAAinfo availableGGATATATCTCTCTTCAAAAACGGATAGATTTCTGAAG27.H3108A03-3Apobec1apolipoproteinH3108A03Mm.3333Chromosome 6TTTTGTTCTCTCB editingCATCTGTTAGCCGTTCTGAGGACTGAATGCAGATTGTCAGCTCAAAAA28.C0180G01-3BI076556ESTs BI076556C0180601Mm.37657Chromosome 16GCCAATCTCAGAACCCACATAGAAGGGTCTGCAGTATTATTCCTGTTTCATGTGTGCACA29.C0938A03-3Sf3a1splicing factorC0938A03Mm.156914Chromosome 11AGTGCAAAATT3a, subunit 1TGGTTTGTTGGTGTGCTTTTCTGGTTTAGGAGCCTGAAACAAGCACACT30.J0703E02-3OgdhoxoglutarateJ0703E02Mm.30074Chromosome 11CATGAGTAAGTdehydrogenaseTGTGAAGGCTG(lipoamide)GACCCACATCTTGATACTTGTTTTCTGCATCTTGGGCA31.C0274D12-3transcribedC0274D12Mm.217705Chromosome 12TAGACGTTGTAsequence withAAAAGGAGCCmoderateAAGTTTATCATsimilarity toTTTGTTCCTTAproteinAATCCGTCATApir:S12207TGTGGG(M. musculus)S12207hypotheticalprotein (B2element)-mouse32.H3097H03-3ExpiextracellularH3097H03Mm.1650Chromosome 11ACTGTGGTGACproteinaseAGCTTCCTAACinhibitorGTGTTTGTGTCTAAAATAAACTATCCTTAGCATCCTTC33.H3074D10-3transcribedH3074D10Mm.103987Chromosome 15TATAAATAGAAsequence withAGTGAACCTGTweak similarityAACCTACCACGto proteinGTATCTATCATref:NP_081764.1AACACTAGACT(M. musculus)TTCAGRIKEN cDNA5730493B19[Mus musculus]34.M14222.1Ctsbcathepsin BM14222Mm.22753Chromosome 14CATCCTACAAAGAGGATAAGCACTTTGGGTACACTTCCTACAGCGTGTCTAACAGTGTGA35.C0176G01-32400006H24RikRIKEN cDNAC0176G01Mm.143774Chromosome MultipleCCTGAAAATCT2400006H24MappingsGTCATGTCCACgeneCTTGGAGCCTGAGTAACTTTGAACAGCTGGTAACTAGT36.H3092F08-5UNKNOWN:H3092F08Chromosome 17AGTCAAGGAGSimilar to MusCCTAAAGATTAmusculusTTATGTCAGAGimmediate-AGACCAGCTTTearly antigenAGATACACCCC(E-beta) geneTGAGCApartial intron 2sequence37.H3054F02-31200003C15RikRIKEN cDNAH3054F02Mm.19325Chromosome 10TTATGCTGCAG1200003C15TTTCACTTGGAgeneAAAGGGACAAGGAGCCTTCTATTGTCCCCTGTTTGTAG38.C0012F07-33010021M21RikRIKEN cDNAC0012F07Mm.100525Chromosome 9GTAACCAAGA3010021M21GCCCTGAATAAgeneGGAATTCATTGTAGTAGTGAAAGGGAAACTAATGCTCTT39.L0955A10-39030409G11RikRIKEN cDNAL0955A10Mm.32810Chromosome 4TCCCATGCCTT9030409G11CCCAGAGGGAgeneATTTTAACAATGTAACAATAAATGCTTGGCCTTGAAGCT40.L0045B05-3transcribedL0045b05Mm.182645Chromosome 9AGGACATCTTCsequence withCCAGATCTCAAmoderateAAGAAGAAGAsimilarity toGAGCCTGTAACproteinCACCTCCATGAref:NP_081764.1CCTAAA(M. musculus)RIKEN cDNA5730493B19[Mus musculus]41.H3049A10-3BG066966ESTsH3049A10Mm.262549Chromosome 6TCCTGTGGGAGBG066966ATCCCATAAATCCTGAACCTCACGTAGTGTTACTTTTCCAGGTCATTCT42.X70298.1Sox4SRY-boxX70298Mm.253853Chromosome 13GGACGACGAGcontaining geneTTCGAAGACGA4CCTGCTCGACCTGAACCCCAGCTCAAACTTTGAGAGCAT43.L0001C09-3transcribedL0001C09Mm.171544Chromosome 12GAAGAGATGGsequence withAAGATGGTAGTweak similarityGCCTTGAACACto proteinAGCCACCCAAref:NP_081764.1GCAAAGTTGA(M. musculus)AGAACAGGRIKEN cDNA570493B19[Mus musculus]44.H3010D12-5UNKNOWN:H3010D12Data not foundChromosome 9GCCTGCAGGASimilar to MusGTTTGTGTTGGmusculusTAGCCTCCAAGRIKEN cDNAGAGCTGAAGAT8430421I07GTGCTGAAGATgeneCCAGGCT(8430421I07Rik), mRNA45.C0923E12-3Ptpns1protein tyrosineC0923E12Mm.1682Chromosome 2CTGTCTTCTAAphosphatase,TTCCAAAGGGTnon-receptorTGGTTGGTAAAtype substrate 1GCTCCACCCCCTTTTCCTTTGCCTAAA46.C0941E09-3D330001F17RikRIKEN cDNAC0941E09Mm.123240No ChromosomeTTCACAGGGTTD330001F17locationCCTGGTGTTGCgeneinfo availableATGCAGAGCCTGAACAAAAGACTCAGGTGGACCTGGAA47.K0534C04-3Tce1T-complexK0534C04Mm.41932Chromosome 17TCTACAAGGAAexpressed geneGCATTCAACCA1CCAAGAGGAGCTTGGACCACGTTCACTCTGTATTCTTT48.H3064E11-3BG068254ESTsH3064E11Mm.173544Chromosome 4GGGCCTGAACTBG068354ATGGCTTAATTTACATTAATTAGTTAACATTAATCACACAGTAAGGAGC49.L0957C02-3E130319B15RikRIKEN cDNAL0957C02Mm.149539Chromosome 2TGTGTTGTGATE130319B15TTCAACTCCCAgeneAGACGCCCTTTATGTCCATTCTGGAAAAATACAATAAA50.L0240C12-3ClqaconplementL0240C12Mm.370Chromosome 4ACTGATGTTTCcomponent 1, qTGCACACTGCCsubcomponent,CAGTGGTTTCTalphaTTAAGCACTTTpolypeptideCTGGAATAAACGATCC51.J0018H07-3Rnf149ring fingerJ0018H07Mm.28614Chromosome 1TCACAGATGTAprotein 149TGTGGAGGGGTTGTTTTCTGAGTACTAGACTACCCTCTGTGGTTATAAA52.K0508E12-3Rin3Ras and RabK0508E12Mm.24145Chromosome 12TCGGGGATGGinteractor 3AGCTGAGATGTTCCCACCACAACCCAAGATCTAAGAGTATTGTTTTGAAGA53.L0208A01-34933437L13RikRIKEN cDNAL0208A01Mm.159218Chromosome 16GGAGACTGAA4933437K13GCTTTTATTGTgeneTTAATGTTGAAGATATTGATCTACAAGGTGGGAATGGTG54.C0239G03-3BM202478ESTC0239G03Mm.217664Chromosome 2AACTGTGGGTABM202478TAATTGTAAGAGCCTGAAACTTCCAGAACTGGAGAAACTGTCACTGGGA55.L0518C11-31700016K05RikRIKEN cDNAL0518C11Mm.221743Chromosome 17GTGTTGTGATT1700016K05GTCGTCCCTGCgeneTTAATGAACCCACCTGAGGGACAGTTAGTGTCTTACCC56.H3054C09-3Oas1c2′-5′H3054C09Mm.206775Chromosome 5CTATATGAACToligoadenylateGAGAAACAACsynthetase 1CACGTATGCTGAACCCCAATTCTACAACAAAGTCTACGCC57.L0811E07-33110087O12RikRIKEN cDNAL0811E07Mm.32373Chromosome 3GGAATATATTA3110057O12TGTAGACTATTgeneCTGGCCTGAACCTTGTGGTTGACTGATGCTCTGCCTCC58.JO948A06-3Mus musculusJ0948A06Mm.261771Chromosome 14TTGGGTGATCCmRNA similarATATTTTTCAAto RIKENACCCATACTCCcDNACAAAAGGAGA4930503E14CCTACTTAAATgene (cDNATTCTCTcloneMGC:58418IMAGE:6708114,) completecds59.C0931B05-3transcribedC0931B05Mm.252843Chromosome 10GTTCCTGAAGCsequence withTCTTGATATTTweak similarityTAGGACAAAAto proteinCCCACCACGACref:NP_081764.1AAAATGAGAA(M. musculus)GGAATTTRIKEN cDNA5730493B19[Mus usculus]60.H3022A09-3Esp812EPS8-likeH3022A09Mm.27451Chromosome 7TGACTTCAAATGTCCCATCCCACCCAAAGAGCCTGTGATAACAGATGTCTCTGGCTATAT61.G0118B03-3Usf2upstreamG0118B03Mm.15781Chromosome 7TGGGTAGGTTCtranscriptionCTAGGTCTCCCfactor 2TGATATCTAACTACAGTTATACTGTAGCTGTGTGACA62.H3156C12-3Ms4a6dmembrane-H3156C12Mm.170657Chromosome 19CCTGTCTCAGAspanning 4-ACTCAAGAATdomains,AAATCCAGTGTsubfamily A,ATCTTCAGAGTmember 6DCACTTTGTAACCCTAC63.H3074G06-39530020G05RikRIKEN cDNAH3074G06Mm.15120Chromosome 6TACTCCCTGGA9530020G05GACTAGAACCgeneGTGGCTATAGCGGAGCATGCTCCAGAGCACAGGACTGAT64.NM_003254.1TIMP1tissue inhibitorNM_003254Hs.5831No ChromosomeGGGACACCAGoflocationAAGTCAACCAmetalloproteinaseinfo availableGACCACCTTAT1 (erythroidACCAGCGTTATpotentiatingGAGATCAAGAactivity,TGACCAAGcollagenaseinhibitor)65.K0647H07-3I17rinterleukin 7K0647H07Mm.389Chromosome 15GAAAACCAAAreceptorACTCTTGGTCAGAGACAATATGCAAAACAGAGATGTCAAGTACTATGTCC66.J0257F12-3Rnf25ring fingerJ0257F12Mm.86910Chromosome 1TCAAGGAGACTprotein 25GTAGACTTAAAGGCAGAACCCCGTAACAAAGGGCTCACAGGTCATCCTC67.H3083G02-3Lcn2lipocalin 2H3083G02Mm.9537Chromosome 2CACCACGGACTACAACCAGTTCGCCATGGTATTTTTCCGAAAGACTTCTGAAAACAAGCA68.M64086.1Serpina3nserine (orM64086Mm.22650Chromosome 12GTACCCTCTGAcysteine)CTGTATATTTCproteinaseAATCGGCCTTTinhibitor, cladeCCTGATAATGAA, member 3NTCTTTGACACAGAAAC69.C0906B05-3CenpccentromereC0906B05Mm.221600Chromosome 5AAGAACTACTGautoantigen CATACAGAACCACTTCAGTTGTTCAGTTAGAATCTTTTTAAGACTCTCTC70.H3094B08-3BG071051ESTsH3094B08Mm.173358Chromosome 2CTTGACCTTTABG071051GATGGAAATTGTACCTAGAGACGAGAAGGAGCCAAACTAAGGTCTGTCA71.K0110F02-3Pstpip1proline-serine-K0110F02Mm.2534Chromosome 9GGAACGGACAthreonineACGTGGCTTTGphosphatase-TCCCTGGGTCGinteractingTACTTGGAGAAprotein 1GCTCTGAGGAAAGGCTA72.L0072G08-3Renbprenin bindingL0072G08Mm.28280Chromosome XTTCGAATGCACproteinATCATTGACAAGTTTCTCTTATTGCCTTTCCACTCTGGATGGGACCCTG73.J0088G06-349304272G13RikRIKEN cDNAJ0088G06Mm.23172No ChromosomeGCCTGGAGACT4930475G13loctionGAAGGCAGTTTgeneinfo availableTACAAAGGAAAACTTAGATTTCTATTCATTTGCTTTTG74.K0121F05-3Fcgr2bFc receptor,K0121F05Mm.10809Chromosome 1CTGGATGAAGIgG, lowAAACAGAGCAaffinity IIbTGATTACCAGAACCACATTTAGTCTCCCTTGGCATTGGGA75.K0124E12-3Wbscr5Williams-K0124E12Mm.23955Chromosome 5TTAATATTGTCBeurenAATGTCAGGGsyndromeGGTTCCCTGTCchromosomeTCAGAGCATTAregion 5TGTGTACTAAChomologTGTAGC(human)76.K0649H05-3F730038I15RikRIKEN cDNAK0649H05Mm.268680No ChromosomeCCAGAGTTTTTF730038I15locationTCCATCATGTTgeneinfo availableTTGCCCCAAAGACCTCGGTTTGTAGAAGCCCAAGGAAA77.K0154C05-3D230024O04hypotheticalK0154C05Mm.90241Chromosome 6GACAGGGTCAproteinATGTTTATTATD230024O04ACATACTGCACTGATGAGAACAATATCATATGTGAAGAG78.C0185E05-3Hmox1hemeC0182E05Mm.230635Chromosome 8ACTCTCAGCTToxygenaseCCTGTTGGCAA(decycling) 1CAGTGGCAGTGGGAATTTATGCCATGTAAATGCAATAC79.L0823E04-3transcribedL0823E04Mm.270136Chromosome 7GACAGGGACTsequence withCCATATGGAAGweak similarityTAAGGACGTTTto proteinACCTCATTACTpir:T26134AAGTCTCGTCA(C. elegans)AAAGAAT26134hypothecticalproteinW04A4.5-Caenorhabditiselegans80.K0310E05-39830126M18hypotheticalK0130E05Mm.266485Chromosome 15CTCGGATCTTCproteinATGTTCTTCAG9830126M18TAAGAATCTCTCTGTGGATTTGGAACAATCGTAAATAA81.C0908B11-3P2ry6pyrimidinergicC0908B11Mm.3929Chromosome 7CTAAGACACCTreceptor P2Y,GTGATTTGGCAG-proteinACTGGTCAATTcoupled, 6CATGCTTGTTACATTCAGAACTCAGGA82.K0438A08-3Ccl2chemokine (C-K0438A08Mm.145Chromosome 11TCCCTCTCTGTC motif) ligandGAATCCAGATT2CAACACTTTCAATGTATGAGAGATGAATTTTGTAAAGA83.H3082C12-3Spp1secretedH3082C12Mm.288474Chromosome 5TTCTCAGTTCAphosphoproteinGTGGATATATG1TATGTAGAGAAAGAGAGGTAATATTTTGGGCTCTTAGC84.H3014A12-3Capgcapping proteinH3014A12Mm.18626Chromosome 6CTGACCAAGGT(actin filament),GGCTGACTCCAgelsolin-likeGCCCTTTTGCCTCTGAACTGCTAATTCCAGATGACTGC85.H3089C11-3BG070621ESTsH3089C11Mm.173282Chromosome 4GATACCTGGCTBG070621TATCTTTTATCAACAGCAAATTATGCAGTGGTGGAAATGTCATCACAGA86.X67783.1Vcam1vascular cellX67783Mm.76649Chromosome 3GTTTGAGAAGAadhesionGACATTATTTAmolecule 1TAAAACCCAGATCCTTAATACTGTTTATTACAGCCCCG87.J0509D03-3AU018874J0509D03Chromosome 13CTCTGATACTGMouse eight-AATAAACCTGAcell stageTGTGATGTACTembryo cDNATATAGTCCTTAMus musculusAGTCTTGAGAGcDNA cloneTTAGAJ0509D03 3′,MRNAsequence88.H3055A11-5UNKNOWN:H3055A11Data not foundChromosome 3GGCAACTACGSimilar toACTTTGTAGAGHomo sapiensGCCATGATTGTKIAA1363GAACAATCACproteinACTTCACTTGA(KIAA1363),TGTAGAAmRNA89.C0455A05-3AW413625expressedC0455A05Mm.1643Chromosome 19ACTTCATAGGAsequenceTTCACAATGGAAW413625GAGGGCTAGGAAGATACTGGACAATTTTCAGCAGTGTG90.NM_019732.1Runx3runt relatedNM_019732Mm.247493Chromosome 4CACCTCTTGTCtranscriptionTCCAGCCATGCfactor 3CCAGGATCAATTCTAGAATCAGAGGCTACCCCTGCCTG91.L0008A03-3AW546412ESTsL0008A03Mm.182599Chromosome 16CGTCAGTGACCAW546412CACTCAATACTGTGGTGGGAAGTAAGATGATGCCAAATCTATAACCTGT92.K0329C10-3Thbs1thrombospondinK0329C10Mm.4159Chromosome 12CGAATGAGAA1TGCATCTTCCAAGACCATGAAGAGTTCCTTGGGTTTGCTTTTGGGAAAGC93.H3115H03-3BC019206cDNA sequenceH3115H03Mm.259061Chromosome 10CCGGCGGGCCCBC019206TAGTTTCTATGTATTTAGAATGAACTCGTGTACATATGTAAAGATCTTT94.C0643F09-3Usp18ubiquitinC0643F09Mm.27498Chromosome 6CAAGCTGGTTGspecificGAGCCTCCAGCprotease 18CTTCAAAATTCTGAATCTAATAAACATTAATGCACACT95.X84046.1HgfhepatocyteH84046Mm.267078Chromosome 5CAATCCTAGAAgrowth factorCAACTACTTGAGTGTTGTGAGTGTTCAGATACTCATTAATATATATGGG96.L0236C05-3Aldh1b1aldehydeL0236C05Mm.24457Chromosome 4TCCCACCTCTCdehydrogenaseTGATGAGTTAT1 family,AGCCAAGAAGmember B1CCTTAGGAGTCTCCATAAGGCATATTCA97H3055E08-3Mcoln2mucolipin 2H3055E08Mm.116862Chromosome 3AAGAAATATTCCCACTTCAGAGTGTGTAAGCAATATTTAAACCCAGATAAAGATGCATGC98.H3009F12-3BG06369ESTsH3009F12Mm.196869Chromosome 5TTTGGGAGTGGBG063639GCTTCATGAATGCGCTCTTACCAAAGGAGCCATGTTTCCATTGTATCAA99.J0208G12-3Cxc11chemokine (C-J0208G12Mm.21013No ChromosomeTTTCATTAAACX-C motif)locationTAATATTTATTligand 1info availableGGGAGACCACTAAGTGTCAACCACTGTGCTAGTAGAAG100.K0300C11-39130025P16RikRIKEN cDNAK0300C11Mm.153315Chromosome 1AAGTGACTCCA9130025P16TTTTCATATGTgeneACTTAAACACAGAGTTCCTGTGGCCTCTGTAAGCTCAG101.H3104F03-5Krt1-18keratin complexH3104F03Mm.22479Chromosome 15CAAGGTGAAG1, acidic, geneAGCCTGGAAA18CTGAGAACAGGAGACTGGAGAGCAAAATCCGGGAACATCT102.L0858D08-3Trim2tripartite motifL0858D08Mm.44876Chromosome 3GCATGTGATTGproteinATTCATGATTTCCCCTTAGAGAGCAAGTGTTACCAAAGTTCTGTTGAGC103.L0508H09-3BY564994EST BY564994L0508H09Mm.290934Chromosome 12TGCTCCAGATGTGAAACTTATAGACGTAGACTACCCTGAAGTGAATTTCTATACAGGAAG104.L0701G07-3BM194833ESTsL0701G07Mm.221788Chromosome 2TGTACAACTGABM194833ACTCACCTCTTGTGAAGAATTATGATTGTCTTACTTGTAAAGAAAGCAC105.K0102A10-3E430015L02RikRIKEN cDNAK0102A10Mm.33498Chromosome 16TTTTGCAGGGGE430025L02TCGAGTGTGATgeneGCATTGAAGGTTAAAACTGAAATTTGAAAGAGTTCCAT106.C0190H11-3SpnsialophorinC0190H11Mm.87180Chromosome 7CAAACAGAAAACAGGGAGATGTAAAACAGTTTCAACTCCATCAGTTATGAAACCATAGCT107.L0514A11-32810457I06RikRIKEN cDNAL0514A11Mm.133615Chromosome 9TCAGCAAATTG2810457I06GCGATTTCGGAgeneATCCTATGACACCTACATCAATAGGAGTTTCCAGGTGA108.J0911E11-3Neflneurofilament,J0911E11Mm.1956Chromosome 14CATGTGCAACClightTCATGGGAAApolypeptideAATAGTAACTTGAATCTTCAGTGGTTAGAAATTAAAGAC109.K0647E02-3Def6differentiallyK0647E02Mm.60230Chromosome 17GTCTCAAGGATexpressed inCTGGGACCAGFDCP 6AACTGGGAAAGAAAAGGAATGACCAAGACAAGATCATAC110.H3091E09-3EiflaeukaryoticH3091E09Mm.143141Chromosome UnTGAATCAGAGtranslationAAAAGAGAGTinitiation factorTGGTGTTTAAA1AGAATATGGGCAAGAGTATGCTCAGGTGAC111.AF286725.1Pdgfcplatelet-derivedAF286725Mm.40268Chromosome 3AAAGGAAATCgrowth factor,ATATCAGGATAC polypeptideAGATTTGTATCTGATGAGTATTTTCCATCTGAACCCGGA112.D31942.1Osmoncostatin MD3194218413Chromosome 11CAGTCCTCTTGAAAGGTCTCAGAAGCTGGTGAGCAATTACTTGGAGGGACATGACTAATT113.L0046b04-3AlcamactivatedL0046B04Mm.2877Chromosome 16AGAGGAGTCTCleukocyte cedlCTTATATTAATadhesionGGCAGGCATTAmoleculeTAGTAAAATTATCATTTCCCCTGAGGA114.K0131D09-3LOC217304similar toK0131D09Mm.297591Chromosome 11GCATGAGTGTAtriggeringTAGGTGAAGGTreceptorTTCACTTTAAGexpressed onATGCTGTCTTCmyeloid cells 5AGTTCTCTTGC(LOC217304),CTATGmRNA115.H3024C07-3HexahexosaminidaseH3024C07Mm.2284Chromosome 9ATCGTCTCTGAATTATGACAAGGGCTATGTGGTGTGGCAGGAGGTATTTGATAATAAAGTG116.L0251A07-3B4galt1UDP-L0251A07Mm.15622Chromosome 4CTGTTCGTGTTGal:betaGlcNAGGGTTTTGTTCc beta 1,4-ATGTCAGATACgalactosyl-GTGGTTCATTCtransferase,TCAGGACCAApolypeptide 1GGGAAA117.C0612G04-3Grip 1glutamateC0612G04Mm.196692Chromosome 10GTGCAATAGAreceptorAATATATGATTinteractingTCAAACACATTprotein 1TCTGAACTGCCAGGGCAAGAAAGTATAG118.C0357B04-3C0357B04-3C0357B04No ChromosomeCTTGTCGTTTTNIA MouseloctionTGGGGGTTGTAUndifferentiatedinfo availableATATCTAAGGGES CellTGAAAAAATTAcDNA LibraryATTTCCAAAGC(Short) MusCAAGAmusculuscDNA cloneC0357B04 3′,MRNAsequence119.L0529E02-3Egfl3EGF-like-L0529E02Mm.29268Chromosome 4CAACTGTTTACdomain,CTGGAAATGTAmultiple 3GTCCAGACCATATTTATATAAGGTATTTATGGGCATCT120.L0218E05-3Dnase2adeoxyribonucleaseL0218E05Mm.220988Chromosome 8CCTTCCAGAGCII alphaTTTGCCAAATTTGGAAAATTTGGAGATGACCTGTACTCCGGATGGTTGG121.H3074C12-3DutpdeoxyuridineH3074C12Mm.173383Chromosome 2TAGGTGAGTTAtriphosphataseGGAATCTGCCATAAGGTCGTTTATAGGATCTGTTTATATGAAGTAATGG122.H3072F09-3Icsbp1interferonH3072F09Mm.249937Chromosome 8ATGACTTTCTCconsensusTGCTTGGTTGGsequenceAGAAGAAGAAbinding proteinTCTTTACTATT1CAGCTTCTTTTCTTTTT123.c0829f05-34632404H22RikRIKEN cDNAC0829F05Mm.28559Chromosome XCCGGGGTGGG4632404H22AAGTTGTTTTTgeneTCCTGGGGGTTTTTTCCCCTTATTTGTTTTGGGGCCCCT124.L0063A12-3similar toL0063A12Mm.38094Chromosome XGGAAGATGGGubiquitin-TAAATAGTAGAconjugatingCTGTGGTGTATenzyme UBCiTTGGAACAAG(LOC245350),GTAGCTTTAAAmRNAGACACAA125.C0143E09-36330548O06RikRIKEN cDNAC0143E09Mm.41694Chromosome 5CCAGGTTCAGA6330548O06GCGGACTGCTAgeneATAATAATGTGTGTATTGATCGAGGAAAAAGTGCGGAG126.K0127G03-3transcribedK0127G03Mm.32947Chromosome 14TGCATGGGAAsequence withATTTCTACGTGweak similarityGCTCACTTCACto proteinCAAGGCTTATTref:NP_000072.1GCACTGGGAA(H. spaiens)AAGAAGAbeige proteinhomolong;Lysosomaltraffickingregulator[Homo sapiens]127.H3109D03-3Lamp2lysosomalH3109D03Mm.486Chromosome XTTAACCTAAAGmembraneGTGCAACCTTTglycoprotein 2TAATGTGACAAAAGGACAGTATTCTACAGCTCAAGACT128.J0034B02-3Dhx16DEAH (Asp-)J0034B02Mm.5624Chromosome 17TCCCCACTACTGlu-Ala-His)ATAAGGCCAAbox polypeptideGGAGCTAGAA16GATCCCCATGCTAAGAAAATGCCCAAAAA129.K0428C07-3Plcb3phospholipaseK0428C07Mm.6888Chromosome 19ATAGGTACTCCC, beta 3CCGATTCCCAAGGAGCAGCTAGTGGAACCCTGGAGTTTTGGGTAGTAGA130.K0119F10-3Ccl9chemokine (C-K0119F10Mm.2271No ChromosomeAGTAGTATTTCC motif) ligandlocationCAGTATTCTTT9info availableATAAATTCCCCTTGACATGACCATCTTGAGCTACAGCC131.J0046B07-3Tuba4tubulin, alpha 4J0046B07Mm.1155Chromosome 1ACCGCTACTTGGAGCCTGTTCACTGTGTTTATTGCAAAATCCTTTCGAAATAAACAGTCT132.C0117E11-3Neu1neuraminidaseC0117E11Mm.8856Chromosome 17TGAACTCTGAC1CTTTTGCAACTTCTCATCAACAGGGAAGTCTCTTGGTTATGACTTAACA133.C0101C01-3Sdc1sydecan 1C0101C01Mm.2580No ChromosomeGTCTGTTCTTGlocationGGAATGGTTTAinfo availableAGTAATTGGGACTCTAGCTCATCTTGACCTAGGGTCAC134.K0245A03-39130012B15RikRIKEN cDNAK0245A03Mm.35104No ChromosomeCCAGCCTGACC9130012B15locationAGATTTTAGTTgeneinfo availableACCTTTTAAGGAAGAGAGATTTATTCTAATGCCATAAA135.H3109A02-3FcerlgFc receptor,H3109A02Mm.22673Chromosome 1CACCTCTGTGClgE, highTTTGAAGGTTGaffinity I,GCTGACCTTATgammaTCCCATAATGApolypeptideTGCTAGGTAGGCTTTA136.L0819C05-3Mapk8ipmitogenL0819C05Mm.2720Chromosome 2CTGAGCTCAGGactivatedCTGAGCCCACGprotein kinase 8CACCTCCAAAGinteractingGACTTTCCAGTproteinAAGGAAATGGCAACGT137.U77083.1AnpepalanylU77083Mm.4487Chromosome 7AGAACAGCAG(membrane)TTAGTTCCTGGaminopeptidaseTTCTGAGAACCACTTGTCCCAGTATGACACCTCTTACTA138.C0164B01-3Tnfaip2tumor necrosisC0164B01Mm.4348Chromosome 12ATGTGTGTACTfactor, alpha-CAGGACAGAAinduced proteinTCCAGAGATTT2CTTTTTTATATAGCTTGATATAAAACAG139.H3085G03-3Cybacytochrome b-H3085G03Mm.448Chromosome 8ACGTTTCACAC245, alphaAGTGGTATTTCpolypeptideGGCGCCTACTCTATCGCTGCAGGTGTGCTCATCTGTCT140.H3074F04-3Abcc3ATP-bindingH3074F04Mm.23942Chromosome 11TTTTTTAATTCTcassette, sub-GCAAATTGTCTfamily CCACAGTGGAAT(CFTR/MRP),GAGGAAATGAmember 3GTTAGAGATCACAGCC141.H3145E02-3Wbp1WW domainH3145Eo2Mm.1109Chromosome 6GTGCTATCTTTbinding proteinACTCACTCCCA1AGACATACACAGGAGCCTTTAATCTCATTAAAGAGACA142.K0609F07-3Cd53CD53 antigenK0609F07Mm.2692Chromosome 3GAGGTCCAAGTTTAAATGTTAGTCTCCTAACAACTGTCAAATCAATTTCTAGCCTCTAAA143.K0205H04-39830148O20RikRIKEN cDNAK0205H04Mm.21630Chromosome 9CTTCTAGATCC9830148O20TTCTGCAGAAAgeneTCATCGTCCTAAAGGAGCCTCCAACTATTCGACCGAAT144.H3095H04-32410002I16RikRIKEN cDNAH3095H04Mm.17537Chromosome 18ACTTATTCATC2410002I16CTTGCCTATACgeneCCACCCCCCAAAAACAGGTTTTATTAATAAAAAATGTG145.C0623H08-3Tm7sfltransmembraneC0623H08Mm.1585Chromosome 13TACAGTAACAA7 superfamilyGCAAGCTATCAmember 1TCCATTTTTACAATAAAGTTGTCAGCATTCATGTCAGC146.L0242F05-32700088M22RikRIKEN cDNAL0242F05Mm.103104Chromosome 15TTATTTACTTT2700088M22ATCTTAGTATGgeneTAACCTTAGCTGACCTGAAACCCACTGGTAGACTAGAC147.C0177F02-3Sdc3sydecan 3C0177F02Mm.206536Chromosome 4CCTGTCCTGAGTTCATGGCCAAAACTTAAATAAGAGAAGGAGGAGAGGGTCAGATGGATA148.L0803B02-3Ppp1r9aproteinL0803B02Mm.156600Chromosome 6AAAGGGGCCTphosphatase 1,GAGTATACGCTregulatoryGTTGCAAGCTG(inhibitor)TATACTTCATTsubunit 9ATCCTTCGGCTGGTTTAT149.H3061D01-3BB172728ESTsH3061D01Mm.254385Chromosome 3TATCCGGACAGBB172728TCTATGTGAAATAGGACCAAGGTCGAAAGCCGGAAAGACATCAACAGAA150.L0259D11-3ClqbcomplementL0259D11Mm.2570Chromosome 4CTGCTTTTCCCcomponent 1, qTGACATGGATGsubcomponent,CGTAATCACGGbetaGGTCAAATTACpolypeptideACCTATCCAACACCAT151.H3011D10-3LcpllymphocyteH3011D10Mm.153911Chromosome 14AACAAAGAGGcytosolicACAGTATGAATprotein 1TTGAATAGCTCCCACTAGATAAGCAATTTCCACGAGAAC152.H3052B11-3Pctk3PCTAIRE-H3052B11Mm.28130Chromosome 1CTGACTGTGAAmotif proteinTGTCGTGACTCkinase 3AGAGCAAAGACAGAGAATATATTTAATTCATGTTGTAC153.k0413h04-3Anxa8annexin A8K0413H04Mm.3267Chromosome 14GCCTGAAGAACATGACAGAACTCTTCTCAATATTCGTTGGGCTTTCAGAATCATAAACAT154.H3054F05-3LyzslysozymeH3054F05Mm.45436Chromosome 10CCTGTGTGAATAAAAATACAAGAACTGCTTATAGGAGACCAGTTGATCTTGGGAAACAGC155.H3060F11-3Cybbcytochrome b-H3060F11Mm.200362Chromosome XGTAAGAAATAT245, betaTAGACTGATTGpolypeptideGAGTTAAAGTAGCACTCTACATTTACCATGGTGTTTGG156.H3012F08-39430068N19RikRIKEN cDNAH3012F08Mm.143819Chromosome 1TGTGAAAGATT9430068N19GTGCATCTGCAgeneTTCAACTACCCTGAACCCTTAGGGAAGAAATGGATTCC157.G0106B08-3Abractive BCR-G0106B08Mm.27923Chromosome 11AGCTGCCTACTrelated geneAGCAGTTTAACAAGGAGCCTTGCTGTCTCAGACAGGTGAAAGAAAATGT158.L0287A12-3Tdrkhtudor and KHL0287A12Mm.40894Chromosome 3CCATGTTTGAAdomainAGTATGTAATGcontainingAAGAGGAGCCproteinTATTAACCATATGAAAGACAGGAATACT159.H3083D01-3AY007814hypotheticalH3083D01Mm.160389Chromosome 7GTGAATTGGATprotein,GCATAGCATGT12H19.01.T7TTTGTATGTAAATGTTCCTTAAAAGTGTCACCATGAAC160.H313F02-3BGO74151ESTsH3131F02Mm.142524Chromosome 8ACCCACTGACTBG074151AGGATAACTGGAAAGGAGTCTGACCTGAATGACGCATTAAACTCCTGCA161.C0172H02-3Lgals3lectin, galactoseC0172H02Mm.2970Chromsome 14CCCGCTTCAATbinding, solubleGAGAACAACA3GGAGAGTCATTGTGTGTAACACGAAGCAGGACAATAACT162.K0542E07-3Cd44CD44 antigenK0542E07Mm.24138Chromosome 2ATATTAACTCTATAAAAATAAGGCTGTCTCTAAAATGGAACTTCCTTTCTAAGGGTCCCAC163.C0450H11-3E430019N21RikRIKEN cDNAC0450H11Mm.275894Chromosome 14TGTGGGTTTTTE430019N21TGAAGAATTAAgeneTGAGCATGTACATAGAAATAGTGACTGCTTGAATCCTG164.K0216A08-3Orc51originK0216A08Mm.566Chromosome 5CTACTCTTAATrecognitionAGATGTTAT-complex,CTTsubunit 5-likeAACACTGAAAT(S. cerevisiaae)TGCCTGAAACCCATTTACTTAGGACTG165.H3122D03-3Pdgfcplatelet-derivedH3122D03Mm.40268Chromosome 3TCAGACCA-growth factor,TTTCC polypeptideTAGGCACAGTGTTCTGGGCTATGGCGCTGTATGGACATATCCTATTTAT166.C0037H07-3Il13ralinterleukin 13C0037H07Mm.24208Chromosome XTCTGAATCTGGreceptor, alphaGCACTGAAGG1GATGCATAAAATAATGTTAATGTTTTCAGTAATGTCTTC167.H30554F04-32610318I15RikRIKEN cDNAH3054F04Mm.34490Chromosome 11GATCCTTAGGT2610318I15CTCCATAGGATgeneGATTTTTGAGGTAGTTAATCAGTGTAAACTCTTACACA168.L0908A12-3BlnkB-cell linkerL0908A12Mm.9749Chromosome 19CTCAGCAGTAACAGAGAAAAGATGAATGAAGCCACTGAGGCTTCGTGAATGAATGAATCT169.G0111E06-3Car7carbonicG0111E06Mm.154804Chromosome 8CTTTGTTCCTAanhydrase 7CCCAGCCACCAAAGCCACCTACATAACAATCCACTCATGTACTAGCAAA170.L0284B06-3Ngfrap1nerve growthL0284b06Mm.90787Chromosome XAAATTGTCTACfactor receptorGCATCCTTATG(TNFRSF16)GGGGAGCTGTCassociatedTAACCACCACGprotein 1ATCACCATGATGAATT171.K0145G06-3TcfectranscriptionK0145G06Mm.36217Chromosome 6ACATGATGTGAfactor ECAAGAATCATTGAAGATCACAGTTGTCTACCGAGTTCAGATTTCCTTACA172.H3001B08-3LynYamaguchiH3001B08Mm.1834Chromosome 4CACCCCCCAGAsarcoma viralAAATGAGACT(v-yes-1)ATTGAACATTToncogeneTCCTTTGTGGThomologAAGATCACTGGACAGGA173.G0117F12-3Prkcshprotein kinaseG0117F12Mm.214593Chromosome 9AGTGATGGGGC substrateACCATGACGA80K-HGCTGTAGCCTGAACCTCAAGGCCTGAACCAGTCTACTGA174.C0903A11-32510004l01RikRIKEN cDNAC0903A11Mm.24045Chromosome 12AAAGGTCCCA2510004L01GGTTTCGATCTgeneGTTTGGAGTTTGGAGTCTAATGGTTGCATAGATAAACAG175.L0062C10-3Rasa3RAS p21L0062C10Mm.18517Chromosome 8TCTATGTGCATproteinTAGGGGGTGAactivator 3CCCAGGGAAATCCAAAGGGAACAGTATTTGATTTCTCAC176.L0939G09-3Cd38CD38 antigenL0939G09Mm.249873Chromosome 5CTACACATGTACTTTAGGATTCTAGGTTTCTCCCTGAGCCCTGCTTTCGATGTAACACTG177.H3115B07-3S100a9S100calciumH3115B07Mm.2128Chromosome 3AAGTCTAAAGbinding proteinGGAATGGCTTAA9 (calgranulinCTCAATGGCCTB)TTGTTCTGGGAAATGATAAGATAAATAA178.K0608H07-3FybFYN bindingK0608H07Mm.254240Chromosome 15GGAAGAAAAAproteinGACCTCAGGAAAAAATTTAAGTACGACGGTGAAATTCGAGTTCTATATTC179.C0104E07-3Tcirg1T-cell, immuneC0104E07Mm.19185Chromosome 19GGATGAAGAAregulator 1ACTGAGTTTGTCCCTTCTGAGATCTTCATGCACCAAGCAATCCACACCAT180.K0431D02-3Wisp1WNT1K0431D02Mm.10222Chromosome 15CTGTTCAGGCTinducibleCAAACAATGGsignalingGTTCCTCCTTGpathway proteinGGGACATTCTA1CATCATTCCAAGGAAAA181.L0837H10-3Igfbp2insulin-likeL0837H10Mm.141936Chromosome 1AGGAGTTCCCAgrowth factorGTTTTGACACAbinding proteinTGTATTTATAT2TTGGAAAGAGACCAACACTGAGCTCAG182.C0159A08-3Mta3metastasisC0159A08Mm.18821Chromosome 17CTCAATAAAAGassociated 3CTCTAAGGAGACATCACAACCCAGTCTTAAGGGTTCATGAGGTTTTAAT183.K0649D06-3Ms4a6bmembrane-K0649D06Mm.29487Chromosome 19ACTTAAAATGTspanning 4-AGACTGTTCATdomains,ACAGTGGGTACsubfamily A,CAGTATGAGTTmember 6BGAATGTGTGTATTACT184.K0609D11-3Manlamannosidase 1,K0609D11Mm.117294Chromosome 10TTTCATAATAGalphaAACCGTCTACCAGTGACCTCTTGATTATGATTTGATTTGACTGCAAAAC185.C0907B04-3Mcoln3mucolipin 3C0907B04Mm.114683Chromosome 3ATCCATGTGGCATCAATTCAATTATGTATAATAATGACTTTACAAGGGCCCCTTAAAACC186.H3020D08-3Edem 1ER degradationH3020D08Mm.21596Chromosome 6CACAAAAGTCenhancer,AAATGTGGATAmannosidaseTCGTACGCTGCalpha-like 1ATCACGTCATAGACAAGTCTAAAGAAGA187.J0039F05-3Gdf3growthJ0039F05Mm.4213Chromosome 6CTATCAGGATAdifferentiationGTGATAAGAAfactor 3CGTCATTCTCCGACATTATGAAGACATGGTAGTCGATGA188.C0906C11-3BM218094ESTsC0906C11Mm.212279Chromosome 6GGAGATCATCABM218094CTCTTGTATGAAATATACTAACTCCAAACCTTTTTAGAGCAGATTAGGC189.L0266E10-3B930060C03hypotheticalL0266E10Mm.89568Chromosome 12ACTATTAAGCAproteinCTCAGGAGAATB930060C03GTAGGAAAGATTTCCTTTGCTACAGTTTTTGTTCAGTA190.H3060D11-3M115myeloid/lymphH3060D11Mm.10878Chromosome 5AAAGAGAAAAoid or mixed-TATGTCAGATGlineageGTGATACCAGTleukemia 5GCAACTGAAAGTGGTGATGAAGTTCCTG191.L0062E01-3Tnctenascin CL0062E01Mm.980Chromosome 4GAGAGAGGAATGGGGCCCAGAGAAAAGAAAGGATTTTTACCAAAGCATCAACACAACCAG192.K0132G08-3A1662270expressedK0132G08Mm.37773No ChromosomeGTTGTACTACTsequencelocationGGAAAGATTTTA1662270info availableGCTGGGACATACAATATGTGTGAGAAAAATAGAGTTGT193.H3114D08-3Arpc3actin relatedH3114D08Mm.24498Chromosome 5AGACCAAAGAprotein 2/3CACGGACATTGcomplex,TGGATGAAGCCsubunit 3ATCTACTACTTCAAGGCCAATGTCTTCT194.C0649E02-3Unc93bunc-93C0649E02Mm.28406Chromosome 19CAGAGCAGGGhomolog B (C.GGCTTTTATTTelegans)TTATTTTTTAATGGAAAATAATCAATAAAGACTTTTGTA195.L0293H10-32510048K03RikRIKEN cDNAL0293H10Mm.39856Chromosome 7CTTGGCAGCTC2510048K03TCCTTACTTCTgeneGGGACATTTGCCACTGTGGTACTGCCAGGAAGGAATCT196.H3024C03-31110008B24RikRIKEN cDNAH3024C03Mm.275813Chromosome 12ACTTATAGAAA1110008B24AGGACAGGTTgeneGAAGCCTAAGAAGAAAGAGAAGAAAGATCCGAGCGCGCT197.H3055002-3Ctsccathepsin CH3055G02Mm.684Chromosome 7TAGTTCAGTGAACAAGTATCTGTCAATGAGTGAGCTGTGTCAAAATCAAGTTATATGTTC198.K0518A04-3BM238476ESTsK0518A04Mm.217227Chromosome 2CATGAATGTCABM238476AAACCTAATTACAAAGCATCGGTCTCTTTGTTGTGAGGTATCAGAACCC199.K0128H01-3Parvgparvin, gammaK0128H01Mm.202348Chromosome 15CCTGTCTCATGGGAGATTTGAATCATAAGGAGAATCACTTTTTGTAACTTTATTGAGGAA200.K0649F04-3Ccr2chemokine (C-K0649F04Mm.6272Chromosome 9AAGTAAATATGC) receptor 2CAAAGGAGAGAAGTTAGAGAAACTCCTCTCATAAGAAAAATGTCTTCCC201.K0603E03-3Vav1vav 1 oncogeneK0603E03Mm.254859Chromosome 17TCGGAACTGTCCCTTAAGGAGGGTGATATCATCAAGATCCTCAATAAGAAGGGACAGCAA202.K0649A02-3Stat1signalK0649A02Mm.8249Chromosome 1TTAGTGGGCTGtransducer andAACCTATCGGTactivator ofTTTAACTGGTTtranscription 1GTCTTAATTAACCATAAACTTGGAGAA203.H3013D11-3Mt2metallothioneinH3013D11Mm.147226Chromosome 8TTTTGTACAAC2CCTGACTCGTTCTCCACAACTTTTTCTATAAAGCATGTAACTGACAATA204.H3013B02-3Atp6vlb2ATPase, H +H3013B02Mm.10727Chromosome 8AGACTTGGAAtransporting,AAGGCTTGGGTV1 subunit B,ACAATTAAGAisoform 2AAAACCCTACATCCCACCCTCCTCTTGAC205.L0541H09-3transcribedL0541H09Mm.221768Chromosome 6TAATAAAGAAsequence withACTGTGGAAATweak similarityACTTGGATTTCto proteinTACTGAAGACApir:S12207AAAGACTTCTA(M. musculus)GGCTGGS12207hypotheticalprotein (B2element) -mouse206.K0516E03-3Mus musculusK0516E03Mm.214742Chromosome 10AGGTTAAACAT12 days embryoATATTCTTGGAembryonicAACATGAAATCbody betweenACAACTCTCAAdiaphragmAAACCGTGAAregion and neckCCACCAcDNA, RIKENfull-lengthenrichedlibrary,clone:9430012B12product:unknownEST, fullinsert sequence.207.H3034A10-3PlaururokinaseH3034A10Mm.1359Chromosome 7CCTCGTGTTGTplasminogenCTTCTTTGGACactivatorCTCAGTTTTTCreceptorCATGAACCAGAAGAGAATTGGAACAAG208.C0910G05-3BM218419ESTsC0910G05Mm.217839Chromosome 10AATAGCAATGTBM218419ATCAAACAATGGATGTGAAAAAGATGCGCTCTATCATCATGAAAATGCC209.C0262H12-3Msh2mutS homologC0262H12Mm.4619Chromosome 17TCTCTGGAGAA2 (E. coli)ATCAGTAACTGCAAAAGGAAGAGAGGGTCTTTAAAGCACATGTAGTAAT210.H3078C11-3BG069620ESTsH3078C11Mm.173427Chromosome 2TGGAATGTTGABG069620AGAATGAAATCTCGAGGGAATTAGAGGTTGAGGTCATCTGGATATTCAG211.L0926H09-36030440G05RikRIKEN cDNAL0926H09Mm.27789Chromosome 6ATAGAACCAAT6030440G05GTAGGAAAATgeneCAGGCAAAATAAAATGATGATCAGTCCATGTCATCATGG212.J0076H03-3C80125 MouseJ0076H03No ChromosomeAGATGGGAAA3.5-dpclocationAAGTACTGTAGblastocystinfo availableGTTCCTGAACTcDNA MusCTGGATCTCAAmusculusGCAGAAATGTcDNA cloneACTGTCTJ0076H03 3′,MRNAsequence213.L0817B08-3transcribedL0817B08Mm.221816Chromosome 18 notAGGAAAACCCsequence withplacedCGGTAGTTAGGstrongACATCTGAATTsimilarity toCTCAATTATTGproteinGATTGCCAAAAsp:P00722 (E.GTGAAAcoli)BGAL_ECOLIBetagalactosidase(Lactase)214.H3065D11-3Crnkl1Cm, crookedH3065D11Mm.273506Chromosome 2GTTTTTGGAATneck-like 1TTGGACCTGAA(Drosophila)AATTGTACCTCATGGATTAAGTTTGCAGAATTAGAGAC215.H3157E02-35630401J11RikRIKEN cDNAH3157E02Mm.21104Chromosome 17TGGGACCTGTG5630401J11AAGCGACTGAgeneAGAAAATGTTTGAAACAACAAGATTGCTTGCAACAATTA216.H3007C11-3BG063444ESTsH3007C11Mm.182542No ChromosomeTCCATTATTACBG063444locationATACAACAATCinfo availableAAGAAAAAGACAGAAAACTACCCTTAGAGAGATCAGGG217.K0517E07-3C53005OH1ORikRIKEN cDNAK0517E07Mm.260378Chromosome 4ATTCAACAGCAC530050H10TTCTAGGAAAAgeneTGGCAAGAAAGTAAATTATCATCCATTTCAGGTCTGTG218.H3150B11-5Ptpn2protein tyrosineH3150B11Mm.260433Chromosome 18CCATATGATCAphosphatase,CAGTCGTGTTAnon-receptorAACTGCAAAGTtype 2ACTGAAAATGATTATATTAATGCCAGC219.C0199C01-39930104E21RikRIKEN cDNAC0199C01Mm.29216Chromosome 18GGGCCATATTT9930104E21TAAAGATAAGgeneGAGAGAGAAACTAGCATACAGAATTTTCCTCATATTGAG220.H3063A09-3Rassf5Ras associationH3063A09Mm.248291Chromosome 1GAAAGGCGTTT(RaLGDS/AF-6)ATTCAGAAAATdomain familyGATGGTAAGAT5TCAGACTTTAAAGCACAGTTAGACCCA221.K0445A07-3HfehemochromatosisK0445A07Mm.2681Chromosome 13TAAGGTGTTTTCTCCAGTTAAGTTCAGTTCCTGAATAGTAGTGATTGCCCCAGTTGCAAC222.H3123G07-3C630007C17RikRIKEN cDNAH3123G07Mm.119383Chromosome 2CCACCATAAAGC630007C17GAAAAAGGACgeneATGTGTATGAGTAGGTGTTCATCTATGTGCATAATTGGC223.H3094C03-3BazlabromodomainH3094C03Mm.263733Chromosome 12GCACAAGATGadjacent to zincGAGTCATTAAAfinger domainATTAAGGCATC1AATCATTTTCAGCATATAACATAGCAGAG224.L0845H04-3BM117070ESTsL0845H04Mm.221860Chromosome 1GATTAAAAACBM117070ATTAGGGATGAGAAATAATAAGGGCTTGCAACTGTGTAGAAGCTAGAGCC225.C0161F01-3BC010311cDNA sequenceC0161F01Mm.46455Chromosome 4TGAAGTACACTBC010311CTCTAAATGAAAATGGGCTATAAATATGTTTGAGTAGGATAGGAGGAAG226.H3034E07-3BG065726ESTsH3034E07Mm.5522Chromosome 9GTGTAAGAAABG065726AGATGGGACTGACAATAAAAATGAAGGTCAGGTAAGAAGTACCAGACTCC227.J0419G11-3Cldn8claudin 8J0419G11Mm.25836Chromosome 16GGGAAATATGCAGCGTTCTATGTTTCCATAAGTGATTTTAGCAGAATGAGGTATTATGTG228.C0040C08-3Cxcr4chemokine (C-C0040C08Mm.1401Chromosome 1GTAGGACTGTAX-C motif)GAACTGTAGAreceptor 4GGAAGAAACTGAACATTCCAGAATGTGTGGTAAATTGAA229.K0612H02-3BM241159ESTsK0612H02Mm.222325Chromosome 16TCATAGGTCTCBM241159CATTTAGTTCAAGTGTTTTATGGACAATCAGCAAGTTTAGGCTCATAGG230.J0460B09-3AU024759J0460B09No ChromosomeTTGGAATATATMouselocationGAATGACAAAunfertilized egginfo availableGAAATGGGAAcDNA MusAAACTGCTGAAmusculusCCCGAGTCTCTcDNA cloneGAATGTCJ0460B09 3′,MRNAsequence231.H3103F07-3Mus musculusH3103F07Mm.174026Chromosome 10CTATCTTGAATtranscribedTGCTAGATTAAsequence withAGAGAAAGAAweak similarityAATGTTAGAGCto proteinAAAATAGGAAref:NP_081764.1CCTGGCC(M. musculus)RIKEN cDNA5730493B19[Mus musculus]232.H3079H09-3BG069769ESTsH3079H09Mm.173446Chromosome 9AATCCCTAGAGBG069769AAAATGGGAATAGAAATAAGCTGCATACAAACTCAAAGACACAGATACT233.H3130D06-3BG074061ESTsH3130D06Mm.182873Chromosome 1AGACTGAAGABG074061AAACCTTAAAATACCCAAAATTCAGGGGAGACATAGCAACTGAGTCTCAT234.H3071D08-3Lcp2lymphocyteH3071D08Mm.1781Chromosome 11AGAGGACTTCCcytosolicTGTCTGTATCAprotein 2GATATTATTGACTACTTCAGGAAAATGACGCTGTTGCT235.K0218E07-3Mus musculusK0218E07Mm.216167Chromosome 10ATGGAGATGTG10 days neonateTAAACAGTAGolfactory brainGACATTTCGATcDNA, RIKENAACTATGTCAGfull-lengthGTCAGTTCTTAenrichedGTTCAGlibrary,clone:E530016P10product:weaklysimilar toONCOGENETLM [Musmusculus], fullinsert sequence.236.C0907H07-3BM218221ESTsC0907H07Mm.221604Chromosome 12GAGGCTATTATBM218221AAATAACCTGAAATGCATATGAGAACTGAACGTGTAATAATTCAGCTCC237.K0605B09-3BM240642ESTsK0605B09Mm.222320Chromosome XAAGTCGGAATBM240642ATGTCTTAGTGTTCTTCTCACTTAGCTCAGTGTAAGATGGTAGCTCAAGT238.C0322F05-3Eya3eyes absent 3C0322F05Mm.1430Chromosome 4CACTTTTCTAThomologGAAGAAAGCC(Drosophila)GTGTGTAAAGTTTCCGTGACAGTAGTAATGGAAATATCT239.J0004A01-3C76123ESTs C76123J0004A01Mm.24905Chromosome 15TGTAAGAATACAAGGTAAAACAAAATAGAGAAATACAGGCATCATATCTGCAAATCGCCG240.K0139H06-3BM223668ESTsK0139H06Mm.221718Chromosome 3CAGAAACAGTBM223668AGTATGGGGTTAAATCACAATGAGGGAAATTATAGGGATATGCAGCCAAG241.L0941F06-3BM120591ESTsL0941F06Mm.217090Chromosome 9ACTGAAAGTTGBM120591GGGAGATACATGTAATTTAATAGGATAGGGTACTTAGGTCCAGACAACC242.C0300G03-33021401C12RikRIKEN cDNAC0300G03Mm.102470Chromosome 15AAGCTGTTGAA3021401C12TATGGACGTAAgeneCTGTAAATCCCAGAGTGTTTTATtTTGAGATGAGAGTT243.C0925E03-3transcribedC0925E03Mm.217865Chromosome 6TTTATCAAACAsequence withTGGAAACATCTmoderateAGAGACTATGsimilarity toGGAGAGAAAAproteinTGGGTTTTTAGpir:S12207ATATGGG(M. musculus)S12207hypotheticalprotein (B2element) -mouse244.H3083B07-5BG082983ESTsH3083B07Mm.203206No ChromosomeGGAAGTTAATABG082983locationGAACTGTTCAAinfo availableAATGTGAAAGTGGAAATAGCGTCAATAAGGAAAGCCCC245.H3056F01-3Gdf9growthH3056F01Mm.9714Chromosome 11AGTGTAGTTTTdifferentiationCAGTGGACAGfactor 9ATTTGTTAGCATAAGTCTCGAGTAGAATGTAGCTGTGAA246.J0259A06-3C88243EST C88243J0259A06Mm.249965No ChromosomeGAAAGTGGGGlocationAATGAAAAGTinfo availableATAACAAAGTAAAAAGAGAATTTCTAGGCCCTTTAGGCCC247.C0124B09-3BC0425 13cDNA sequenceC0124B09Mm.11186Chromosome 11GGTTTTCTCTTBC0425 13GTTTTATCATGATTCTTTTTATGAAGCAATAAATCCATTTCCCTGTTGG248.L0933E02-3L0933E02-3L0933E02No ChromosomeCTTTTTGAGGTNIA MouselocationTTATTTTTCCANewbominfo availableCAGTTTTCATTKidney cDNATGTTCATTAGGLibrary (Long)CATTTTCCCTTMus musculusTTACTcDNA cloneL0933E02 3′,MRNAsequence249.H3072B12-3BG069052ESTsH3072B12Mm.250102Chromosome 9AGTGTTTTTCTBG069052TTAATTCTTGAGGTTGTTATTGTAATATTTACATATAGTGCAAGAATGT250.L0266C03-3D930020B18RikRIKEN cDNAL0266C03Mm.138048Chromosome 10TAAAGTATCCAD930020B18CTGAAGTCACTgeneATGGAAAACAGCCTTTTGATTTATGGACTATTTAGCTC251.K0423B04-3Zfp91zinc fingerK0423B04Mm.212863Chromosome 19GCCTAGTTTTTprotein 91TCAGCATCAATTTTGGAAAACCTTAGACCACAGGCATATTTCGTCAAGT252.J0403C04-3AUO21859J0403C04No ChromosomeTCATTTTTCAAMouselocationGTCGTCAAGGGunfertilized egginfo availableGATGTTTCTCAcDNA MusTTTTCCGTGACmusculusGACTTGAAAAcDNA cloneATGACGJ0403C04 3′,MRNAsequence253.J0248E12-31700011103RikRIKEN cDNAJ0248E12Mm.78729No ChromosomeCTGAAAATCAC1700011103locationGGAAAATGAGgeneinfo availableAAATACACACTTTAGGACGTGAAATATGTCGAGGAAAAC254.J0908H04-3Rpl24ribosomalJ0908H04Mm.107869No ChromosomeGCGAGAAAACprotein L24locationTGAAAATCACGinfo availableGAAAATGAGAAATACACACTTTAGGACGTGAAATATGGC255.K0205H10-3MaddMAP-kinaseK0205H10Mm.36410Chromosome 2AGAAAGCTATactivating deathGGACTGGATAdomainGGAGGAGAATGTAAATATTTCAGCTCCACATTATTTATAG256.C0507E09-3Gpr22G protein-C0507E09Mm.68486Chromosome 12ACAAAAAGGTcoupledTACCTATGAAGreceptor 22ACAGTGAAATAAGAGAGAAATGTTTAGTACCTCAGGTTG257.J0005B1 1-3Mus musculusJ0005B11Mm.249862Chromosome 7CTAAGGGAGGtranscribedAAATGTTGGTAsequence withTAAAATGTTTAweak similarityAAAGAACTTGto proteinGAGGCAAACTTref:NP_083358.1GGAGTGG(M. musculus)RIKEN cDNA5830411J07[Mus musculus]258.L0201E08-3AW551705ESTsL0201E08Mm.182670Chromosome 6CCACATCATTGAW551705GAAAGAAATACACTTATCTTAATTGCCATGGAATAGGAGCATGAAAGTC259.J0426H03-3AU023164ESTsJ0426H03Mm.221086Chromosome 4ATGAGAAATAAU023164CACACTTTAGGACGTGAAATATGGCGAGGAAAACTGAAAAAGGTCTATTC260.C0649D06-3Cdkn2bcyclin-C0649D06Mm.269426Chromosome 4CCTGTGAACTGdependentAAAATGCAGAkinase inhibitorTGATCCACAGG2B (p15,CTAAATGGGAinhibits CDK4)AACCTGGAGAGTAGATGA261.J0421D03-3Rpl24ribosomalJ0421D03Mm.107869No ChromosomeGCGAGAAAACprotein L24locationTGAAAATCACGinfo availableGAAAATGAGAAATACACACTTTAGGACCAGAAATATGGC262.K0643F07-3ESTsK0643F07Mm.25571Chromosome XTGGAGGAAATTBQ563001GATTGAAAAACGATTGGTCAAATCGAAAATGGAGAAAACTCATGTTCAC263.H3103C12-3SlamflsignalingH3103C12Mm.103648Chromosome 1CTTCATCCTGGlymphocyticTTTTCACGGCAactivationATAATAATGATmoleculeGAAAAGACAAfamily memberGGTAAATCAA1ATCACTG264.J0416H11-3PscdbppleckstrinJ0416H11Mm.123225No ChromosomeACTGAAAATCAhomology, Sec7locationTGGAAAATGAand coiled-coilinfo availableGAAACATCCACdomains,TTGACGACTTGbinding proteinAAAAATGACGAAATCAC265.AF015770.1Rfngradical fringeAF015770Mm.871Chromosome 11CAAGCACTGTGgene homologCTGCAAAATGT(Drosophila)CGGTGGAATATGATAAGTTCCTAGAATCTGGACGAAAA266.C0933C05-3ESTsC0933C05Mm.217877Chromosome 1TTTGAGAAGAABQ551952AGGCATACACTTGAAATAAAGGCAAAAACATTATACTGTCTACCGAGAC267.C0931A05-3E130304F04RikRIKEN cDNAC0931A05Mm.38058Chromosome 13GAAGAAAACGE130304F04AGGTGAAGAGgeneCACTTTAGAACACTTGGGGATTACAGACGAACATATCCGG268.J0030C02-3C77383ESTs C77383J0030C02Mm.43952Chromosome 13ATCATAAAAACTGTGGAAATCCATATTGCCCTTTTAAAAGAAAACTATGGGGATGGAGAG269.H3061A07-3Srpk2serine/arginine-H3061A07Mm.8709Chromosome 5AAATGGCAGArich proteinAGAAAGGGTTspecific kinaseAATGGCTGGA2AAAATGGATCAGTAGTCTTGCAGAGGAACC270.J0823B08-3AUO41035J0823B08Chromosome 10ATTUAGGGGGMouse four-CTTTATTGUAcell-embryoCTTGACGTGGAcDNA MusATTTGAAAACTmusculusAAAAAGATGAcDNA cloneGTCTGGJ0823B08 3′,MRNAsequence271.L0942H08-3Mus musculusL0942H08Mm.276728Chromosome 11GTGGAAATCAtranscribedGAGATCTAAGTsequence withACGTTTATGCAmoderateTAGGAGTAGGsimilarity toAATGAGGGGTTproteinATTAAAGref:NP_081764.1(M. musculus)RIKEN cDNA5730493B19[Mus musculus]272.C0280H06-3Mrp150mitochondrialC0280H06Mm.30052Chromosome 4AAACCCCCCAAribosomalGTAGCCCAAAprotein L50GGCCCGCTTCCCACCAAAATGTTTTTTATGTTTTAAGGA273.L0534E07-34632417D23hypotheticalL0534E07Mm.105080Chromosome 16ATTATGATGCCproteinTGTAACACACA4632417D23GAAGTATCTGACTGTGAACGAATCAACCTCATGGATGA274.U22339.1Il15rainterleukin 15U2233916169Chromosome 2AGAAGAGATAreceptor, alphaCTGAGCCAATGchainAACCCTTTCGTGACAAAACCAAACTCAG275.L0533C12-3L0533C12-3L0533C12No ChromosomeCTGCCTTCCCANIA MouselocationTAAAAATAAANewborn HeartAGGCATGCAAcDNA LibraryAACCAATTTTTMus musculusGGCCAGGCCCcDNA cloneAGTTAAGAL0533C12 3′,MRNAsequence276.C0909E04-3MvkmevalonateC0909E04Mm.28088Chromosome 5ACAAGCCCTGGkinaseGCCTCTGAGACCACCCGACACACCATCCTACCAAGAAGCCTCTAAGTAT277.J0093B09-3Bhmt2betaine-J0093B09Mm.29981Chromosome 13CAAGTCAGCAhomocysteineAGAAGCCAACmethyltransferaseCTTGGTGAAAT2AATTCTGGTTGTTTGAAAGCTAGGTCTTG278.H3066D09-3BG068517ESTsH3066D09Mm.250067Chromosome 1GGTCAAGAGABG068517GTGCCAACTAGCTTTGTTTAAAAAATCCTAGTCCTGAATCCACAAGCCTG279.C0346F01-3BM197260ESTsC0346F01Mm.222100Chromosome 9AGTGGAAGCCTBM197260TATAAGCATTGAACCCAGGATGAGTCGCTCGTATTTCCACCTTACTCAT280.K0125A06-3Hdac7ahistoneK0125A06Mm.259829Chromosome 15CTTCCCACAACdeacetylase 7ACCCACCGTACCTTGTCTATGTATGCATGTTTTTGTAAAAAAGAAAAAAG281.J0214H07-3C85807 MouseJ0214H07No ChromosomeTGCCTGACTCCfertilized one-locationAAGAAAAGAAcell-embryoinfo availableGCCAGAACTCGcDNA MusGAACCATAGTCmusculusATCTTTAAAGAcDNA cloneTCTTCTJ0214H07 3′,MRNAsequence282.C0309H10-35930412E23RikRIKEN cDNAC0309H10Mm.45194No ChromosomeGTTAATATTAT5930412E23locationTAACTGAGCCTgeneinfo availableGCCCATACCCCCCGTGGTCATTGGTGTTGGGTGCAGTG283.C0351C04-32610034E13RikRIKEN cDNAC0351C04Mm.157778Chromosome 7GGAGGACGAC2610034E13ATCCTCATGGAgeneCCTCATCTGAACCCAACACCCAATAAAGTTCCTTTTAAC284.K0204G07-3Arf3ADP-K0204G07Mm.295706Chromosome 15 notTCTGAACCTCAribosylationplacedACCCATCACCAfactor 3ACCCCGTGTCTTCAACATTACTTCCAAAAAAGTCTGG285.L0928B09-3transcribedL0928B09Mm.217064Chromosome 10AGGAGCCTGTGsequence withTCCTTATAGAGstrongTTGGAATTAACsimilarity toTTCAGCCCTCTproteinATCTCACTTCCpir:S12207TCTGT(M. musculus)S12207hypotheticalprotein (B2element) -mouse286.H3059A09-3C430004E15RikRIKEN cDNAH3059A09Mm.29587Chromosome 2GAAAAAAGATC430004E 15GAGATCTCCTCgeneCATGACAAGAGCCTGCATACAACATTTGAGTACCCTTCT287.C0949D03-3UNKNOWNC0949D03Data not foundNo ChromosomeTTTGATTTTAGC0949D03locationCAGAAACCACinfo availableCACCAAAATTGTGCCTTAGCTGTATTTCTGTTTAGGGGA288.K0118A04-3Rgs1regulator of G-K0118A04Mm.103701Chromosome 1AGATACTATGGproteinTACTGTCATGAsignaling 1AATGCAGTGGGACTCTATTCAAACAACCCTCCAAAATG289.H3123F11-3transcribedH3123F11Mm.157781Chromosome 7AGAGAACCCAsequence withCACTCCTTTCAmoderateTCAAGACTTGCsimilarity toAGAGCATCCCAproteinCAACCAAGATref:NP_081764.1GCTATTT(M. musculus)RIKEN cDNA5730493B19[Mus musculus]290.H3154A06-3Gng13guanineH3154A06Mm.218764Chromosome 17TATGAGCCTGAnucleotideCCCACACTCTCbinding proteinTGTAAGGTGTG13, gammaACTTTATAAATAGACTTCTCCGGGTGT291.L0534E01-3L0534E01-3L0534E01Chromosome 9ATACCCCACCANIA MouseCAACCTCTCAANewbom HeartAAGAGGGCTCTcDNA LibraryTAACTTGGAAGMus musculusGATAAAATAAcDNA cloneATCAGGL0534E01 3′,MRNAsequence292.L0250B10-3Ap4m1adaptor-relatedL0250B10Mm.1994No ChromosomeTATCCTCCCACproteinlocationAAAGATGAGAcomplex AP-4,info availableGGAGCCCATCCmu 1AGTGTTACTGTTAGAAGTCACAGTGAAA293.L0518G04-3BM12304SESTsL0518004Mm.221745Chromosome 3TATTGTCCAATBM123045GAAACCCACAAACTACCCTCTATCTGGAGTTGGAACATTTATCTGCATT294.J1020E03-3transcribedJ1020E03Mm.250157Chromosome 9TAAGGAGACTsequence withGCCCTACAAAAmoderateCTACGATACTAsimilarity toCTATCACTTTAproteinAAAATTAGTGTpir:S12207AAAGGG(M. musculus)S12207hypotheticalprotein (B2element).mouse295.X12616.1Fesfeline sarcomaX12616Mm.48757Chromosome 7TCAAGGCCAAoncogeneGTTTCTGCAAGAAGCAAGGATCCTGAAACAGTACAACCACCCCAACATTG296.J0026H02-3C77164expressedJ0026H0297587Chromosome XGATTGCCAGAGsequenceACTTACACTTAC77164ATAGAGTCATAAAGCCCATAGAGCCTGAGTGAGAGCCA297.H3154D11-5Taf71TAF7-likeH3154D11Mm.103259Chromosome XTTATTCCTGAARNAGCCCCCGCTACpolymerase II,AGATGTTTCCATATA boxCAACCGAAGAbinding proteinAGCGGTCTCCA(TBP)-AAGAGCassociatedfactor298.H3054H04-3Kcnn4potassiumH3054H04Mm.9911Chromosome 7AGCTCCACATGintermediate/smAACTCACAGAall conductanceAGAACCAGGCcalcium-TAAGTACCCAAactivatedGGACCGAGCTCchannel,AAGGACAsubfamily N,member 4299.J0425B03-3R75183expressedJ0425B03Mm.276293Chromosome 15ACCATTATTCTsequenceTTTAAAAAACCR75183CAAAAACCACCAGCAAGGGGGCCTTTGGTTGGCCTCAA300.C0930C02-30610037D15RikRIKEN cDNAC0930C02Mm.218714No ChromosomeCTTCATCTTAA0610037D15locationAACTCCAGAACgeneinfo availableAACTCCCTTCCTAACCTGGAACCCAGCAGCTTTCAGTT301.L0812A11-3ESTs B1793430L0812A11Mm.261348No ChromosomeCTGCACGCCCClocationAGGAGCCTGGinfo availableGTGAAGCATCACAGCACTAAGTCATGTTAAAAGGAGTCT302.J0243F04-39530020D24RikRIKEN cDNAJ0243F04Mm.200585Chromosome 2CACTGGAGCAC9530020D24TGAACATGATGgeneTACAAGTATCACACAGAAAAGCAGCACTGGACTGTACT303.C0335A03-31110035014RikRIKEN cDNAC0335A03Mm.202727Chromosome 12ATAAGAACTTA1130035014TAGGAACCCCAgeneACTCCCCATGAAAAATATAAGACCTCAAGGCCTGGGGA304H3003B10-3BG063111ESTsH3003B10Mm.100527Chromosome 3GCCCACCAACTBG063111CTAATTTGTGCTACTTATATATATTCCTGGGAGTAGGACTGTCCTCCTG305U97073.1Prtn3proteinase 3U97073Mm.2364Chromosome 10CAGTCAGGTCTTCCAGAACAATTACAACCCCGAGGAGAACCTCAATGACGTGCTTCTCCT306.K0300D08-3AfmidarylformamidascK0300D08Mm.169672Chromosome 11CGTAGCTCGCTGGTAGAAAGCCTGACCACCATGCATACGATCCTGGGTTTCAACAAGGAA307.H3029H06-3Sf3b2splicing factorH3029H06Mm.196532Chromosome 19GAGCCTGAGAT3b, subunit 2CTACGAGCCCAATTTCATCTTCTTCAAGAGGATTTTTGAGGCTTTCAAG308.H3074D09-3Drg2developmentallyH3074D09Mm.41803Chromosome 11GAGTCTGTGGGregulatedTAUCGCCTGAGIP bindingACAAGCATAAprotein 2GCCCAACATCTATTTCAAGCCCAAGAAA309.K0647G12-3PlekpleckstrinK0647G12Mm.98232Chromosome 11AGCATCAAACAAAGCACATAAACTCGTACATAAGCAAGGGATGTCCTTATTGGTCAAACA310.H3137A08-3Mus musculusH3137A08Mm.197271Chromosome 2GGGAAAAAATtranscribedAGCAAAACCCsequence withCAAACTCCACAmoderateACCACAAAAAsimilarity toCCTGTTAATTAproteinTGGTGGCApir:S12207(M. musculus)S12207hypotheticalprotein (B2element) -mouse311.C0166D06-3Slc38a3solute carrierC0166D06Mm.30058Chromosome 9ACACAGAGCCfamily 38,AGAAAACCCAmember 3GGCCTGAAGACATCCCCTAGTCCTGCTGAGAGACCACAGT312.K0406B07-3Sirt7sirtuin 7 (silentK0406B07Mm.259849Chromosome 11CGACCAATCTGmating typeCCTGGGAAACinformationAACACCCCACAregulation 2,GAACGGGGCTThomolog) 7 (S.CAGAAACACGcerevisiae)TGAGTGA313.H3085D10-3GdaguanineH3085D10Mm.45054Chromosome 19GTTTAGGTGAGdeaminaseTTTTCCATTGTATCTTATAACAGAGAAACCCATTAGGCAGTAGTTAGTTC314.H3099C09-3Igf1insulin-likeH3099C09Mm.268521Chromosome 10TCGAAACACCTgrowth factor 1ACCAAATACCAATAATAAGTCCAATAACATTACAAAGATGGGCATTTCC315.H3099B07-52610028H24RikRIKEN cDNAH3099B0776964No ChromosomeTGCTACCCTCC2610028H24locationAGGACCAACGgeneinfo availableATGGATGCACCACGGAGTCCCAAGAGCTGAAAAGCAGAA316.H3114H10-3Rec8L1REC8-like 1H3114H10Mm.23149Chromosome 14CGGAGCTCTTC(yeast)AGAACCCCAACTCTCTCTGGCTGGCTACCCCCAGAACTCCTAGGTTTAT317.L0703E03-3Lipclipase, hepaticL0703E03Mm.362Chromosome 9ATAAAGAGAATTCCCACCACCCTGOGCGAAGGAATTACCAGCAATAAAACCTATTCCTTC318.H3074H08-3BG069302ESTsH3074H08Mm.11484Chromosome 7:notACTTTCAAGTCBG069302placedTGAATCCTATGAGCCTGAAGTGAGATCTTATTTAGAAACAGAACCCCAA319.K0443D01-3BazlbbromodomainK0443D01Mm.40331Chromosome 5GACAAGCCCTTadjacent to zincAGGGAGCCAGfinger domain,AAAAAGAGCA1BGGAAGAAGTTAAAATGTTTAATTTTTTAA320.J0409E10-3AU022163ESTsJ0409E10Mm.188475Chromosome 16GCCCAAGAGCTAU022163AGAAAACCTACTCTATGTGTAGAGATACTTCCTATTAAAATAATAGTAC321.L0528E01-3BM123655ESTL0528E01Mm.216782Chromosome 9CTCCACTTTTABM123655AAGTCTGTAGGAATAGGAGCCGATTAGACAACTCTCGGTCTCATGCTCA322.L0031B11-3AlcamactivatedL0031B11Mm.2877Chromosome 16TTTCTGGGATCleukocyte cellCCACTGCACCGadhesionCCATTTCTTCCmoleculeCAGATTTATGTGTATAACTTAAACTGG323.G0115A06-3Femlafeminization 1G0115A06Mm.27723Chromosome 17ATACAGTAGAThomolog a (C.GCTGAACACACelegans)TTGAGTCCATCATGAGGGGGTAATAAGTCTCACCAGCA324.L0947C07-3Malmyelin andL0947C07Mm.39040Chromosome 2TCTTATACTTTlymphocyteCAACAAAGCTprotein, T-cellGAACCCTAACAdifferentiationTTACACTAACCproteinAGCAGCTCAACACGAGT325.H3101A05-3AU040576expressedH3101A05Mm.26700Chromosome 7CTGAATGTATAsequenceCACACCCACAGAU040576GAGACTGTGGCTGAGCGTTCATCCAAATAAATTTGAAT326.H3064E10-3BG068353ESTsH3064E10Mm.35046Chromosome 4GTTCCTGTTCABG068353GAGTGCCTGAAAACCCAAAGTGTCTGAGAGTCTGAAGGAATTCAACTGT327.K0505H05-3Ian6immuneK0505H05Mm.24781Chromosome 6AAACACCCACassociatedACTTGAAACTTnucleotide 6CCATGAACCCACTCAAATTCATTTCTATCCCCCTTTGGA328.H3082E12-3Ptpreprotein tyrosineH3082E12Mm.945Chromosome 7TCATGGAGATAphosphatase,TAACTATAGAGreceptor type, EATAAAGAGCGACACCCTGTCTGAAGCAATCAGCGTCCG329.H3088A06-32310047N01RikRIKEN cDNAH3088A06Mm.31482Chromosome 4GGACACTGTGA2310047N01ACACTGTGTGGgeneACAGAGCCCACAACTTCTCCATTTGTGTCTGGCAGCAA330.K0635B07-3Ccr5chemokine (C-K0635B07Mm.14302Chromosome 9AGGAAAGAAAC motif)GGGGTTAGAATreceptor 5CTCTCAGGAGATTAAAGTTTTCTGCCTAACAAGAGGTGTT331.C0153A12-31110025F24RikRIKEN cDNAC0153A12Mm.28451Chromosome 16CTCAAGACTTT1110025F24GCCAACATGTTgeneCCGTTTCTTACACCCTGAACCCTGATCGGAACATTCAT332.C0143E02-3BC022145cDNA sequenceC0143E02Mm.200891Chromosome 11TCTGTACATGGBC022145CCGAAAATCAGAGTCCACCATATTCTTTTGAATATCCAGGGTTCTCTGA333.L0863F12-3Nr2c2nuclear receptorL0863F12Mm.193835Chromosome 6TTCTGGCTCCTsubfamily 2,TATTTCAGTTCgroup C,TCTTTAAAACCmember 2AGTTCAACACCAGTGTGTTAAAAAGAA334.H3045F02-3LOC214424hypotheticalH3045F02Mm.31129Chromosome 9GCAGATTTAACproteinAACTAGCAACTLOC214424CTGTCATCTTTTTCTAAAAATGACCAACTGCTGATTAC335.H3035005-3BG065832ESTsH3035G05Mm.154695Chromosome 17CTTAAAAAGGBG065832GAGATACAGTTTTACTCTGATCCAGCAAATCTAGTTAAGACACTAGAATG336.H3137D02-3HnrplheterogeneousH3137D02Mm.9043Chromosome 7CTTCCTGAACCnuclearATTACCAGATGribonucleoproteGAAAACCCAAin LATGGCCCGTACCCATATACTCTGAAGTT337H3097F07-3AU040829expressedH3097F07Mm.134338Chromosome 11GTAACGGAGCsequenceCTGGGGGTTGAAU040829AGGTTATCTTTACATATATGTACAAACTGTTGTCAAGAG338.J0029C02-3Frag 1-pendingFGF receptorJ0029C02Mm.259795Chromosome 7TCCCCACCACTactivatingCATGGGGATCTprotein 1TCAAGAAGCATCACCATTCACTGAAAGGTCCTAAAAAA339.BB416014.1Mus musculusBB416014Mm.24449Chromosome 10GCGCAGAGGCB6-derivedAAACCAACGTCDII + veGGAGCCAGACdendritic cellsATTGGTGAACCcDNA, RIKENCAACCTATCCAfull-lengthCACCTTCAenrichedlibrary,clone:F730035A01product:similarto SWI/SNFCOMPLEX170 KDASUBUNIT[Homosapiens], fullinsert sequence.340.H3087E01-3Anxa4annexin A4H3087E01Mm.259702Chromosome 6CTTATTTTAGACAGATCCAAAGTTCTCACAAGCCCCCTTTCTTTGCTCTGCCTATCATCG341.H3088E08-3BG070548ESTsH3088E08Mm.11161Chromosome 8AACCTCTGAACBG070548CTAATCACTGTGGATTCCCACCAACACCATATATGAAAATGCAGGCCGA342.AF179424.1Mus musculusAF179424Mm.1428Chromosome 14TGCGGAAGGA13 days embryoGGGGATTCAAmale testisACCAGAAAACeDNA, RIKENGGAAGCCCAAfull-lengthGAACCTGAATAenrichedAATCTAAGAlibrary,clone:6030408M17product:GATAbinding protein4, full insertsequence343.J0258C01-3Mus musculusJ0258C01Mm.275718Chromosome 2CCCTAGTCCGTmRNA forTTTCTGATCAGmKIAA1335TCAGAACCCACproteinAATAACTACTAGTAGTCCTGTGGCTTT344.K0507B09-3ESTsK0507B09Mm.218038Chromosome 9GTAGCCACCAABM238095GCCACAAGTAACAAATGATCTCTGTGAATGCCATATGGAAACTTTTATT345.L0846F07-3BM117131ESTsL0846F07Mm.216977Chromosome 9GGCTCCATTTCBM117131TGAACTCTGTGTTAAGCTAATAAGATTTTAAATAAACGCTGATGAAAGC346.U48866.1CEBPECCAAT/enhancerU48866Hs.158323No ChromosomeTGCTGGGGGCCbindinglocationTAGAACCCTGAproteininfo availableGACATAGACC(C/EBP),ATGGATAAATGepsilonGCAACCGGGGTGGCAAA347.K0301B06-3FechferrochelataseK0301B06Mm.217130Chromosome 18AACGCAAAGAGCAAGAACCAAACAAAGACAGGAACAACTCGCAGAAGAAATCCCGCCTGG348.NM_009756.1Bmp10boneNM_009756Mm.57171Chromosome 6TGTTTTCTGATmorphogeneticGACCAAAGCAprotein 10ATGACAAGGAGCAGAAAGAAGAACTGAACGAATTGATCA349.NM_010100.1Edarectodysplasin-ANM_010100Mm.174523Chromosome 10CCCACCACTGAreceptorATATAGACCATACTGTGAGAGGACCATAATTAGGTCCTGAATTTTTAAT350.G0115E06-3C430014D17RikRIKEN cDNAG0115E06Mm.103389Chromosome 3GTATGACTTCCC430014D17AACCAGAAAAgeneAGGCTCTAAAAGCTGAACACACTAACCGGCTGAAAAACG351.L0266D11-3Ppp3caproteinL0266D11Mm.80565Chromosome 3CTTCTGGCTCCphosphatase 3,CTTACATGAAGcatalyticGACTGATTTAAsubunit, alphaGAAACCAGACisoformCATTCCTTTACTTTGAA352.L0526F10-3Mus musculusL0526F10Mm.215689Chromosome XGCAGGGTGCTT10 days neonateACTTTCTCAGAcortex cDNA,GCCTGAAGTTARIKEN full-CTTCCATTGTTlength enrichedTTGGCACTGAAlibrary,TAACAclone:A830020C2 Iproduct:unknownEST, fullinsert sequence.353H3047C10-3Slc6a6solute carrierH3047C10Mm.200518Chromosome 6TTAGCACAAGAfamily 6GAAAAGCTGA(neurotransmitterGAACGTGGGTTtransporter,TTGCCTCCTTCtaurine),AGAAATATGTCmember 6TGGCTC354K0322G06-3BC042620cDNA sequenceK0322G06Mm.152289Chromosome 17ACACAGCACCCBC042620ACAACTAATCTTGGGACACCCCTATCTGGTTGGAAGAGAGTAAACTAAT355.NM_009580.1Zp1zona pellucidaNM_009580Mm.24767Chromosome 19CAATGGCCTATglycoprotein 1TCTGTCAGATGGGTGTCCTTTCAAGGGTGACAACTACAGAACACAAGTA356.H3150E08-3Map4k5mitogen-H3150E08Mm.260244Chromosome 12AAAGTAGGTTCactivatedACACAGTAAAprotein kinaseGGGATAATACCkinase kinaseATCTGGAACAAkinase 5TGATCAGTGTAGAGTTA357.J0059G03-3C79059ESTs C79059J0059G03Mm.249888Chromosome 4CACCTGGGTCTACAGCTACTCTGATTCTACAAAGACAGGGTCAAGCATCTCTAACAAAGT358.U93191.1Hdac2histoneU9319115182Chromosome 10TATTAAACCCAdeacetylase 2GGAGATACAAGGAGTCTGCCATTAACCTCTCTGTAACTCAAGAGTAGTT359.H3033C04-5H3033C04-5H3033C04No ChromosomeTTCCTCCCAAANIA MouselocationATGGAGTTTCC15K cDNAinfo availableTCTTCAAACCAClone Set MusCAGCTCCCCCAmusculusAGATCTATCCTcDNA cloneGATATH3033C04 5′,MRNAsequence360.H3085C01-32700038N03RikRIKEN cDNAH3085C01Mm.21836Chromosome 5TATGTCTTGAT2700038N03ACTGGACCCACgeneACTACTGGGGCACTCCAAAAAACCGTTGTGAACTACAA361.J0412G02-3BB336629ESTsJ0412G02Mm.208743Chromosome 11AGTAAAGGGCBB336629ACCGGAAATGTTAAATCCTTGTTTAGGATATGAAAGGAATTAGGGGATGG362.K0527H09-3BM239048ESTsK0527H09Mm.217288Chromosome 11GAATGTCTGATBM239048ACATGACCCATCAGTTAGGAACCACTGAACTAGAGGAGTAGCTAAACTC363.H3009C10-3Serpinb9bserine (orH3009C10Mm.45371Chromosome 13GCTTCTACTGGcysteine)CTCTTGTATGCproteinaseATATGTGCACTinhibitor, dadeTATCCAGACTGB, member 9bAGGATTTTACAAAGCA364.H3142D11-3Mus musculusH3142D11Mm.113272Chromosome XCTGTCTAAGCGmRNA similarCTGAACCACTTto hypothelicalAGCAGAAATGproteinACACCCATATGFLJ2O811AGAGCTTGTGC(cDNA cloneCAAATAMGC:27863IMAGE:3492516), completecds365.H3094B07-3Mus musculusH3094B07Mm.173357Chromosome 14AAAGGAGACTtranscribedGCATCAGGTATsequence withTCTGATAGAGAweak similarityGCTGAGGAAGto proteinAGATTGAGGTAsp:P11369TGGGATT(M. musculus)POL2_MOUSERetrovirusrelated POLpolyprotein[Contains:Reversetranscriptase;Endonuclease]366.J0068F09-3C79588ESTs C79588J0068F09Mm.234023No ChromosomeTGACTGGAATClocationACCACCCTTGCinfo availableCTGAGTTTGCGATCTCACAGTTGGAACTGAGAGTTTCC367.H3039B03-5EO30024M05RikRIKEN cDNAH3039B03Mm.5675Chromosome 12GGATCAGATGE030024M05ATGCACCATUGgeneCTTTCCATTTGCTACATTTAAAATCTTTTACTAGTCAACC368.H3068B03-3BG068673ESTsH3068B03Mm.11978Chromosome 1TTGAGACCTTABG068673AAGAAATAACAAACTCAAGGAAGATTAGGGTCCAGTGTTTAAGTCATGG369.C0250F05-3BM203195ESTsC0250F05Mm.228379Chromosome 12GTCTCCTTTGTBM203195GTTATTGCCTTCCCAACACTTCTAAGTCCCAGCTCAACAGCTACTTCTA370.H3110C11-3MlphmelanophilinH3110C11Mm.17675Chromosome 1CACAGCTGCTTGTAGTCATCATTCCAGTGAGGAGTAAGAAGAATTTTATGTGTGTCTCTA371.H3121F01-3Wnt4wingless-H3121F01Mm.20355Chromosome 4AACTTAAACAGrelated MMTVTCTCCCACCACintegration siteCTACCCCAAAA4GATACTGGTTGTATTTTTTGTTTTGGT372.J1012G09-3Brd3bromodomainJ1012G09Mm.28721Chromosome 2CAGCAGAAAAcontaining 3GGCTCCCACCAAGAAGGCCAACAGCACAACCACAGCCAGCAGGATGTGTT373L0952B09-3Usp49ubiquitinL0952B09Mm.25072Chromosome 17GGCTTCACATCspecificTAAGTGGGGAprotease 49CTATTTTAACTTATTTACAGGTATATGGTGTGGAAATAA374.K0131B12-3I14rainterleukin 4K0131B12Mm.233802Chromosome 7CGCTCAGTTGTreceptor, alphaAGAAAGCAACAAGGACACAAACTTGATTGCCCAAAGTCACTGCCAGTTA375.H3046E09-3Nfatc2ipnuclear factorH3046E09Mm.1389Chromosome 7GTCTGAACACAof activated T-CTATTATGTATcells,CCATCCAATCTcytoplasmic 2CAACTGAATAAinteractingAGGGAGATGCproteinCTTTTG376.K0520805-3transcribedK0520B05Mm.221547Chromosome 14AAAGAATTTCAsequence withAGAACGAAGCweak similarityATAGGTGGTTAto proteinTGTAGTTTGATpir:158401TACAGAAAAG(M. musculus)AGATGCC158401 proteintyrosine kinase(EC2.7.1.112)JAK3 - mouse377.K0315G05-3Stat5asignalK0315G05Mm.4697Chromosome 11AAACCACCTTCtransducer andAGTGTGAGGAactivator ofGCCCACGTCAGtranscriptionTTGTAGTATCT5ACTGTTCATACCAACAAT378.H3086F07-3BC003332cDNA sequenceH3086F07Mm.100116Chromosome 6GCACTCCAGCCBC003332TGATTCTTTGAGACTTTGGGGTACACATATTGAAAGTACTTTGAATTTG379.H3156A10-5Ctsdcathepsin DH3156A10Mm.231395Chromosome 7ACTGTATCGGTTCCATGTAAGTCTGACCAGTCAAAGGCAAGAGGTATCAAGGTGGAGAAA380.C0890D02-3C0890D02-3C0890D02Chromosome 18GTGTTTGAATTNIA MouseAAAACCCCCACBlastocystCCTCGGAGGCCcDNA LibraryTTTAAAGAAAT(Long) MusGGTTTTTGTCCmusculusGTTGTcDNA cloneC0890D02 3′,MRNAsequence381.L0245G03-36430519N07RikRIKEN cDNAL0245G03Mm.149642Chromosome 6CTCTCGACAAA6430519N07ATATAAATGGAgeneCAGTACCAAACTAAGAGGGATATAAGTGGGAGCAAAGG382.J0447A10-3Mus musculusJ0447A10Mm.202311Chromosome 11TATGGTACGAGcDNA cloneTTTAGGGCTTAIMAGE:12820GTCAGTTTACA81, partial cdsATGGGGATTGAATTTTGTGTCAAAACC383.J1031A09-3Mus musculusJ1031A09Mm.235234No ChromosomeCTGGCTCCTACtranscribedlocationTGGCAACAGGsequence withinfo availableCATACTTGTGGweak similarityTTTAATACAGAto proteinGAAACAAAACpir:158401ATTCATA(M. musculus)158401 proteintyrosine kinase(EC2.7.1.112)JAK3 - mouse384.L0072H04-3A630084M22RikRIKEN cDNAL0072H04Mm.27968Chromosome 1TTTGACCTAATA630084M22GAAATACCCATgeneTTCATCTGTGACAACACATAGCCCAGTAAACATCACTG385.J0050E03-3transcribedJ0050E03Mm.37806Chromosome 14CCTGTTCCTAGsequence withTATCCTGOCGTweak similarityCCACATATACCto proteinCAAAGTTAGGCref:NP_081764.1ATACTAACCAA(M. musculus)GAGATRIKEN cDNA5730493B19[Mus musculus]386.H3039C11-3Tyro3TYRO3 proteinH3039C11Mm.2901Chromosome 2CTGGAACTCAGtyrosine kinaseCACTGCCCACC3ACACTTGGTCCGAAATGCCAGGTTTGCCCCTCTTAAGT387.C0324F11-36720458F09RikRIKEN cDNAC0324F11Chromosome 12CCTGGAGGTCT6720458F09CCACCTGAAGTgeneTCCCTGATGCAGGGTCAGTCCAGCCTTGGTAAGGGCCA388.L0018F11-3AW547199ESTsL0018F11Mm.182611Chromosome 12AAATGAGAACAW547 199CAGATTACCAAAATTACCACTACCACCAAAATAACCCCTCTGATTCCTTG389.X69902.1Itga6integrin alpha 6X69902Mm.225096Chromosome 2CAGATAGATGACAGCAGGAAATTTTCTTTATTTCCTGAAAGAAAATACCAGACTCTCAAC390.H3105A09-3transcribedH3105A09Mm.174047No ChromosomeGGTGCCAAATGsequence withlocationCGGCCATGGTGweak similarityinfo availableCTGAACAATTTto proteinATCGTCAGAGGref:NP_416488.1GGAAGAACAG(E. coli)TTGACCputativetransportprotein,shikimate[Escherichiacoli K12].391.H3159F01-5UNKNOWNH3159F01Data not foundNo ChromosomeCCAAAACAGAH3159F01locationGCCAACACCACinfo availableCGACAACAACCCCACAGCAAACCCGGAGAGAAACCCAAA392.K0522B04-3F5coagulationK0522B04Mm.12900Chromosome 1TTTCAACCCGCfactor VCCATTATTTCCAGATTTATCCGCATCATTCCTAAAACATGGAACCAGAG393C0123F08-3A1843918expressedC0123F08Mm.143742Chromosome 5TGGAGACTGAsequenceGTTCGACAATCA1843918CCATCTACGAGACTGGCGAAACAAGAGAGTATGAAGTTT394H3067G08-3BG068642ESTsH3067008Mm.250079Chromosome 11GATACAACAGBG068642CATCTGTTTTCCAAGGAGAAATCATTTGAGGAACAAAACCTATCAAGAGA395.K0349B03-3Stam2signalK0349B03Mm.45048Chromosome 2AACTAGAAAAtransducingCATAGATGCACadaptorAGGACTCGGATmolecule (SH3CCATGATATTTdomain andACACTGGGAAITAM motif) 2ATGTTCT396.C0620D11-3BidBH3 interactingC0620D11Mm.34384Chromosome 6ATCTCAAGATTdomain deathTCTATCCAAGTagonistGGAAACAAACTGAATCATGCACACGACTTATCTGTGTG397.C0189H10-34930486L24RikRIKEN cDNAC0189H10Mm.19839Chromosome 13AGAGGAGCCA4930486L24CACTTGATGTGgeneAATTAAACTCATAAACATTATGCCACTAACAGCTTTTAT398.H3140A02-3Slc9a1solute carrierH3140A02Mm.4312Chromosome 4CTGCCGCCTGTfamily 9ACAAAGGAAA(sodium/hydrogenCTGAACCTTTTexchanger),TCATATTCTAAmember 1TAAATCAATGTGAGTTT399.K0645B04-3Smc411SMC4K0645B04Mm.206841Chromosome 3AAGCTGAGATTstructuralAAACGGCTACmaintenance ofACAATACCATCchromosomesATAGATATCAA4-like 1 (yeast)CAACCGAAAACTCAAGG400.C0300008-36720460106RikRIKEN cDNAC0300008Mm.28865Chromosome 4GACTTGGGAA6720460106AACAATGCAAgeneCTCCCATAAACCAAAACTCCAATTCCATGCCTAACTTGCT401.M59378.1Tnfrsf1btumor necrosisM59378Mm.2666Chromosome 4AGCAGGGAACfactor receptorAATTTGAGTGCsuperfamily,TGACCTATAACmember 1bACATTCCTAAAGGATGGGCAGTCCAGAA402.NM_009399.1Tnfrsfl 1atumor necrosisNM_009399Mm.6251Chromosome 1AGCTCCAACTCfactor receptorAACAGATGGCTsuperfamily,ACACAGGCAGmember 11aTGGGAACACTCCTGGGGAGGACCATGAA403.C0168E12-32810442122RikRIKEN cDNAC0168E12Mm.103450Chromosome 10ACTAGCTGCAT2810442122TGTAAAGAAAgeneCAAATCGAAACTGAGTCTTTTCACATATTGTGACGGACA404.L0228H10-3CLrcomplementL0228H10Mm.24276Chromosome 6GTAGGGTCATCcomponent 1, rATACACCCAGAsubComponentCTACCGCCAAGATGAACCTAACAATTTTGAAGGAGACA405.H3088B10-3BG070515ESTsH3088B10Mm.11092Chromosome 11TCCCCACCACGBG070515AATTATCGTGGCTAGTGGATGAAGGCCACTAATACAGGTTCAAATTGTT406.K0409D10-3Lrrc5leucine-richK0409D10Mm.23837Chromosome 5TATGTGCATAGrepeat-GCTGGAGTTTTcontaining 5GGTTATACATGGTACACTTTTGGGCCAATATAATAGGA407.H3056D02-3transcribedH3056D02Mm.9706Chromosome 12CCACACTCCCTsequence withGGAGACAATGmoderateTCTGCCATTTTsimilarity toTGCATCACTTGproteinTCAAACCACTAref:NP_079108.1ACTTCT(H. sapiens)hypotheticalproteinFLJ22439[Homo sapiens]408.J0430F08-3AU023357ESTsJ0430F08Mm.173615Chromosome 6TCGGTTGACCTAU023357GATTCCACCAAGGAGAAGGAGATCAAGGAAGAGTAAACTGTAAGAGCAT409.H3158C06-32810457106RikRIKEN cDNAH3158C06Mm.133615Chromosome 9GAGTGCTTTGA2810457106TGGTTGTTAGGgeneGACCGTAAGAATAGTCCTGTGTCAGACAGCAGATTCTA410.M85078.1Csf2racolonyM85078Mm.255931Chromosome 19AACTGTCATAAstimulatingAATCCAACGTGfactor 2CCTTCATGATCreceptor, alpha,AAAGTTCGATAlow-affinityGTCAGTAGTAC(granulocyte-TAGAAmacrophage)411.C0145E06-3Satb1special AT-richC0145E06Mm.289605Chromosome 5ACTCTCATCTGsequenceTAAAGCCTTCCbinding proteinCATCTCATTAT1TCCTTGCACTAACCACAGCCACTAGGA412.H3015B08-3BG064069ESTsH3015B08Mm.197224Chromosome 11CAGACTGAAABG064069GGAAATTCCAAAGAAAACAAAAACCTTTCAATCTATGAACTCAATGGCTG413.C0842H05-3Fbln1fibulin 1C0842H05Mm.219663Chromosome 15CTGAGAATAACCTACTACCACCTCTCTTTTCCCACCAACATCCAAGTGCCAGCGGTGGTT414.G0117D07-3Otx2orthodenticleG0117D07Mm.134516Chromosome 14AGCGACATGChomolog 2AACCAAATACC(Drosophila)ACTCAAAACAAAAATCCAGCAAAACTGAGTTGTGAGGGA415.L0806E03-3Stmn4stathmin-like4L0806E03Mm.35474Chromosome 14GTTTGTACATGTAAAAGATTGACCAGTGAAGCCATCCTATTTGTTTCTGGGGAACAATGA416.H3073B06-3BG069137ESTsH3073B06Mm.173781Chromosome 3ACTTAGACCACBG069137AACAGCATCTAAGCATCATTACCTTAAGTACTAAAGCAAAAATCTAGTC417.H3082G08-3Myo10myosin XH3082G08Mm.60590Chromosome 15TAAACCACTCTTAAACTGCTGGCTCCAGTGTTTTTAGAATGATATGAAGTCATTTTGGAG418.C0141F07-3C3arlcomplementC0141F07Mm.2408Chromosome 6AGTAAGTGCCAcomponent 3aTTATCCACCCAreceptor 1ACTACCAACCAATGCCTAAGCAGATTCTATATCTTAGC419.K0525G09-35830411120hypotheticalK0525G09Mm.31672Chromosome 5GCTTCTGGCAGproteinAGATCTGTTTA5830411120GCATAGTGTGGTATTAATTATAGCAAATGTTAAGGTAG420.H3064D01-3transcribedH3064D01Mm.250054Chromosome 15GTTGTCTGAATsequence withAATAGCACCCAweak similarityAGAAAAAGTGto proteinTGGAGATCAGTref:NP_001362.1AGGTATTCATT(H. sapiens)AAGCATdynein,axonemal,heavypolypeptide 8[Homo sapiens]421.C0120F08-36330406L22RikRIKEN cDNAC0120F08Mm.5202Chromosome 10TAAAGGAGCTT6330406L22TCCACATGAACgeneTCACAATTTTCTTGAAATAAACTTCTTAACCAACTGCC422.H3105G04-3Map4k4mitogen-H3105G04Mm.987Chromosome 1GTCACTTGGATactivatedGGTGTATTTATprotein kinaseGCACAAAAGGkinase kinaseGCTCAGAGACTkinase 4AAAGTTCCTGTGTGAAC423.J0800D09-32310004L02RikRIKEN cDNAJ0800D09Mm.159956Chromosome 7GTCATGAACCC2310004L02AATACACTGTGgeneGAAATGTGTGATTCTTTATATTAAACGTCTGCTGTTCA424.L0226H02-35830411120hypotheticalL0226H02Mm.31672Chromosome 5TGTCGATACCAproteinTCTAAAGACCA5830411120CAACTTCTAGCCATAGGGTATTTCATATATGTCCATTT425.L0529D10-3BM123730ESTsL0529D10Mm.221754Chromosome 7ATGCAAACCTABM123730AAAAGCACCCAAAAAATTCACATTGGACTGAAGAAGAGTGATCCAAGCA426.H3088E05-3Glagalactosidase,H3088E05Mm.1114Chromosome XTTTGAGACCCTalphaTTCATAAGCCCAATTATACAGATATCCAATATTACTGCAATCATTGGAG427.K0621H11-3K0621H11-3K0621H11Chromosome 13ACCTAAATTTCNIA MouseCACAGGCAACTHematopoicticTACTTTGTTATStem Cell (Lin-TAAATTTGGGG/c-Kit-/Sca-1+)ATCATATCCTGcDNA LibraryTGCCC(Long) MusmusculuscDNA cloneNIA:K0621H11IMAGE:30070846 3′, MRNAsequence428.C0846H03-3D330025I23RikRIKEN cDNAC0846H03Mm.260376Chromosome 9TTTTTTCAGACD330025I23TTAAGAACAGCgeneTAAACAAAACCTTCCTCTAGCTTTTTCATCACATCCAG429.J0058E06-3C78984ESTs C78984J0058E06Mm.249886Chromosome 17ATAATGATGATGATAACAACAAGAAAACAGACTCGAACCTAAAGACGCTGGTCTCAGATA430.K0325E09-3Ibspintegrin bindingK0325E09Mm.4987Chromosome 5CGCAAACATACsialoproteinCCTGTATAAGAAGGCTCCTAACGAGAGATTTATTAACAACACTATATAT431.K0336F07-3Pycspyrroline-5-K0336F07Mm.233117Chromosome 19TTTGACTGGGAcarboxylateCCAGCCCAGCCsynthetaseATTCTCAGCCT(glutamateCTCGACATGTAgamma-ATTTCATTTCTsemialdehydeTTTACsynthetase)432.H3013B04-3B230106124RikRIKEN cDNAH3013B04Mm.24576Chromosome 3AGGACTCATAGB230106124ACTTACAGAATgeneGATGCCGAATGGAATGTTTTGTGCATGACCTTTTAACC433L0238A07-3MidnmidnolinL0238A07Mm.143813No ChromosomeCCACCTCGCCClocationAAGTCTCCTTTinfo availableTACTGAAATAAAATTTGAGGGGAAGAGAAAAAATTTAC434.L0929C04-3Tnfrsfl lbtumor necrosisL0929C04Mm.15383Chromosome 15: notGATGTTCTTCTfactor receptorplacedGTAAAAGTTACsuperfamily,TAATATATCTGmember 1 lbTAAGACTATTA(osteoprotegerin)CAGTATTGCTATTTAT435.L0020F05-36330583M11RikRIKEN cDNAL0020F05Mm.23572Chromosome 2CTTAAGATTCA6330583M11GGAAAATGGTTgeneCTTTCTGCCCTTCCTAGCGTTTACAGAACAGACTCCGA436.H3012H07-3Cd44CD44 antigenH3012H07Mm.24138Chromosome 2TATATTGACATCCATAACACCAAAAACTGTCTTTTTAGCTAAAATCGACCCAAGACTGTC437.K0240E11-3Myo5amyosin VaK0240E11Mm.3645Chromosome 9TCTTTAGTGCTGCATTTAAGTGGCATACAAAATACAATCCCATATGTATGAACTGTTGTG438.K0401C06-3Col8a1procollagen,K0401C06Mm.86813Chromosome 16AATCTATGCCAtype VIII, alphaGATACTGTATA1TTCTACCATGGTGCTAATATCAGAGCTAAATGATACTC439.C0917F02-3Frzbfrizzled-relatedC0917F02Mm.136022Chromosome 2AATTTACACATproteinGTGGTAGTAGTAGGTCCAGATTCCTAAGTTACAGTGTGCTGAAAAATAA440.H3104C03-31500015O10RikRIKEN cDNAH3104C03Mm.11819Chromosome 1ATGAGGCTAA1500015O10ATTTGAAGATGgeneATGTCAACTATTGGCTAAACAGAAATCGAAACGGCCATG441.K0438D09-3Col8alprocollagen,K0438D09Mm.86813Chromosome 16TCTACTACTTTtype VIII, alphaGCTTATCATGT1TCACTGCAAGGGAGGCAACGTATGGGTTGCTCTCTTCA442.H3152C04-3Usp16ubiquitinH3152C04Mm.196253Chromosome 16GTACTGAACTCspecificACAAGCGTATCprotease 16TCCTATTTTATGAGAGAATACTGTGATAACAAAAAGTG443.H3079D12-3Pld3phospholipaseH3079D12Mm.6483Chromosome 7TTGGCCCACCCD3CCAAAGGGCCAAGATTATAAGTAAATAATTGTCTGTATAGCCTGTGCTT444.L0020E08-3ClqgcomplementL0020E08Mm.3453Chromosome 4CTGGGAACCACcomponent 1, qCTAATGGTATTsubcomponent,ATTCCTGTGGCgammaCATTTATCAATpolypeptideACCTTATGAGACTATT445.J0025G01-3Yarstyrosyl-tRNAJ0025G01Mm.22929Chromosome 4TCCTCTGGGGTsynthetaseAAATGAGCTTGACCTTGTGCAAATGGAGAGACCAAAAGCCTCTGATTTT446.L0832H09-3Mafbv-mafL0832H09Mm.233891Chromosome 2GCCGCAACGCmusculoaponeuAACAGAAATTroticGTTTTTAATTTfibrosarcomaCATGTAAAATAoncogeneAGGGATCAATTfamily, proteinTCAACCCB (avian)447.C0451C02-32700094L05RikRIKEN cDNAC0451C02Mm.25941No ChromosomeACTTTTGGGTC2700094L05locationTTTAGAACTGAgeneinfo availableGCCCACCTACTGAGTCTCAGTTTCTGTTGGTGTGACCT448.H3063A08-3LgmnlegumainH3063A08Mm.17185Chromosome 12TGCTTACTAAGAAGCCAGTTTGGGTGGGTAAAGCTCTCTGGAAGAAGGAACTTTGCTTCT449.K0629D05-3Evi2aecotropic viralK0629D05Mm.3266Chromosome 11TCCCAATGTGTintegration siteAGAATTCAACT2aATGTAACGCAATGGTACATTCTCACTGGATGAGATAGA450.G0111D11-3Cts1cathepsin LG0111D11Mm.930Chromosome 13CTTATGGACACTATGTCCAAAGGAATTCAGCTTAAAACTGACCAAACCCTTATTGAGTCA451.H3077D05-3Npc2Niemann PickH3077D05Mm.29454Chromosome 12GCCATATGATGtype C2AACAGAATTTCAAGAATGCTGTTTTATGCCTTTTAACCTCCAAAGCAGT452.G0104C04-3Dab2disabledG0104C04Mm.288252Chromosome 15TCATTTTCCTGhomolog 2TCTAGGCTAAA(Drosophila)GCTAAACTTAAACTATGGCTTTACGTAAATTAAGCTCC453.L0502D10-3PlalaphospholipaseL0502D10Mm.24223Chromosome 16CAACATCTAACA1 member AGCTTTACATAAATGCCCTTTTAGCTTCTCTATTTCGACACAACTGTGAT454.H3126B08-3Pla2g7phospholipaseH3126B08Mm.9277Chromosome 17TTACCCAAATAA2, group VIIAGCATTTTTTA(platelet-AATATACCCTGactivating factorTACTGTAGGATacetylhydrolase,AGTGATGAACplasma)GCCTAG455.J0034A07-3CregcellularJ0034A07Mm.459Chromosome 1ATAAGCCGTATrepressor ofCTGGGTCTTGGEIA-stimulatedACTACTTTGGTgenesGGACCTAAAGTAGTGACACCTGAAGAA456.H3114B07-3Slcl2a4solute carrierH3114B07Mm.4190Chromosome 8AAGTGGAATGfamily 12,GAGCCGGCCAmember 4AGCTGAGCCTGACTTTTTTCAATAAAACATTGTGTACTTC457.K0339H12-3Thbs1thrombospondinK0339H12Mm.4159Chromosome 2CTTAAAACTAC1TGTTGTGTCTAAAAAGTCGGTGTTGTACATAGCATAAAAATCCTTTGCC458.H3028C09-3AdkadenosineH3028C09Mm.19352Chromosome 14CAGCTGCCTAAkinaseCCCGCAACATTTGCATTATGTTCAGACTGTAACCTGCTTACTGATGGTA459.L0277B06-3PsapprosaposinL0277B06Mm.233010Chromosome 10CTGTGGTACCAAGGAGTTATTTTGGATGATTAGAAGCACAGAATGATCAGGCCTTTAGAG460.H3013F05-3Sdc1syndecan 1H3013F05Mm.2580Chromosome MultipleTTGTTTTTGTTTMappingsTTAACCTAGAAGAACCAAATCTGGACGCCAAAACGTAGGCTTAGTTTG461.H3084A06-3SpinspindlinH3084A06Mm.42193Chromosome 13TGCCTGAAAACACTTAACACTGATTGTCTAAGAGATGAAAGTCCTCCAAAGATGACACAG462.H3077F04-3Osbpl8oxysterolH3077F04Mm.134712Chromosome 10ACTTCAGTTAAbinding protein-TGGGTTTATAAlike 8AGTCAAGCACTGGCATTGGTCAGTTTTGTATGATAGGA463.K0324A06-3Itgal 1integrin, alphaK0324A06Mm.34883Chromosome 9TCCCCTATGCG11GTACGACCTTTACTGTCAGAAATATATTTAAGAAAATGTTCTAAACGGT464.C0115E05-32010110K16RikRIKEN cDNAC0115E05Mm.9953Chromosome 9GATCCAGCCTT2010110K16CTATGAAGAATgeneGCAAACTGGAGTATCTCAAGGAAAGGGAAGAATTCAGA465.C0668G11-3Fabp5fatty acidC0668G11Mm.741Chromosome MultipleCATGACTGTTGbinding proteinMappingsAGTTCTCTTTA5, epidermalTCACAAACACTTTACATGGACCTTCATGTCAAACTTGG466.L0030A03-3Alox5aparachidonate 5-L0030A03Mm.19844Chromosome 5CTTGTAATCAGlipoxygenaseACACGTGTTTTactivatingCCTAAAATAAAproteinGGGTATAGACAAAATTTAAGCCCATGG467.H3009E1 1-3Socs3suppressor ofH3009E11Mm.3468Chromosome 11TGTCTGAAGATcytokineGCTTGAAAAACsignaling 3TCAACCAAATCCCAGTTCAACTCAGACTTTGCACATAT468.L0010B01-3AbcalATP-bindingL0010B01Mm.369Chromosome 4TACTCCCATTAcassette, sub-CTATTTGCTGGfamily ATAATAGTGTAA(ABC1),CGCCACAGTAAmember 1TACTGTTCTGATTCAA469.G0116C07-3Ctsbcathepsin BG0116C07Mm.22753Chromosome 14CAGCCGATGCTTTTTCAATAGGATTTTTATGCTTTGTGTACCTCAACCAAGTATGAAGAG470.K0426E09-3Eps8epidermalK0426E09Mm.2012Chromosome 6GGGACACTTAAgrowth factorTTTACATGTACreceptorTTTAACCCCATpathwayGAAAGAGTCTsubstrate 8AGATAGAGAGAAGACAC471.H3102F08-3AsahIN-H3102F08Mm.22547Chromosome 8GCCTGCCAGTAacylsphingosineACCCCAGGAAamidohydrolaseGAGTCTAGCTT1CAAAAACCCACAAACTCATTATTTTTAA472.L0825G08-3Dcamk11double cortinL0825G08Mm.39298Chromosome 3AATCTAGATGTandTAGAAATCAATcalcium/calmodGTGTATGATGTulin-dependentATTGTATTTAGprotein kinase-ACCATACCCGTlike 1GACCG473.K0306B10-3Fgf7fibroblastK0306B10Mm.57177Chromosome 2ACGATGAGCAgrowth factor 7GTGTTTGAAAGCTTTCCAGTGAGAACTATAATCCGGAAAAATGAATGTTT474.H3127F04-3Chst11carbohydrateH3127F04Mm.41333Chromosome 10GATGCGTGAAsulfotransferaseATGTTCCTCCA11GGAAAAGCCATTCAAGCCTGATTATTTTTCTAAGTAACT475.L0208A08-31200013B22RikRIKEN cDNAL0208A08Mm.100666Chromosome 1CATCTTAGATC1200013B22TCAGAGACTTGgeneAACCTTGAAGCTGTTCCTAGTACCCAGATGTGGATGGA476.H3026G09-3Col2a1procollagen,H3026G09Mm.2423Chromosome 15CGTGTCCTACAtype 11, alpha 1CAATGGTGCTATTCTGTGTCAAACACCTCTGTATTTTTTAAAACATCAA477C0218D02-3Madh1MAD homologC0218D02Mm.15185Chromosome 8AAGGAGCCAC1 (Drosophila)GATAATACTTGACCTCTGTGACCAACTATTGGATTGAGAAACTGACAAGC478.J1031F04-3Dfna5hdeafness,J1031F04Mm.20458No ChromosomeGTTTATAGGTAautosomallocationGACCTAAGAGdominant 5info availableATAAAACTGCAhomologGGGTATCACAT(human)TAACGTTGGTTAAAAGA479.L0276A08-3Rail4retinoic acidL0276A08Mm.26786Chromosome 15AAACTTGAGACinduced 14ATTTTGTAGGACGCCTGACAAAGCGTAGCCTTTTTCTTGTGTCAGGATG480.C0508H08-3Sptlc2serineC0508H08Mm.565Chromosome 12CTCATACCAAApalmitoyltransfGAAATACTTGAerase, longCACTGCTTTGAchain baseAGGAGATAGAsubunit 2TGAAGTTGGGGATCTGC481.J0042D09-3C78076ESTs C78076J0042D09Mm.290404Chromosome 12AAATCCAGCCTTTAAAAGCTCAGTTTCTTCCTCTAAGTGAATGTCATTACTCTGGTATAC482.J0013B06-3Akrlb8aldo-ketoJ0013B06Mm.5378Chromosome 6ACCAGGAACTCreductaseTGGTAACATTTfamily 1,GAGGGCATGCmember B8AGATAAAATAATAAAGAATGAGAACATT483.H3158D11-3Mmp2matrixH3158D11Mm.29564Chromosome 8TCAACATCTATmetalloproteinaseGACCTTTTTAT2GGTTTCAGCACTCTCAGAGTTAATAGAGACTGGCTTAG484.H3001D04-3Hist2h3c2histone 2, H3c2H3001D04Mm.261624Chromosome 13GACCGAGAGCCACCACAAGGCCAAGGGAAAATAAGACCAGCCGTTCACTCACCCGAAAAG485.C0664G04-3PpicappeptidylprolylC0664G04Mm.3152Chromosome 11TTCTACCTCACisomerase C-TAACTCCACTGassociatedACATGGTGTAAproteinATGGTACATCTCAGTGGTGGTGATGCA486.H3091E10-3Nupr1nuclear proteinH3091E10Mm.18742Chromosome 7TTGGAGAAATT1AGGAGTTGTAAGCAGGACCTAGGCCTGCTTGATTCTTTCCCACCTAAGT487.X98792.1Ptgs2prostaglandin-X98792Mm.3137Chromosome 1TTATTGAAAAGendoperoxideTTTGAAGTTAGsynthase 2AACTTAGGCTGTTGGAATTTACGCATAAAGCAGACTGC488.L0908B12-3Ptpn1protein tyrosineL0908B12Mm.227260Chromosome 2CACCATTTCCAphosphatase,ACTTGCTGTCTnon-receptorCACTAATGGGTtype 1CTGCATTAGTTGCAACAATAAATGTTT489.H3081D02-3BokBcl-2-relatedH3081D02Mm.3295Chromosome 1AACAAGAGATovarian killerCCTGTGGATGAproteinGGGGGTCTGTATAAGTTATACTCCAATAAAGCTTTACCT490.C0127E12-3Cln5ceroid-C0127E12Mm.38783No ChromosomeTTTTGACCAGTlipofuscinosis,locationTGAACCCATTTneuronal 5info availableTGTTTTCCTAGCGAACACTAGCATAATATTGGAAAAGC491.K0310G10-3Col5a2procollagen,K0310G10Mm.257899Chromosome 1GTGAGGATTGGtype V, alpha 2AATTAGAACATTCATAAGAAAATATGACCCAACATTTCTTAGCATGACC492.H3023H09-3Ftl 1ferritin lightH3023H09Mm.7500Chromosome 7CGCCCTGGAGCchain 1CTCTGTCAAGTCTTGGACCAAGTAAAAATAAAGCTTTTTGAGACAGCAA493.G0104B11-3Slc7a7solute carrierG0104B11Mm.142455Chromosome 14AAGATGGAGAfamily 7GTTGTCCAAAC(cationic aminoAAGATCCCAAacid transporter,GTCTAAATAGAy+ system),GCAAGGGATTCmember 7TGAGGTG494.C0123F05-3B4galt5UDP-C0123F05Mm.200886Chromosome 2GTTTTAAAAGGGal:betaGlcNAcTGCCAGGGGTAbeta 1,4-CATTTTTGCACgalactosyltrans-TGAAACCTAAAferase,GATGTTTTAAApolypeptide 5AACAC495.H3082D01-31801105C04RikRIKEN cDNAH3082D01Mm.25311Chromosome 15TCTGAGGTATT1810015C04AAAATATCTAGgeneACTGAATTTTGCCAAATGTAAGAGGGAGAAAGTTCCTG496.C0121E07-3AW539579ESTC0121E07Mm.282049No ChromosomeAAGTATTGCTAAW539579locationGACTGAAACCinfo availableACTTGAACTTCTCAGAGAGGTTAGACTGACAGAAGGTGT497H3153H08-3Hs6st2heparan sulfateH3153H08Mm.41264Chromosome XACATTTTTGTC6-O-ATCATCATGTAsulfotransferaseAATCCCACGAT2TTCAAACTGTAAACATCTGTTCAGTGG498.J0238C08-34930579A11RikRIKEN cDNAJ0238C08Mm.24584Chromosome 11CTGGGGAAATT4930579A11GATCTTTAAATgeneTTTGAAACAGTATAAGGAAAATCTGGTTGGTGTCTCAC499L0942B10-3Msr2macrophageL0942B10Mm.45173Chromosome 3AGGACTCAAAscavengerACTATATTAATreceptor 2CTGCTCTGAGATAATGTTCCAAAAGCTCCAAAGAAAGCC500.J0915B05-3Cdcalcell divisionJ0915B05Mm.151315Chromosome 1GCTCCAACATGcycle associatedCCATGTATTGT1ATAGACTTTTACTACAATTCAAATAACGTGTACAGCTT501.H3058B09-3Lypla3lysophospholipaseH3058B09Mm.25492Chromosome 8CAGCTGAATGG3GTTTTGGTTTGCAGGAAAACAGTCCAGAGCTTTGAAAAGGCTCCTAAGA502.C0197E01-3D630023B12hypotheticalC0197E01Mm.227732Chromosome 3TGTTTTTATTGproteinTGTTTGGTGGAD630023B12GAAGAATAATACACTTCTTGCCTAAATCCAGAAGCCCC503.J0802G04-30610011104RikRIKEN cDNAJ0802004Mm.27061Chromosome 6TCCAGTTCCCG0610011104AAGAAGCTGAgeneTAGGAATTGCCCTTGTGCATATACTACACAAGCATGCTA504.H3039E08-3Sh3d3SH3 domainH3039E08Mm.4165Chromosome 19CATAAAGACATprotein 3AGTGGAGGTTCTGTTTACTCAGCCGAATGTGGAGCTGAACCAGCAGAAT505.L0210A08-3B130023014RikRIKEN cDNAL0210A08Mm.27098Chromosome 5GGATTCGGCTCB130023014GATGAATGAAgeneGCACTTTATGGACTGCGGGGATCAGTTACTGCCACACCC506.H3114C10-3PpgbprotectiveH3114C10Mm.7046Chromosome 2TGCTTTTACCAprotein for beta-TGTTCTCGAGGgalactosidaseTTCCTGAACAAAGAGCCTTACTGATAGTTCCGCTGCAA507.C0322A01-32810441C07RikRIKEN cDNAC0322A01Mm.29329Chromosome 4TGAAGCAAAA2810441C07AACATAAAACgeneCTCACCACTGCCTGCTGAACCTAGAACCTTTTGTTGGGGC508.L0256F11-3AdfpadiposeL0256F11Mm.381Chromosome 4GAATCCTTAGAdifferentiationTGAAGTTATGGrelated proteinATTACTTTGTTAACAACACGCCTCTCAACTGGCTGGTA509.L0939H06-3Mgat5mannosideL0939H06Mm.38399Chromosome 1GATATTAGTAGacetylglucosamiTATATCATAAAnyltransferase 5ACTTGAGAAATAAAGATGCGCTCACCCCCTATCTGTTG510.C0503B05-3Dcanikl1double cortinC0503B05Mm.39298Chromosome 3TGTGATAAAGTandTGTGACATACGcalcium/calmodTATTAGTTGGCulin-dependentACATATTTAAGprotein kinase-CTCCAAATCAGlike 1TTTGC511.H3136H11-3Map4k5mitogen-H3136H11Mm.260244Chromosome 12TAAAAGTTAAAactivatedGTAAGCGAAGprotein kinaseAAAGGAAGCTkinase kinaseGTATCTACACTkinase 5GCTTTCCAGTTTAATCAG512.K0349A04-3Fnlfibronectin 1K0349A04Mm.193099Chromosome 1GGAGATTTTTCTCTTCAGGGTGTCTACATACCTTACACACACTTGTGTCTTAATAAGCAA513.C0177C04-3Ctszcathepsin ZC0177C04Mm.156919Chromosome 2AATCCATGGGAGGGGGGAACAAGTCCAGACTGCTTAAGAAATGAGTAAAATATCTGGCTT514.C0668D08-3GrngranulinC0668D08Mm.1568Chromosome 11AATGTGGAGTGTGGAGAAGGGCATTTCTGCCATGATAACCAGACCTGTTGTAAAGACAGT515.C0106D12-3Anxalannexin A1C0106D12Mm.14860Chromosome 19TGACATGAATGATTTTACCAGAAGAAGTATGGAATCTCTCTTTGCCAAGC516.H3078E09-3HexbhexosaminidaseH3078E09Mm.27816Chromosome 13ACTGGATACTGBTAACTATGAGAATAAAATATAGAAGTGACAGACGTCTACAGCATTCCAG517.L0033F05-32810442122RikRIKEN cDNAL0033F05Mm.275696Chromosome 10ATACAAGCAA2810442122GCTGTTAAAGAgeneTCTTGGATCCCATTCTATAGTGTGTATACCTAAATCAAC518.K0144G04-3Ifi203interferonK0144G04Mm.245007Chromosome 1 notAGCATCAACTGactivated geneplacedTCCTGTCAAGC203ACAAAAAATGAAGAAGAAAATAATTACCCAAAAGATGG519.H3144E05-34933426M11RikRIKEN cDNAH3144E05Mm.27112Chromosome 12CCTCTGTTCTG4933426M11AGGAACATTCTgeneAGCATAGAAAATGGAATATGCTGCAAACATTTCTAGAT520.K0336D02-3Ifi16interferon,K0336D02Mm.212870Chromosome 1GTGTAGAAGCCgamma-TATTGAAATATinducibleCAGTCCTATAAprotein 16AGACCATCTCTTAATTCTAGGAAATGG521.H3004B12-3HpnhepsinH3004B12Mm.19182Chromosome 7CTGATCCCGCCTCATCTCGCTGCTCCGTGCTGCCCTAGCATCCAAAGTCAAAGTTGGTTT522.K0617G07-3Atp6vlb2ATPase, H+K0617G07Mm.10727Chromosome 8TGTAGAAAATGtransporting,TGGCCTCTCGTV1 subunit B,TATAAATGAAAisofonn 2ATAAATGTTTAATTTAATGGGAGTTTC523.L0849B10-3PltpphospholipidL0849B10Mm.6105Chromosome 2GGTGCCACAGtransfer proteinAGAAGAGCCCAGTTGGAAGCTATACCCGATTTAATTCCAGAATTAGTCAA524.L0019H03-3Fnlfibronectin 1L0019H03Mm.193099Chromosome 1CAGTGTTGTTTAAGAGAATCAAAAGTTCTTATGGTTTGGTCTGGGATCAATAGGGAAACA525.J0099E12-3Slc6a6solute carrierJ0099E12Mm.200518Chromosome 6ATAACTATATAfamily 6TACTTAGAGTC(neurotransmitterTGTCATACACTtransporter,TTGCCACTTGAtaurine),ATTGGTCTTGCmember 6CAGCA526.J0023G04-3BC004044cDNA sequenceJ0023G04Mm.6419Chromosome 5CCTTGGGACATBC004044TTTTGTGGAGTAGTTTGCAGTGAGATAACAGTGCAATAAAGATACAGCA527.C0913D04-34933433D23RikRIKEN cDNAC0913D04Mm.46067Chromosome 14TCTATACCTGG4933433D23ATAAAAAGAAgeneACCTACACTTCACTGTAAAACTTCATGTTTCAAGGCAAG528.H3020C02-3Mt1metallothioneinH3020C02Mm.192991Chromosome 8CCTGTTTACTA1AACCCCCGTTTTCTACCGAGTACGTGAATAATAAAAGCCTGTTTGAGTC529.C0217B11-3Sema4dsema domain,C0217B11Mm.33903Chromosome 13ACCGTGTAGACimmunoglobulinACTCATATTTTdomain (Ig),GCATGACATGAtransmembraneTCTACCATTCGdomain (TM)GTGTAAACATTand shortTGTGTcytoplasmicdomain,(semaphorin)4D530.C0917E01-3Bhlhb2basic helix-C0917E01Mm.2436Chromosome 6GCCAAAGGAAloop-helixAATGTTTCAGAdomainTGTCTATTGTcontaining,ATAATTACTTGclass B2ATCTACCCAGTGAGGAA531.H3132B12-5DeafideformedH3132B12Mm.28392Chromosome 7TCCAGAAGCTGepidermalCATTGCCAACAautoregulatoiyTCACACCCCAAfactor 1AATTGTCCTGA(Drosophila)CATCGCTGCCCGCATT532.L0270C04-3MpplmembraneL0270C04Mm.2814Chromosome XAAGGACTCTGAprotein,GGCCATCCGTApalmitoylatedGTCAGTATGCTCATTACTTTGACCTCTCTTTGGTGAAT533.J0709H10-3transcribedJ0709H10Mm.296913Chromosome 13ATCTCCCAAGGsequence withCAAAGAACTGmoderateAAACTCAGAGsimilarity toCTGTCTGGATTproteinGAAGAAATGTpir:A38712GTGTTGTT(H. sapiens)A38712fibrillarin[validated]-human534.C0166A10-3Car2carbonicC0166A10Mm.1186Chromosome 3ATGAAGGTAGanhydrase 2GATAATTAATTACAAGTCCACATCATGAGACAAACTGAAGTAACTTAGGC535.L0511A03-3BM122519ESTsL0511A03Mm.296074Chromosome 1GGTGTAGCCATBM122519ACAATACACAAATACAATAGATATTCTCTCTACAATCTTTATGGTGTGG536.H3029F09-3Atp6v1e1ATPase, H+H3029F09Mm.29045Chromosome 6GGAGAAGCAGtransporting,ATTATCTGTGTVI subunit EGGCTTCCTCTTisoform 1TCTGTTCTAATACTGGTAATCAGTGGAC537.J0716H11-3Kdtlkidney cell lineJ0716H11Mm.1314Chromosome 6GTGAACACCAderivedGAATTTAATTTtranscript 1CCATACTTGTACAGGTAGGACTATTCTTCAGCTCTCTAC538.C0102C01-3Acp5acidC0102C01Mm.46354Chromosome 9GGCTTCACACAphosphatase 5,TGTGGAGATAAtartrate resistantGCCCCAAAGAAATGACCATCATATATGTGGAAGCCTCT539.C0641C07-3Pdgfbplatelet derivedC0641C07Mm.144089Chromosome 15GTTTGTAAAGTgrowth factor,TGGTGATTATAB polypeptideTTTTTTGGGGGCTTTCTTT-TTATTTTTTAAATGTAAAG540.C0147C09-3Tct7tetratricopeptideC0147C09Mm.77396Chromosome 17ATGGAATTCTGrepeat domainTTAGAGTAAAA7AAGAGAAAAGCAGATACTATTGGCTGGCCTTGGAGGTC541.K0301G02-394300025M21RikRIKEN cDNAK0301G02Mm.87452Chromosome 1AATAGTGCTGA9430025M21ATTTGTCTAAAgeneCAGAATTGAGAGGTCATAGAAATCCTTAACAGGGTAAC542.H3002D05-3TpbpbtrophoblastH3022E05Mm.297991Chromosome 13TATGAAGATTTspecific proteinGGGAAAGAACbetaAGCTATCTGACACCTGGAAGGCTCAGCCAGAGTAACAGT543.H3007C09Sh3bgr13SH3 domainH3007C09Mm.22240Chromosome 4GAGGCAACATTbindingCCTTATTCACCglutamic acid-AACTAGTCTCArich protein-likeAAAGATTGTCT3TAAGCCCTGACGATGG544.L0820G02-3Igsf4immunoglobulinL0820G02Mm.248549Chromosome 9TAATGAAGGATsuperfamily,GTATAATTGATmember 4GCCAAATAAGCTTGTTCTTTAGTCACGATGACGTCTTG545.C0120H11-34933433D23RikRIKEN cDNAC0120H11Mm.46067Chromosome 14CAGTTTGCGAA4933433D23GTAGAATTTTGgeneTTTCTAAAAGTAAAAGCTAAGTTGAAGTCCTCACGAG546.J1016E08-31810046J19RikRIKEN cDNAJ1016E08Mm.259614Chromosome 11TAGAAAAGAT1810046J19CACCAACAGCCgeneGGCCTCCCTGTGTCATCCTGTGACTAAGAAATGATTCTT547.L0822D10-3Prkcbprotein kinaseL0822D10Mm.4182Chromosome 7TATCTAAGAGCC, betaCAAGTCTATGGCATTAGCTGTGAGAAGTAGTTACCACTGTAATTCACCT548.H3050H09-3Ppp2r5cproteinH3050H09Mm.36389Chromosome 12AAATTATCACTphosphatase 2,TGATACGGAregulatoryGGAACATGACTsubunit BAGGCACATTTT(B56), gammaATGAATACTCCisoformAAATCC549.J0442H09-3Mus musculusJ0442H09Mm.11982Chromosome 10AACTATGGTGhypotheticalGTATATTTTTGLOC237436AACACAGGTTA(LOC237436),ACTGTGGAGGTmRNATATCTGCTAATAGCAA550.H3141E06-3Sra1steroid receptorH3151E06Mm.29058Chromosome 18ACCTCTGGAACRNA activatorAGGCATTGGA1GGACTGCCATGGTCACACAAAGAAACAGAACTTTTACAT551.C0171H06-3Adss2adenylosuccinateC0170H06Mm.132946Chromosome 1CCAGTATACCTsynthetase 2,ACAAAATGACnon muscleCCACAAGTAACCCGCATGAGTCCAAGTTGTCAGCCATAT552.K0344C08-3Emp1epithelialK0344C08Mm.30024Chromosome 6GTAAAGGGACmembraneCATTACTAAGTprotein 1GTATTTCTCTAGCATATTATGTTTAAGGGACTGTTCAAG553.J0907F03-3NplN-J0907F03Mm.24887Chromosome 1CTCTAAGTCATacetylneuraminateTCATTTTGTAApyruvatelyaseAATTATTATAGAGAAATCTCTACTTATACAGATGCAAT554.J1008C10-3Ptpn1protein tyrosineJ1008C10Mm.2668Chromosome 2TCTAATCTCAGphosphatase,GGCCTTAACCTnon-receptorGTTCAGGAGAtype 1AGTAGAGGAAATGCCAAATACTCTTCTT555.K0103F09-32500002K03RikK0103F09Mm.29181Chromosome 6ATTCAGATCAG2500002K03GAAAGGTTGAgeneAATGGTCTTCGTTACCAGGAGGTCTACATTTATTAATTT556.C0837H01-3Adam9a disinegrinC0837H01Mm.28908Chromosome 8CAGTTATGGGCandTTCCATTTTCAmetalloproteinaseAATATCTTTTCdomain 9AACTGTAATGA(meltrinCTATGACAGGAgamma)ACTGA557.J0207H07-3Runx2runt relatedJ0207H07Mm.4509Chromosome 17GCTTTCTATGCtranscriptionACGTATTGTACfactor 2AAATTGTGCTTTGTGCCACAGGTCATGATCGTGGATGA558.J0246C10-3Tpd52tumor proteinJ0246C10Mm.2777Chromosome MultipleTGGCTAGATTTD52MappingsAATTGAGGATAAGGTTTCTGCAAACCAGAATTGAAAAGCCACAGTGTCG559.H3158E12-3BC003324cDNA sequenceH3158E12Mm.29656Chromosome 5AGAGGACCATTBC003324ATGAAGAAGCTGTTCTCTTTCCGGTCAGGGAAGCATACCTAGACTGAAA560.H3094A04-3Dnajc3DnaJ (Hsp40)H3094A04Mm.12616Chromosome 14AGAAAAGAAAhomolog,AAGCAGAGAsubfamily C,AAAAGTTCATTmember 3GACATAGCAGCTGCTAAAGAAGTCCTCTC561.L0231F01-3EvlEna-vasodilatorL0231F01Mm.2144Chromosome 12ATATTTGCTTAstimulatedTTTAAGCGTACphosphoproteinGTTCCTTTGGTTTATAGAGAACACCCCCAAATCACCTG562.K0512E10-3Myo5amyosin VaK0512E10Mm.222258Chromosome 9GACTCTCCCAACTTACAGACTTTTATCAGATATGGAGAAGATAATGTTAAGAGACTTCACA563.K0608H09-3Ptprcprotein tyrosineK0608H09Mm.143846Chromosome 1TAAAATCCCATphosphatase,TGAAAGTGGAreceptor type, CCTCAGTTGTAAGAATAACAATGTGTACCATTCTGGAATG564.L0842E04-3Prkcbprotein kinaseL0842E04Mm.4182Chromosome 7CCAATGAACCGC, betaACAGTGTCAAAACTTAACTGTGTCCAATACCAAAATGCTTCAGTATTTG565.H3121G01-3BG073361ESTsH3121G01Mm.182649Chromosome 11TCAAATCAGTTBG073361TCAACTTTCATAAAATGGATTCTTTAATGGATGGAGACTTACTCGTCGG566.C0947F04-35830411K21RikRIKEN cDNAC0947F04Mm.160141Chromosome 2CTATACACAAG5830411K21ATATGCTAGGAgeneGATGTGAAAGATAATGGAGACTTTCCAGTAAGCACTTT567.H3009D03-5Plac8placenta-H3009D03Mm.34609Chromosome 5CTGAGATTTTTspecific 8CAAATCTTTGGCAACTGAGATGGGATGGATCCATTTAATTAGAGAACGG568.H3132E07-3LxnlatexinH3132E07Mm.2632Chromosome 3AAATGTCTTTCCAACAGTAATGGTACTATGTCTATCCCCTAATAAAACTTCACTTCAGCC569.H3054C01-3Nr2e3nuclear receptorH3054C01Mm.9652Chromosome XTGAACATTCACsubfamily 2,AGGATTTCTAAgroup E,CTATACTGATAmember 3TAAACCCAGTGTTTTCTGGACTCAGGG570.H3013h03-3Manlamannosidase 1,H3013H03Mm.117294Chromosome 10CAACAAAGTTGalphaATTTACATGTATAATCCACACCCTTAAAGATGAACAGTTAGAGTAGCAC571.J0058F02-3ankprogressiveJ0058F02Mm.142714Chromsome 15TGGACACAGTTankylosisCACTAAATTCCTGATTTAGTCAAAGTAACTAGACTGAAAGAACCTAAAC572.L0829D10-3Sncasynuclein, alphaL0829D10Mm.17484Chromosome 6TTGTTGTGGCTTCACACTTAAATTGTTAGAAGAAACTTAAAACACCTAAGTGACTACCAC573.H3037H02-31110018O12RikRIKEN cDNAH3037H02Mm.28252Chromosome 18TGAACACATCA1110018O12AGTATTCTGGAgeneGCTTCAGCGGCAGTTAAATGCCAGTGACGAACATGGAA574.K0105H12-3Cdk6cyclin-K0105H12Mm.88747Chromsome 5AAGGTCCAAAdependentATACAGACATTkinase 6TTTGCTAGGGCCTAGAAATCGACCATAAAACACACTGCA575.C0105D10-3C0105D10-3C0105D10No ChromosomeGACTGAAATGNIA MouselocationAAAGTTCCACTE7.5info availableAACGGTATTTGExtraembryonicCTCTAGTGATAPortion cDNATGTGGACATTGLibrary MusTGATATmusculuscDNA cloneC0105D10 3′,MRNAsequence576.L0229E05-3PrkxputativeL0229E05Mm.106185Chromosome XTCAAATAAAAserine/threonineAACCCTTAATCkinaseAGGCTGTAAATCAAATGACACTATGCGATGTCACTACAG577.L0931H07-3ESTsL0931H07Mm.221935Chromosome 1GCACTATAAATBQ557106TTCATCTTTTGAAGGTTGTTGACTACAAGGGTACAAAAATGATACAGGC578.K0138B11-3Trim25tripartite motifK0138B11Mm.4973Chromosome 11CTTGCATGAGTprotein 25GCGTGTTTAAGTTCTCGGAATTTCCTGAGAGGATGGAGTGCCATTGTTA579.H3019H03-3Lass6longevityH3019H03Mm.265620Chromosome 2AGTGTTAGCTGassuranceCAAAGCTACAhomolog 6 (S.AAGCTCTGGAcerevisiae)TGGTTACATTATGATTCTGGAACGTTCG580.J0051F04-3Ifi30interferonJ0051F04Mm.30241Chromosome 8TCCAGACTTCTgammaCAGAGACAAGinducibleGATCTTGCCTTprotein 30ATTTTCAAATGGTGCTAAATTTAAATTC581.H3106G04-3CacnaldcalciumH3106G04Mm.9772Chromosome 14AGTGACTTCCAchannel,CCTTTTAATGTvoltage-CATTAAAAGCAdependent, LGGAGCTTAAACtype, alpha 1DTAAAAGCAGCsubunitATTCCA582.L0701D10-3ArhgdibRho, GDPL0701D10Mm.2241Chromosome 6ACATACATTTCdissociationATCACCAATATinhibitor (GDI)GTTTTATCTTAbetaCCCCATCTCTCAGAGTGTTCCCTGCAA583.H3137A02-3Mus MusculusH3137A02Mm.21657Chromosome 4TTTTTTGTATT10 days neonateATTGTGTTTTGcerebellumTGCTACTGTAGcDNA RIKENTTTTGGTGTGGfull-lengthCACTATTATAAenrichedTTAAAlibraryclone:B930053B19product:unknown EST, fullinsert sequence.584.L0043D10-3A5310090O1RikRIKEN cDNAL0043D10Mm.40298Chromosome 15CTTAGGGAGACA530090O15TACTAACATGGgeneAGAGAATGCCGTGTATACCTCACGTACTGTGTGCTTTA585.H3087D06-3EtfleukaryoticH3087D06Mm.3845Chromosome 18CATACATAGAAtranslationGCAAAATACTTterminationTAACTGCTGTAfactor 1AACCTTCAAAAGTTAGTAGACGTGAGG586.C0827E01-3Mus musculusC0827E01Mm.45759Chromosome 10ACTTCCTGCAA15 days embryoTACATCCCAGThead cDNAAGGTACACCTARIKEN full-GTTTACAATTTlength enrichedAAACTAGTTTGlibrary,TGAAAclone:D930031H08product:unknownEST, fullinsert sequence.587.H3053E01-3B130024B19RikRIKEN cDNAH3053E01Mm.34557Chromosome 10GGAGGCACATB130024B19AATTCCAAGCAgeneATACAGGCTGTTAAAATATAAATAATGGGAACTGTGATT588.K0117C08-3BM222243ESTsK0117C08Mm.221706Chromosome 1AAGCGTTAGGBM222243AAGGAAATTTCCTGGAAGGATAGGTTGTCTTCCTAGCAGCCTCGTCAATA589.H3056D11-3PtgfmprostaglandinH3056D11Mm.24807Chromosome 3TTTTTTAACTTF2 receptorCACTCATGACAnegativeACAGAGGAAGregulatorAAAGGAATTGAGGTTTAGGTAAGTTCTC590.C0228C02-32510004L01RikRIKEN cDNAC0228C02Mm.24045Chromosome 12AGGCATATCTC2510004L01ATAGAGCCTTAgeneAGTTAGAATCTTACTCTTATGGAAGGAGTTATTTCCTA591.H3144F09-3Rab711RAB7, memberH3144F09Mm.34027Chromosome 1GATCACCTCATRAS oncogeneTCCTCGACTGTfamily-like 1GAGATGAGTTTATGAAAAGAATTAAAAGTGAGCACTTG592.H3052B06-3Abcb1bATP-bindingH3052B06Mm.6404Chromosome 5TAAAGGTAACTcassette, sub-CCATCAAGATGfamily BAGAAGCCTTCC(MDR/TAP),GAGACTTTGTAmember 1BATTAAATGAACCAAAA593.L0273B08-3TgifTG interactingL0273B08Mm.8155Chromosome 17GGCCAGGTATAfactorTGTGTACCAGTGCTCTTCAAAGGGAGAACCATTAAAACCAACATGGAAT594.K0406A08-3Siat4csialytransferaseK0406A08Mm.2793Chromosome 9CCAAGAGATTA4C (beta-TTTAACATTTTgalactosideATTTAATTAAGalpha-2,3-GGGTAGGAAAsialytransferase)ATGAATGGGCTGGTCCC595.AF075136.1Sap30sin3 associatedAF075136Mm.118Chromosome 8AGTGAACGAApolypeptideAAAGACACCTTAACATGTTTCATCTACTCAGTGAGGAACGACAAGAACAA596.K0644H12-3Prkchprotein kinaseK0644H12Mm.8040Chromosome 12GATATTTATTGC, etaAGTGTCAAATAAAAAGGTGCCATAATCTTCAGTAGCGTACACAGTAGAG597.H3108A04-3CluclusterinH3108A04Mm.200608Chromosome 14GTGTTACCAGAAGAAGTCTCTAAGGATAACCCTAAGTTTATGGACACAGTGGCGGAGAAG598.H3020F06-3Snx10sorting nexin 10H3020G06Mm.29101Chromosome 6TGTCTTTATTTTAATGCCAAAAGGAAGTGATTATGCAGCTGTGTGTAGAGTTTCAGAGCA599.L0066C05-3Uxs1UDP-L0066C05Mm.201248Chromosome 1AGAACAAACTglucuronateGGAATTTTATTdecarboxylase 1CTGAAGCTTGCTTTAAAGACACTGATGTGCCTAAACGCT600.L0025F08-3Rgs19regulator of G-L0025F08Mm.20156Chromosome 2TATGGTCTTTCproteinAGTCACAGTGTsignaling 19AGTCACAGTGTCATCTTAATCTTACTGATCCAATAAAAC601.H3076F06-3Siat4asialytransferaseH3076F06Mm.248334Chromosome 15ATCCTCCTGAT4A (beta-TGGTCTGAATGgalactosideCATTTCCAATGalpha-2, 3-ATGTCAGGGAsialytransferase)TCAGCC602.C0354G01-3Mus musculus,C0354G01Mm.259704Chromosome 13TAAGCCCTGTCSimilar to IQTTCTGGGAAATmotifATCAGTTTTAAcontainingAGAGAACTTTTGTPaseGTGCAATTCCAactivatingAATGAprotein 2, cloneIMAGE:3596508, mRNA,partial cds603.C0191H09-3Atp6vla1ATPase, H+C0191H09Mm.29771No ChromosomeGGAAGATTAATtransportinglocationTTTCCAGGGATV1 subunit A,info availableTGTATCAATCAisoform 1GGACCATTTTTGTGGGGCACTTGGGAC604.H3050G04-3Dpp7dipeptidyl-H3050G04Mm.21440Chromosome 2ATGTGATCTACpeptidase 7AGTGGTGTGACAACTTGCCTTGTATCTGATGGACTGTCCAGATTTATGG605.L0219A09-3GatmglycineL0219A09Mm.29975Chromsome 2AAACGAAGTGamidinotransferaseACTTTCCATGA(L-arginine:ATGCCTTTAACglycine amidino-ATTCTTGTGTCtransferase)AACATTTGGTACTAAAC606.J0821E02-3AU040950expressedJ0821E02Mm.17580Chromosome 13AATACTCATTAsequenceTGCTGTGTGGGAU040950AATTTCCTGATTACTAGAAGCTGACCTCTGCTATCCTG607.H3080a02-3Cbfbcore bindingH3080A02Mm.2018Chromsome 8GAATTATTATAfactor betaAACAATAATGTGTTACAGAAGCTGATGCTGACCTTGTGTTACTGAGCAC608.C0276B08-3Plscr1phospholipidC0276B08Mm.14627Chromosome 9TTCTTGAGGTTscramblase 1TAAGGACGACAACTTTATGGACCCTGAATGGAAACTGAGGAATCACAAG609.C0279E04-3Srd5a21steroid 5 alpha-C0279E04Mm.86611Chromosome 5GTCACATGCCAreductase 2-likeATAAAAACAGGAAACTCTGAAAATAATATGAATGTACAGTATCAGACCG610.K043D04-3PgdphosphogluconateK0434D04Mm.252080No ChromosomeCCCTATTGCAAdehydrogenaselocationATTGATTTGTTinfo availableTTCCCTTAACCCTGTTCCCTTTTAACCCCGGCTTTTTT611.C0174H01-3Ddx21DEAD (Asp-C0174H01Mm.25264Chromosome 10CATTGCATCGTGlu-Ala-Asp)TTTCCAACATAbox polypeptideCTTTTAGATTT21ACAAAGTAAAACCAACCATGGATCTGC612.H3085A07-3BG070224ESTsH3085A07Mm.173217Chromosome 17TTGAGAAATTABG070224AAAACAAATATCCAAAATCGACTTTTCCTCAAGGCTATGTGCTTCGTCC613.K0208E10-3MmabmethylmalonicK0208E10Mm.105182Chromosome 5ACGACTCTTGTaciduriaTAATGTGCGTT(cobalaminTCTCATGGAGTdeficiency) typeAATTTTCAGAGB homologCCTGAACTTGT(human)AGCAC614.H3006F10-3Cops2COP9H3006F10Mm.3596Chromosome 2GTTGGTGTGTC(constitutiveCTGAAAGGGAphotomorphogenic)TGGAGTTATGGhomolog,CAGAAGTGCTTsubunit 2TTGTGATCAAC(Arabidopisi-TGGTTTthaliana)615.C0108A10-3Nek6NIMA (never inC0108A10Mm.143818Chromosome 2CAGAAAACTCmitosis gene a)-AAGTCATGGACrelatedTATGCGAGTCAexpressedAGAATTAAAATkinase 6ACAACTGTATTATGTGC616.H3028H10-3Ppicpeptidylprolyl-H3028H10Mm.4587Chromosome MultipleAAATTTCTCATisomerase CMappingsTTAATTTTCCAGTCTCGATTGCAGTAACAAAGTCAACCACACAGTCAGA617.H3121E08-3Ralgdsral guanineH3121E08Mm.5236Chromosome 2GGAGGAAGACnucleotiedAACTGAACATTdissociationTGTATAAAACGstimulatorTAAAAGTTTACTGATTGGGGTGGGACA618.L0266H12-3Opaloptic atrophy 1L0266H12Mm.31402Chromosome 16CAGCAGCTTAChomologAAACACTGAA(human)GTTAGGCGACTAGAGAAAAACGTTAAAGAGGTATTAGAA619.K0635G02-32310046K10RikRIKEN cDNAK0635G02Mm.68134Chromosome 14GAGAAATGTTA2310046K10GTAAAATGGTAgeneAAAGGGAATCACGTGACATTCAGGGTAGGAAGAGCTTG620.L0704C05-32613018G18RikRIKEN cDNAL0704C05Mm.180776Chromosome 3TCAGGAAAAA2610318G18TGTCATAAGCCgeneATCTGGTAAGTTTTCTTAAAGGATGTTGTTAAGAAGTCC621.C0303D10-3UNKNOWNC0303D10Data not foundNo ChromosomeCAAAACAAATC0303D10locationACATATTATAAinfo availableAATAAAAGAAAAGGCGTGATAAATGGATGTGACAAAATT622.K0605C04-3BM240648ESTsK0605C04Mm.265969Chromosome 15GTAGGGAAAAMN240648TATGTCCATAGGTTTTAGGAAACACTTAGCCTTTAATATACTGGTTGTAG623.H3071G06-3BG069012ESTsH3071G06Mm.26430Chromsome 4GTATACAGATGBG069012GTAGTTAGAAATACTGGATGAACTGATCAGTTATTGTGTGTAGAAAGTG624.C0600A01-3Coro2acoronin, actinC0600A01Mm.171547Chromosome 4TTGTATCCCAAbinding proteinAGGGAAACGG2AGAATCAAGATACGGACCTATGCTTTTCATATGAAACCGT625.NM_007679.1CebpdCCAAT/enhancerNM_007679Mm.4639Chromosome 16TGCAGCTAAGGbindingTACATTTGTAGproteinAAAAGACATTT(C/EBP), dataCCGACAGACTTTTGTAGATAAGAGGAA626.H3048A01-3Kras2Kirsten ratH3048A01Mm.31530Chromosome 6GGCAATGGAAsarcomaAATGTTGAAAToncogene 2,CCATTCGTTTexpressedCCATGTTAGCTAAATTACTGTAAGATCC627.C0267D12-3Tpp2tripeptidyl-C0267D12Mm.28867Chromosome 1CCCCAAAGAApeptidase IIAACTGGAAAAATTGTTTTCCACTCCTGAAATTTCTTGGATGGGCCCCCTG628.J1012C06-3AU041997ESTsJ1012C06Mm.181004Chromosome 5CCAGACAGTGTAU041997ATTCTTCGGACAAATGGTGTGAAAGTGAAATAAGAATTCATAATGTAAC629.L0072f04-3Vav2Vav2 oncongeneL0072F04Mm.179011Chromosome 2AGCAAAAGTATGTATATTTTAGCTTGTCATGAAATGTCAACGAAGGACACTGAGAAAGAG630.L0836H04-3C030038J10RikRIKEN cDNAL0836H04Mm.212874Chromosome 6TAGAATGGGAC030038J10ATTTTCTGTCTgeneCATAGTGACATATTGCTATGTTTAACAGTGAACACTCAC631.K0614A10-3Sh3kbp1SH3-domainK0614A10Mm.254904Chromosome XTGACGGTATATkinase bindingTTGCAAAAAGprotein 1AGAAAGAAAAATCTGGTATTTGCAATGATCTGTGCCTTC632.H3156B08-36620401D04RikRIKEN cDNAH3156B08Mm.86150Chromosome 16GAAATATCATT6620401D04TGTAGCTTTAAgeneGGCTAGAAAATGAAAAAGAATCCAAGCCAGTAGAAGGC633.C0334C11-3B230339H12RikRIKEN cDNAC0334C11Mm.275985Chromosome 8ATACCAGGAAB230339H12AATAAAAGTAgeneCCAGTAAGGAAGCATCAAATCAAGATGTCATAGTCAGTGG634.H3103G05-3BG071839ESTsH3103G05Mm.17827Chromosome 3CAGTGTAAATABG071839TAGCATATGGTTAGGTGGTGAGAAAATGATCTTGAGACTGATAAGAATC635.C0205H05-31600010D10RikRIKEN cDNAC0205H05Mm.86385Chromosome 3ATCCTTTAGAT1600010D10GTTAGTACAGTgeneGTTTATGAGAAAACTGTTACTAGAAGCTGAAGAACAGC636.L0513G12-3QkquakingL0513G12Mm.2655Chromsome 17AGTGTTCTATATGTGTAAATTAGTATTTTCAACTGGAAAATGTTGGCTGGTGCAAAAGGC637.C0100E08-3Pdap1PDGFAC0100E08Mm.188851Chromosome MultipleGTCTGGGCTAGassociatedMappingsTGCCCGTTTTTprotein 1AACCCTACCCATTGATCATTTCAAGAAACCTCTGGTTA638.J0055B04-3transcribedJ0055B04Mm.228682Chromsome 16TGTAAGACCATsequence withTTCTAAATTGCstrongTGGTAATAGAAsimilarity toACTCATGGCAGproteinTAAAAATGTAApir:S12207CCTCG(M. musculus)S12207hypotheticalprotein (B2element)-mouse639.J0008D10-3Mbpmyelin basicJ0008D10Mm.2992Chromosome 18ACTGGAATAGproteinGAATGTGATGGGCGTCGCACCCTCTGTAAATGTGGGAATGTTTGTAACTT640.K0319D09-3Mtm1X-linkedK0319D09Mm.28580Chromosome XTCTACTAGAAGmyotubularGGTTAAAAGCCmyopathy geneATATGAATGCA1AGAAATCATTTGAGGCTTAAAATGCTG641.C0243H05-3Galnt7UDP-N-acetyl-C0243H05Mm.62886Chromosome 8GGACACCATTTalpha-D-TTCATGTTAAAgalactosamine:TAGATTTTAACpolypeptide N-CTCGTATCTATacetylgalactosaGCATAGGCTAAminyltransferaseGGTGG7642.L0841H10-3BM116846ESTsL0841h10Mm.65363Chromosome 2TAGATAAAGCCMN116846CGTATGAGAAGAGAAAACCAAATTAATCCACTTCAGCAAAAAGAAAGCC643.K0334D05-3Ccn1cyclin D1K0334D05Mm.22288Chromosome 7CAATGTCAGACTGCCATGTTCAAGTTTTAATTTCCTCATAGAGTGTATTTACAGATGCCC644.L0209B01-3L0209B01-3L0209B01No ChromosomeCTTTGGGGGGGNIA MouselocationGTTTTGGAAAANewborn Ovaryinfo availableCCGGTTTTTTCcDNA LibraryGGGGGGGTTTCMus musculusCTTTTGGGGGGcDNA cloneTTTTTL0209B01 3′,MRNAsequence645.K0151H10-3BB129550EST BB129550K0151H10Mm.283461No ChromosomeGCCATACAGCTlocationTATATTTGTACinfo availableTGGTATGTCCAGAAATCATGGAGGAAAGAAAAGTAAAA646.L0505B11-3Ammecr1AlportL0505B11Mm.143724Chromosome XTGGTGTTTTGAsyndrome,TTACAGTGAGAmentalCATCACAGGTTretardation,ATCTAAAAGCCmidfaceCTTCGTTATAAhypoplasia andCCAGCeliptocytosischromosomalregoion gene 1homolog(human)647.L0944C06-3BM120800ESTsL0944C06Mm.217092Chromosome 3: not placedTATTTGGTGGTBM120800AAAGAATATGGTTGAAAATTGTCATCCACATGCATGCATCAAGTAACAC648.J0027C07-3Mrps25mitochondrialJ0027C07Mm.87062Chromosome 6CGAGGAGTTATribosomalTAGGGAGAATprotein S25CATGGAGCCACATAAGAAAATCTTGGGCAAGAAAAGAGG649.L0855B04-3Wdr26WD repeatL0855B04Mm.21126Chromosome 1TGGTGACAGGdomain 26ATTACGTGAAAATCTCTGACATTGTGATAAACTGGATAAAGGCTTAAGAG650.H3060H05-3Mus musculusH3060H05Mm.11778Chromosome 1ACCCTTTGCTTcDNA cloneAAATAGTGGGMGC:28609AAAACGTGAAIMAGE:42185TGTTTAGCATA51, completeATATAAAAACcdsATGCAGGC651.K0330609-35830461H18RikRIKEN cDNAK0330G09Mm.261448Chromosome 14GTTGGACTCTA5830461H18ATACAACTGACgeneCATTGAAAAATGAACAACGGCTTATTGTTTTGTAAACAG652.L0803E07-3Dpys14dihydropyrmid-L0803E07Mm.250414Chromosome 7TTCTACAAAGinase-like 4TGTGTTTCTATAGGATTACTAGAGTAGCGGTTTTGTACTGTGAGGAAAC653.L0283B01-3Ivns1abpinfluenza virusL0283B01Mm.33764No ChromosomeTAGATAACAGTNS1A bindinglocationGACTATTGACGproteininfo availableATTTTAGTAAAAGAAAGTTGACATGCGTACCGCTACCT654.L0065G02-36530401D17RikRIKEN cDNAL0065G02Mm.27579Chromosome XGGGGGGACAG6530401D17TTAATATCGTTgeneTGTTAGATACCATAAGTGGTGGAAATAAAGTGACTAAAG655.C0949A06-3Mus musculusC0949A06Mm.71633Chromosome 13AAAGAGGAAA0 day neonateCTGTCCTATTTskin cDNA,CTCAACTGATARIKEN full-AGTACTCCTGGlength enrichedTAAGATGTAATlibraryATTTGCclone:4632424N07product:unknownEST, fullinsert sequence.656.H3100C11-3BG071548ESTsH3100C11Mm.173983Chromosome Un: notCAAATGTACTGBG071548placedAGAAACAAAATCATGAACGACCTTGAAATCACCTTCTTATTTCAGCTCC657.C0142H08-33110020O18RikRIKEN cDNAC0142H08Mm.117055Chromosome 5AACATAAATCA3110050O18AAATATACTTAgeneGGAATATTTACAATTAAACATGATGTTTTAAACTTAGT658.L0945G09-3Bcl2111BCL2-like 11L0945G09Mm.141083Chromosome 2GACTATTTATT(apoptosisAGATTAGAAAfacilitator)GTCATGTTTCACTCGTCAACTGAGCCAAATGTCTCTGTG659.L0848H06-3E130318E12RikRIKEN cDNAL0848H06Mm.198119Chromosome 1ACAAACACATE130318E12GAAAAAATCAgeneAGTAGGAACTGGAGAAACGTCTCACAGTTAAGAATGTTTG660.K0617B02-3Bmp2kBMP2K0617B02Mm.6156Chromosome 5AATTCACAGATinducibleGGCTTACATTTkinaseATGTAAAGAATTCCTGTAAGGCACTCATGTTTGACATC661.C0203D07-3Pftk1PFTAIREC0203D07Mm.6456Chromosome 5TATACCAAACTprotein kinase 1GAAAACGTTTAAATCTCAAATGAAGTAAGCAAGGTTTTGTTCTCCCTGC662.L0267A02-32210409B22RikRIKEN cDNAL0267A02Mm.30015Chromosome 4TAGCCATTTAG2210409B22GAGATGTCCCTgeneTCAAAGTGACGTGATGATGGACTTGCACTTGGGAATCA663.J0086F05-3transcribedJ0086F05Mm.31079No ChromosomeGCTCAGCTTAGsequence withlocationGCTAGACTTTGmoderateinfo availableACCAGGTAAGsimilarity toCAGAAGAAATproteinGAGAAACAAAsp:P00722 (E.ACTCAGCAcoli)BGAL_ECOLIBeta-galactosidase(Lactase)664.C06606A03-3Rps23ribosomalC0606A03Mm.295618Chromosome XTATCACTGGAAprotein S23TATTGAAAGGTTGTATGTAGTATGGGAGATCAACTTTCTTCCCTAAGGT665.L0902D02-3Ncoaoipnuclear receptorL0902D02Mm.171323Chromosome 4ACTGCTGAGAAcoactivator 6AAACAAAATTCinteractingACTACATACCTproteinCAATAGTTATTTACCATGAGATTGGCG666.H3060C12-3BG067974ESTsH3060C12Mm.173106Chromosome 1GAAGGAAATGBG067974CAAACACCTTTGAACTTCAATTCTTTCAGTAGGAAAACAAGAATTGTCCC667.C0611E01Tor3atorsin family 3,C0611E01Mm.206737Chromosome 1AGAAAAACACmember ATAAACTCCAAATTAGTATAATAACGAGCACTACAGTGGTGAAAAAGCTCC668.U54984.1Mmp14matrixU54984Mm.19945Chromosome 14AAAGGAATCTTmetalloproteinaseAAGAGTGTAC14ATTTGGAGGTG(membrane-GAAAGATTGTTinserted)CAGTTTACCCTAAAGAC669.H3089F08-30610013E23RikRIKEN cDNAH3089F08Mm.182061Chromosome 11GAAATGGATTT0610013E23TGAGGCTTTGAgeneAAATGAAAATGGCTAGTQTCTCAAAGATGTCAGTATCC670.K0633C04-3Ebi2Epstein-BarrK0633C04Mm.265618Chromosome 14ACTATTTCTTGvirus inducedTCAATAGTTTGgene 2GCAAAAGACGACTAATTGCACTGTATATTGCCAGTGTA671.J0943E09-3Nup62nucleoporin 62J0943E09Mm.22687Chromosome 7TCCTCTAAAGATGTGTCTTATATACATGATTGTCATTGGTGGGCTCAAACAATAAGGGTG672.L0267D03-3Dcndecorin:0267D03Mm.56769Chromosome 10TTGGAAACTACAAGTAACCCTCAGACGGCCTAATTCTTATAATCCGGAAAAACACCCCAA673.L0250B09-3111031E24RikRIKEN cDNAL0250B09Mm.34356Chromosome 8GTGTGATAATC1110031E24TTTTCATGTTTTgeneCTAGAGCAAAGACAAAGCAGTTACTCTTCTATCGCAA674.L0915B12-3Etv3ets variant geneL0915B12Mm.34510Chromosome 3GGCTTTAGAGA3AAACTTCGGTCTTCAAAGAACTCTTCTAATTAGTTCCTTCTTGGAAAAA675.NM_009403.1Tnfsf8tumor necrosisNM_009403Mm.4664Chromosome 4AAAGTAGGAGfactor (ligand)ATGAGATTTACsuperfamily,ATTTCCCCAATmember 8ATTTTCTTCAACTCAGAAGACGAGACTG676.C0308F04-32700064H14RikRIKEN cDNAC0308F04Mm.24730Chromosome 2AGTCCTCTGCA2700064H14TGTTTCCAAAAgeneTTTCCTTTACATGAAGGCTATATTGGATCAGAGCTTAC677.C0288G12-36030400A10RikRIKEN cDNAC0288G12Mm.159840Chromosome 5AAGAATAAAT6030400A10CACTTGAAATCgeneATACTGTTTTTGGAAATCCAAACTGTTTAAAGAAAACTT678.H3005A11-3Fancd2FanconiH3005A11Mm.291487Chromosome 6GTTAGATGCCAanemia,TTGAAGGGGAcomplementationAATAACTTTGGgroup D2CTAATAGCTTGGAAAACTCAGTACTAAG679.H3121H07-32810405I11RikRIKEN cDNAH3121H07Mm.73777Chromosome 18AGCAGATATGT2810405I11GACTTCTCATAgeneTACACAGTTACGCTAACTCAGGTGTATGATGAATACAG680.K0124A06-3BM222608ESTsK0124A06Mm.221709Chromosome 19TGTCTATGGGABM222608GAAGTAATAGCCTGAAATAAGATAAGGCTCAAACAAACACTACTTACTT681.NM_010835.1Msx1homeo box,NM_010835Mm.259122Chromosome 5GGGAAGAAAAmsh-like 1AGAATTGGTCGGAAGATGTTCAGGTTTTTCGAGTTTTTTCTAGATTTACA682.K0134C07-3Falzfetal AlzheimerK0134C07Mm.218530Chromosome 11CTTGAAGAAAantigenAGTATATCACGTAGGCATAGATGAGAAAGCCGTTTGATCAAGTCTGGTTA683.K0424H02-3Pfkpphosphofructok-K042H02Mm.108076Chromosome 13TCCTTCAGTCAinase, plateletGATATCTGTCCCAGAGAAAGGAAAATAAGGAGCATGGTAAGAAATGAGT684.H3153G06-38030446C20RikRIKEN cDNAH3153G06Mm.204920Chromosome 13TATGGAATGGA8030446C20GAAATAAATAgeneCATCTGTGTTGAAGAACCTTTTGATGGAACTAATACCGC685.H3071C09-3BG068971ESTsH3071C09Mm.162073Chromosome 6AGGTCAATGTTBG068971AAGTTTTCTGAGTTTAATATATAGTTAGGGTGAAAGACTTAGCACACGG686.L0243B07-3PossiblyL0243B07Data not foundNo ChromosomeAATGCTTAACTintronic inlocationTTGAGTCACACU008124-info availableTGTTTACCCTTL0243B07CCTATGAGGTTGCATTTTGACAACAAC687.C0143D11-3IiIa-associatedC0143D11Mm.248267Chromosome 18TAAAGGGAACinvariant chainCCCCATTTCTGACCCATTAGTAGTCTTGAATGTGGGGCTCTGAGATAAAG688.L0512A02-3Snx5sorting nexin 5L0512A02Mm.20847No ChromosomeCCCCTTTTGTlocationAACTGGGATATinfo availableAAATCCTTGAAAGAAAGGAGAATTTAGAGTTTTGCCCC689.K0112C06-3Atp8a1ATPase,K0112C06Mm.200366Chromosome 5GTCAGTGAGTTaminophospholipidGGTTTCCTTTCtransporterCATCAGGAAA(APLT), class I,AATGGATTCTGtype 8A,TAAAGAGTCAmember 1GGGCGTT690.H3053A01-3Tnfsf13btumor necrosisH3053A01Mm.28835Chromosome 8GAAAGCCGTCfactor (ligand)AGCGAAAGTTTsuperfamily,TCTCGTGACCCmember 13bGTTGAATCTGATCCAAACCAGGAAATAT691.C0668F08-3Atp6ap2ATPase, H+C0668F08Mm.25148Chromosome XGAAATATGTTAtransporting,ACTAAGAGCAlysosomalGCCCAAAAATaccessoryACTGGATATGCprotein 2TTATCCAATCGCTTAGTT692.K0417E05-3Osmroncostatin MK0417E05Mm.10760Chromosome 15GTATACAATGCreceptorTATTTTTAGGTTAAGGCCTAAACTTCTGAAGATCTTGGTAACAGCAGAG693.NM_010872.1Birclbbaculoviral IAPNM_010872Mm.89961Chromosome 13GGATGAAGTGrepeat-GAAGATTACTGcontaining 1bGCAGGTCCAAAAACCTGATTTTCTAGTACATTTCACTCT694.L0262G06-3CfhcomplementL0262G06Mm.8655Chromosome 1TTCAATCAAGAcomponentAAGTAGATGTAfactor hAGTTCTTCAACATCTGTTTCTATTCAGAACTTTCTCAG695.J0249F06-32210023K21RikRIKEN cDNAJ0249F06Mm.28890No ChromosomeAAATTTTCTTA2210023K21locationAAGCTATGAACinfo availableTCTGACTTTTGATTTTGTGTTTCCATTTAGTAGAAACT696.C0170A02-3Serpinb9serine (or)C0170A02Mm.3368Chromosome 13AGAATCTCACTcysteine)ACTAAAGTCAAproteinaseGTATAGAAATAinhibitor, cladeACTGTTCTTATB, member 9GTTTTCCTCCAAGGCC697.H3076C12-3Fac14fatty acid-H3076C12Mm.143689Chromosome XATCTTTGGCTACoenzyme ATATTTTCCTGGligase, longTAGCATATGACchain 4AAATGTTTCTACAGTGAGAAGCTGAGA698.H3155C07-31810036L03RikRIKEN cDNAH3155C07Mm.27385Chromosome 15GGGTTATAATG1810036L03CACTGAGATCCgeneAGAAGTTGGGAAAACTCAATAAATGTACAAAGGAAAGC699.K0331C04-3Sdccag8serologicallyK0331C04Mm.171399Chromosome 1TACTTGTGTGAdefined colonCAAGCTAGAGcancer antigenAAGTTACAGA8AGAGAAATGACGAACTAGAAGAGCAATGC700.J0538B04-3Laptm5lysosomal-J0538B04Mm.4554Chromosome 4TAAATAATCCCassociatedTTCCCATGAGCproteinCCACTGCTCTGtransmembraneAATGGACAAG5CTGTCCTTATCTTCAAT701.H3014E07-31810029G24RikRIKEN cDNAH3014E07Mm.27800Chromosome 18AAATAGTTGTT1810029G24TTTAAGGTTGAgeneAGGAAGAGACATTCCGATAGTTCACAGAGTAATCAAGG702.K0515H12-32900064A13RikRIKEN cDNAK0515H12Mm.268027Chromosome 2TGAATCTACAG2900064A13GCAACTCTTCAgeneTCTCTGTAATGCTACCTGACTTCTCTTGTGAGGAGCTG703.H3159D10-3BG076403ESTsH3159D10Mm.103300Chromosome 14TGGCAAAGAGBG076403TAGATGAGAAAATGTTGGATTTAAATCAGCAGACTCATTTCATACTTTGC704.K0127F01-3Prgproteoglyan,K0127F02Mm.22194Chromosome 10ACCACGTTTAAsecretoryATGACCAGTCTgranuleCAGGATAAAGAGTTTTACAGAAAATTTAAAATGCCTGG705.L0919B08-3Bnip31BCL2/adenovirusL0919B08Mm.29820Chromosome 14GACATCGTTTTE1B 19kDa-CTCTCTAAATTinteractingCAGTAGCAGTTprotein 3-likeTCATCGACAGTGCCATTGAACTATGGG706.J0904A09-31110060F11RikRIKEN cDNAJ0904A09Mm.4859Chromosome 4TCTGTGGGGTT1110060F11CTCATGCCAGTgeneGTCTGAAATCTCACCTCACTAGAGATGTTTCTCGAATT707.L0270B06-3D11Ertd759eDNA segment,L0270B06Mm.30111Chromosome 11TTCCAGTTCTCChr 11,ATGTCTTGAGAERATO DoiTTTCAAGTAAA759, expressedGATGTGTTAGTGTAAGCTCAGATCCGA708.K0230D06-3EaflELL associatedK0230D06Mm.37770Chromosome 14AACCATTGGGAfactor 1AAATGCAATACAGATAAACTAGAGATTCGTATAATGCCACGTGTTAGCT709.K0611A03-3AI447904expressedK0611A03Mm.447Chromosome 1GTGAATGGAGTsequenceGTTTACTGTATAI447904GTAAGAAAGAAGAAAAGTGGAACTACATTTGCTATGAG710.H3155A07-3BG076050ESTsH3155A07Mm.182857Chromosome 5TTCACAATTTABG076050GACACAAGATTTGGAAGATTGAAACTGACATGAAAGTCTTCTTCCTGAG711.H3028H11-3Ctshcathepsin HH3028H11Mm.2277Chromosome MultipleGAAGATTTTTTMappingsGATGTATAAAAGTGGCGTCTACTCCAGTAAATCCTGTCATAAAACTCCA712.L0001D12-34833422F06RikRIKEN cDNAL0001D12Mm.27436Chromosome 15AGAATGAACC4833422F06AGAATGGAGAgeneAAACGTAAAATTTGAAGAATCTCGTTGAAGAGCTATTTGC713.L0951G01-3BG061831ESTsL0951G01Mm.133824Chromosome 10TCGACAAGAGBG061831GTAATCCGAGAAATGGAGCAGAAAACCTCCTTGCACTTCAGTGATATACA714.H3035G02-3A1314180expressedH3035G02Mm.27829Chromosome 4TATATGCAACTsequenceTCATAGATCCTA1314180CTGCAATATGTACTTAGCTACCTAAGCATGAAATAGAC715.C0925G02-3Fer113fer-1-like 3,C0925G02Mm.34674Chromosome 19CGTCATATATCmyoferlin (C.CTATTTGTAATelegans)CAAGAGGAAAGACTACATTAAGAAGATAGGGTGCATAG716.C0103H10-3Il17rinterleukin 17C0103H10Mm.4481Chromosome 6CTCAGATCAGTreceptorTCTTTAGAAAGAGCTGGTATAGAAATGGGTGATGTAAAACTTGAGAAGC717.H3129F05-3Mrpl16mitochondrialH3129F05Mm.203928Chromosome 19AATGAAAATCTribosomalGCGTCTAACTTprotein L16TTGAAAGTAAGTGTTAACTTACTTGAATGCTGGTTCCC718.L0942B12-3Mus musculusL0942B12Mm.214553Chromosome 15AATCTTCGACC12 days embryoAGACATTGGATspinal ganglionATTTGAACTATcDNA, RIKENCCTGAAACATTfull-lengthTTAGAAATATCenrichedCAGGClibrary,clone:D130046C24product:unknownEST, fullinsert sequence719.L0009B09-3Plcg2phospholipaseL0009B09Mm.22370Chromosome 8TACCCCATTAAC, gamma 2AGGCATCAAATCCGGGTTTAGATCAGTCCCTCTGAAGAATGGGTACAGT720.C0665B08-3Sh3bp1SH3-domainC0665B08Mm.4462Chromosome 15TTTTTTCTCTTGbinding proteinCCAATGTATTT1TTGTAAGGCTCGTAAATAAATTATTTTGAACAAAACA721.H3102F04-3Rgs10regulator of G-H3102F04Mm.18635Chromosome 7CACACCCTCTGproteinATGTTCCAAAAsignalling 10GCTCCAGGACCAGATCTTCAATCTCATGAAGTATGACA722.K0547F06-3transcribedK0547F06Mm.162929Chromosome 19CCCAGGTATTTsequence withCTAAGCATGCTmoderateAGGTTTGAGGTsimilarity toCATTTACCATGproteinTTCAAATAAAAsp:P00722 (E.GACGGcoli)BGAL_ECOLIBeta-galactosidase(Lactase)723.H3087C07-3Glb1galactosidase,H3087C07Mm.255070Chromosome 9GGAGCAAAACbeta 1TTGAATAATGTCCTTTATCCTGATTTGAAATAATCACGTCATCTTTCTGC724.J0437D05-3AU023716ESTsJ0437D05Mm.173654Chromosome XTGGAATAAGAAU023716AAGAATCTGTGGTAGAAATAATAGACTTGCTACATAGGGTTAGCTAAGGC725.H3156A09-3Pex12peroxisomalH3156A09Mm.30664Chromosome 11ACCACAGTTTAbiogenesisTCAGCATTTGAfactor 12AGATTTCCTTGATGATCCATACTTGTCTTGGGATAGGG726.G0108H12-3Ly6elymphocyteG0108H12Mm.788Chromosome 15AGGGTCAGCGantigen 6CCGAATCTTGTcomplex, locusGGACACACTGEACAAGGATGTCTAATCCAAATAGATGTAT727.H3098D12-5Map2k1mitogenH3098D12Mm.248907Chromosome 9AGTGGAGTATTactivatedCAGTCTGGAGTprotein kinaseTTCAGGATTTTkinase 1GTGAATAAATGCTTAATAAAGAACCCT728.C0637C02-3Zmpste24zincC0637C02Mm.34399Chromosome 4TTTGGGCCCTTmetalloproteinase,AAAAACATATTSTE24TCAGTTTTGCChomolog (S.CAAGTGAGGCcerevisiae)CTTAAAAATTGCCCATG729.H3119B06-3Atplb3ATPase,H3119B06Mm.424Chromosome MultipleAAAGGAAAATNa+/K+MappingsAAAGTGGATCTtransporting,GAAAGTAGACbeta 3TCTGCTTCTGCpolypeptideGCATGTGTGAGTGGTGCC730.C0176B06-3Ubl1ubiquitin-like 1C0176B06Mm.259278Chromosome MultipleTTCACTCCTGGMappingsACTGTGATTTTCAGTGGGAGATGGAAATTTTTCAGAGAACTGAACTGTG731.C0626D04-39130404D14RikRIKEN cDNAC0626D04Mm.219676Chromosome 2CACCATCCTTC9130404D14CAGAATATGGTgeneATGAAAAATCTATGCAAACTGTGTAAGCTTTTGCTCAT732.H3155E07-3Dock4dedicator ofH3155E07Mm.145306Chromosome 12TTGTGGAGTGTcytokinesis 4GAAATAAAGGATAATTGCCTACCTCTAGCAAGTGGATCTTATTATGTTG733.C0106A05-3H2-Eb1histocompatibilityC0106A05Mm.22564Chromosome 17ACCAGAAAGG2, class IIACAGTCTGGACantigen E betaTTCAGCCAACAGGACTCCTGAGCTGAGATGAAGTAACAA734.H3037B09-3Mus musculusH3037B09Mm.274876Chromosome 7GATACTGCCGG12 days embryoCTTTGAAAATGspinal cordAAGAACAGAAcDNA, RIKENGCTAAAATTCCfull-lengthTGAAGCTTATGenrichedGGTGGClibrary,clone:C530028D16product:2310008H09RIKPROTEINhomolog [Musmusculus], fullinsert sequence.735.H3003b09-3F730017H24RikRIKEN cDNAH3003B09Mm.205421Chromosome 14CCATTTGAGCCF730017H24TCACTGCAATGgeneTTAGTGCAGAGGAGAAAACAATTTTTAATGTAATCTTG736.C0909E10-3Pignphosphatidylino-C0909E10Mm.268911Chromosome 1GGCAACTTGTAsitol glycan,AAGTGTGTTCAclass NTTCTAACTGTTAAACTGAGAAAACTTGAGAACATACTG737.H3045G01-3BG066588ESTsH3045G01Mm.26804Chromosome 14CAGAAGAGATBG066588TCTGAAAATGTTAGTTGTGGTGACTCTAATGTAGATCCATAATCTGAAAAG738.H3006E10-3transcribedH3006E10Mm.218665Chromosome 15TATCGTAAGTTsequence withGCACCTATTGTweak similarityTAAGTGGAAAto proteinATGCTCTGATTsp:Q9H321ACACTCAGGA(H. sapiens)AGCTGGGVCXC_HUMAN VCX-Cprotein(Variablycharged proteinX-C)739.H3098H09-32310016E02RikRIKEN cDNAH3098H09Mm.21450Chromosome 5TGTTTTGTCCC2310016E02TAAATCACCACgeneCACTCACTATTTCTCCCAGGGTCTGATAATGCCTTTAC740.J0540D09-3Adam9a disintegrinJ0540D09Mm.28908Chromosome 8AGCCACTTTAAandCTCTAAACTCGmetalloproteinaseAATTTCAAAGCdomain 9CTTGAGTGAAG(meltrinTCCTCTAGAATgamma)GTTTA741.L0208C06-3Pknox1Pbx/knotted 1L0208C06Mm.259295Chromosome 17GCTTTGTTTAAhomeboxATGGTCAGACTCCCAAACATTGGAGCCTTTTGAATGTGTTCTGAGACCT742.H3154G05-3NapgN-H3154G05Mm.154623Chromosome 18CCTTAGAAAGAethylmaleimideTGGTAATTCACsensitive fusionTTTAGGTAAAAproteinGTACTATTTCAattachmentCGCCATTATGAprotein gammaAACCC743.L0854E11-31500032M01RikRIKEN cDNAL0854E11Mm.29628Chromosome 19TAAAATGAGG1500032M01CTTTTGGAAAGgeneAAAGATGAAAACGTAGAATGTAGTGCTAAGAACGTTTCC744.H3014C06-3B2mbeta-2H3014C06Mm.163Chromosome 2GCAGTTACTCAmicroglobulinTCTTTGGTCTATCACAACATAAGTGACATACTTTCCTTTTGGTAAAGCA745.K0538G12-3Ccr2chemokine (C-K0538G12Mm.6272Chromosome 9TGCTTAGAACTC) receptor 2ACATAGAATCAGAAGCAAAATGGATGCCTTAGCACTGAGGAAAGGTTTC746.J0819C09-3C030002B11RikRIKEN cDNAJ0819C09Mm.70065Chromosome 10GGTTTTCGAACC030002B11CACGTACCTTTgeneATGCCTCGTGATTGTGAAACATTGACTTTTGTAAACCC747.C0175B11-3Histlh2bchistone 1, h2bcC0175B11Mm.21579Chromosome 13GTTCACTGTAGAAATTTGTGATAAGAAAGACACACAGACGTAGAAAATGAGAATACTTGC748.H3009B11-3Nufip1nuclear fragileH3009B11Mm.21138Chromosome 14AAGACTTTTTX mentalTGGACTTAATAretardationCTGATTCTGTGproteinAAAACTGAAGinteractingAAGTGTAGATGproteinTCTCCC749.H3135D02-3Lamp2lysosomalH3135D02Mm.486Chromosome XCTGGTGTGGGAmembraneTATTTTCCACAglycoprotein 2CTTTAGAATTTGTATAAGAAACTGGTCCATGTAAGTAC750.K0540G08-31200013B08RikRIKEN cDNAk0540g08Mm.247440Chromosome XTAAAGGTTTTA1200013B08GTGTCCTAACTgeneCCCCAGGATCAGGAGATTATCCCAACTATTTCTGGGGT751.H3089H05-3Lnx2ligand of numb-H3089H05Mm.34462Chromosome 5CTGAATTTTGAprotein X 2TCACTTGTGGTTTCTCATGGTGACCTCCATTTGCAACAAAAAGATGTCT752.J0203A08-3C85149ESTs C85149J0203A08Mm.154684Chromosome 2TGTGCTTTACCAAAATGGGAAATAATTCTGCTTTAGAGGATACTATCAAGACAACCTTAC753.H3119F01-3Mcfd2multipleH3119F01Mm.30251Chromosome 17TCTGTGAGATGcoagulationTTGTAGACATTfactorCCGTAAGAGAdeficiency 2ATCCAGAATGATAGCAGGATCAGGAAAG754.H3134C05-3Mglapmatrix gamma-H3134C05Mm.243085Chromosome 6CTTACATGATCcarboxyglutamateTCCTAAAAGGA(gla) proteinTGGGCCCCTCCTTCCTTTTGCGGGTTGAAAGTAATGAA755.C0147D11-3B230215M10RikRIKEN cDNAC0147D11Mm.41525Chromosome 10CTGTTTAAAAAB230215M10ATGAAATCAGgeneGAAGCTTGAAGAAGACGATCAGACGAAAGACATTTGAGC756.C0949H10-3Sulf1sulfatase 1C0949H10Mm.45563Chromosome 1TGAATATAGTAGGGCCATGAGTATATAAAATCTATCCAGTCAAAATGGCTAGAATTGTGC757.K0114E04-3BM222075ESTsK0114E04Mm.221705Chromosome 19GGGGGAAATTBM222075CTATATGAGCTTCGTTTTCTAATGACTTACATGGATAGTATGGAAACTTC758.H3012C03-3Cappa1capping proteinH3012C03Mm.19142Chromosome MultipleAAACTTGAAAalpha 1MappingsACACAGACATTGAAGGAATCATAGGTATTTTTGCTTTATGCTCTCTGGCA759.C0507E11-3BE824970ESTsC0507E11Mm.139860Chromosome 16AATAAGCAGGBE824970AAGAATTTGACTTGGAAAACTAATACACGCATGTTAGGCATTCTCAAGGC760.H3158D06-3Lnklinker of T-cellH3158D06Mm.200936Chromosome 5TCCCACTGTTTreceptorACAGATGTAGTpathwaysTCTTGTGCACAGGTGCCACTAGCTGGTACCCTAGGCCT761.C0174C02-3Pold3polymeraseC0174C02Mm.37562Chromosome 7TATTTTTGTCA(DNA-TTGCCTCTAGTdirected), deltaGATTTTTGTAA3, accessoryATGGGAATGGsubunitAAAAGTACAAGGCAACC762.C0130G10-3Cklfst7chemokine-likeC0130G10Mm.35600Chromosome 9TTAACTGGCCTfactor superGTCAAACTGGTfamily 7CTTGAAGCGTCTCTAAGTGAAGAGCCAGAAGAAACCCT763.C0137F07-3Rik3cbphosphatidylino-C0137F07Mm.213128Chromosome 9CAATGTGATTTsitol 3-kinase,TTCAATGGTATcatalytic, betaTAGTTCAAATTpolypeptideGACGTGGATTCATGCCACATGGAAATC764.H3115F01-32610027O18RikRIKEN cDNAH3115F01Mm.46501Chromosome 12AACTGAATAA2610027O18AGTTGACCAGAgeneAAGTGAAAGTCTTTAACATGGATGGAAAAGACTTCATCC765.H3097F03-3Mus musculus,H3097F03Mm.227202Chromosome 3GGATATAAAGTcloneGTATTTCTTTCIMAGE:53723AGTGATTTCTC38, mRNAAGTGCATAAGAAGTGCATAAGTCTCAG766.H3059A05-3Mad211MAD2 (mitoticH3059A05Mm.43444Chromosome 6TAGCTTTTTAAarrest deficient,AAGAAGTTTTThonolog)-like 1CTACCTACAGT(yeast)GACCATTGTTAAAGGAATCCATCCCAC767.L0935E02-3Sykspleen tyrosineL0935E02Mm.248456Chromosome 13ATTTGCAAGGTkinaseCAGAAACTAGCCAAGGTCCTTCTCAGGCATCTATCCTTAACTTGGTCTC768.C0946F08-31110014L17RikRIKEN cDNAC0946F08Mm.30103Chromosome 11TTGGAATTTGA1110014L17GGAGGAGAAAgeneTGAAAAAACAGTGTGTCCCTGGTGTCACCCTGGCATCAT769.H3079F02-5PossiblyH3079F02Data not foundChromosome 10TCTTATGATTTintronic inAAGTGATTGGTU011488-GGATAAATGTAH3079F02TAGGAATTTTACACTCCAGCAGCATGG770.H3137E07-3III0rainterleukin 10H3137E07Mm.26658Chromosome 9GCCTCAAATGGreceptor, alphaAACCACAAGTGGTGTGTGTTTTCATCCTAATAAAAAGTCAGGTGTTTTG771.C0143H12-3GalnsgalactosamineC0143H12Mm.34702Chromosome 8CCGTACACAAA(N-acetyl)-6-AGTGAAGATTTsulfate sulfataseCAGCGAAATGCCAAGGAAGTGCCATCTATCTGGCTTCT772.H3114D03-3Man2a1mannosidase 2,H3114D03Mm.2433Chromosome 17AAGAAATGCalpha 1TGTATGATGTTAGAAGACATTGTAATTATCATCCCGTGTCTTTGCTGTAC773.H3041H09-3BG066348ESTsH3041H09Mm.270044Chromosome 8GGCATTTCAGTBG066348TTATCTTGGGTTTGTAATTAGTTAAAACAAAAACCAACCTAGGTCTGTG774.C0628H04-3Slc2a12solute carrierC0628H04Mm.268014Chromosome 10ATTAGCCAAGGfamily 2,AGTCCGGACATmemeber 12AATATTTATCCAGATCTCTAAGCAGTTAGCTTTAAATT775.K0125E07-3IfngrinterferonK0125E07Mm.549Chromosome 10TACATTAGCTAgamma receptorATACTAACCACATAGAATATCAGACTTAGATACGTGAATAGGGATCCTG776.G0115E02-3SdcbpsyndecanG0115E02Mm.276062Chromosome 4AAGATTTTCTAbinding proteinGTCACTGCATAAAGGAAACGCCTAAGAGTTGCCGTATTGCTTTCTGAGA777.C0032B05-3Rap2bRAP2B,C0032B05Mm.26939Chromosome 3ACAAGAATTCAmember ofTTCTTAACATTRAS oncogeneTGAACGAGTGTfamilyATTTGCTTAGGTCGATGAAAGTGTTGC778.H3141C08-3Ofd1oral-facial-H3141C08Mm.2474889Chromosome XAGGATTTTCTCdigitalATGAAGAACCsyndrome 1AGATGACATGTgene homologGGTAATAACAT(human)TAGCTGTCTAGTTTCTC779.H3157C05-3BG076236ESTsH3157C05Mm.182877Chromosome 1TAGAGTCTGABG076236AGAACAGAAATTCAAGGTCATTTTCAATTACAGAGTGAGGTTAGAGCCA780.H3076A01-35031439G07RikRIKEN cDNAH3076A01Mm.121973Chromosome 15TCTAAAACATG5031439G07CCAAATGACTTgeneATGTCACAAAGAATAGGTCCTAATATACTGTATACCCC781.H3080D06-3BC01807cDNA sequenceH3080D06Mm.139738Chromosome 13GTGTTTCTTCCBC018507CATTTGTAAATGTCCTGAACCATAAATTACTATCAGGATTAACTGACAG782.L0518D04-3Uap1UDP-N-L0518D04Mm.27969Chromosome 1GAAGCTGGAAacetylglucosamineGCATTTGTTTTpyrophosphorylaseTGAAGTTGTAC1ATATTGATAAGTCAGCGTATGTGTCAGA783.K0541B11-3BM239901ESTsK0542B11Mm.222307Chromosome 2TTACATGGCAABM239901ATCTGAAAGGAAGACTTAAGCAGGGTAAAGTTAATTGAAAGGAGGAGCT784.L0959D03-3Tnfrsflatumor necrosisL0959D03Mm.1258Chromosome 6AGCAATCTTTGfactor receptorTATCAATTATAsuperfamily,TCACACTAATGmember 1aGATGAACTGTGTAAGGTAAGGACAAGC785.H3035C07-3BG065787ESTsH3035C07Mm.24933Chromosome 1GGTGTAGGAABG065787ATAAAGTTTAGTCAATGTTGAAAATCTCTCCTGGTTGAATGACTTGCTC786M29855.1Csf2rb2colonyM29855Mm.1940Chromosome 15CTTTCAGTCTCstimulatingCTTCTGTGTCTfactor 2CGAACCTTGAAreceptor, beta 2CAGGATGTGATlow-affinityAACTTTTCTAG(granulocyte-ACCACmacrophage)787.C0352C11-3BM197981ESTsC0352C11Mm.215584Chromosome 2GACTGTTTCTGBM197981GGAAAATAAGTATGTGAAGTGATGCAGAAAATCCATCTAGACAGTTGAG788.L0846B10-3BM117093ESTsL0846B10Mm.216113No ChromosomeTGGTGGCTTGAMN117093locationTTGATTTGATCinfo availableTGAGAGCAGTTTATAACATAATGGAGAACTGTTTGCAG789.L0227C06-3Serpinb6aserine (orL0227C06Mm.2623Chromosome 13AGAAGTCTACCcysteine)TTTAAGATGACproteinaseCTATATTGGAGinhibitor, cladeAGATATTCACTB, member 6aAAGATTCTGTTGCTTC790.J0214H09-3Serpina3gserine (orJ0214H09Mm.264709No ChromosomeACTCTCTGGTCcysteine)locationATGATGGTTTTproteinaseinfo availableCCGAAATCAGinhibitor, cladeGTTCCTGACCTA, member 3GGAAAATTTGGGTTAATC791.H3077F12-3Arhhras homologH3077F12Mm.20323Chromosome 5GTTTTTCAT-gene family,GCTmember HTTGGAAGTCTTTTCTTTGAAAAGGCAAACTGCTGTATGAGGAGAAAATA792.C0341D05-3BM196992ESTsC0341D05Mm.222093Chromosome 1GTGTGTAGGAABM196992AATGTAATTAAGTACAAGGCTTGTTTATGGGTGGCTATGGAATGCAGTC793.H3043H11-3BG066522ESTsH3043H11Mm.25035Chromosome 6GTTTCCTCATCBG066522AGGTGTAATGGCGTGTCCTAATGAAGCTATTCTTATGTATAACAGAGA794.K0507D06-3Mus musculus,K0507d06Mm.103545Chromosome 11TGAAAAAATGcloneAAAAGAATCAIMAGE:12632GAGATGAAAT53, mRNAAGGAGCGCTCAGAAGTTTTTATGTTCTCCC795.J0535D11-3AU020606ESTsJ0535D11Mm.26229Chromosome 11AAAGAAATGAAU020606AAACCGTCATTTGCGATTTTCAGGGTACGTTTCTAATGTATCCAGAAGTC796.H3152F04-3Sepp1selenoprotein P,H3152F04Mm.22699Chromosome 15TTTCCAGTGTTplasma, 1CTAGTTACATTAATGAGAACAGAAACATAAACTATGACCTAGGGGTTTC797.L0701F07-3H2-Ab1histocompatililityL0701F07Mm.275510Chromosome 17TTTTGACTCAG2, class IITTGACTGTCTCantigen A, betaAGACTGTAAG1ACCTGAATGTCTCTGCTCCGAATTCCTG798L0227H07-3Clca1chlorideL0227H07Mm.275745Chromosome 3CCCGAGTTACTchannel calciumAACAACATTCTactivated 1TTTGCTATATGTAGATCAAGATTAACAGTTCCTCATTC799.J1014C11-32900036G02RikRIKEN cDNAJ1014C11Mm.80676No ChromosomeGTTTTGGTGCA2900036G02locationAAAGTCGTCCTgeneinfo availableGTGTCTCTTGTTCCCTTCATTAGAAAACATGCTAGAGG800.H3134H09-3BG074421ESTsH3134H09Mm.197381Chromosome 12AGGAAGGAAABG074421ATAGGCTTTGTTGTATGTACATAAGTGGAATTAACAAGAGTCTTTAGTCC801.G0116A07-3Atp6vblc1ATPase, H+G0116A07Mm.276618Chromosome 15TACAGGGAATtransportingGGTCTAAGCATV1 subunit C,ACCATTTCATTisoform 1CACTGTATTAGTAGACATAACTGTTGAG802.L0942F05-3Ostm1osteopetrosisL0942F05Mm.46636Chromosome 10GAAACGGGCTTassociatedTGTTGTAAAGGtransmembraneTAATGAATAGGprotein 1AAACTCCTCAGATTCAATGGTTAAGAA803.C0912H10-30610041E09RikRIKEN cDNAC0912H10Mm.132926Chromosome 13AAGTTAAGGA0610041E09AATACTGAGAgeneATCGGTCAGTTAACACTCTGAAAAGCTATTCAAAGCATAG804.C0304E12-3Pde1bphosphodiesteraseC0304E12Mm.62Chromosome 15AAATACATGCA1B, Ca2+-TTTGTACAGTGcalmoduinGGCCCTGTTCTdependentTGTGAAGTCCATCTCCATGGTCATTAG805.L0605C12-34930579K19RikRIKEN cDNAL0605C12Mm.117473Chromosome 9CCGTTTTATTG4930579K19ATTGGAAATGTgeneAAGACTCAAAGAACTCAGGTTTACTGGCCAAGATGGCA806.K0539A07-3Cd53CD53 antigenK0529A07Mm.2692Chromosome 3GGAAAGAGAGATCAAACTAGGAACCTACAAGATAGTTCACTAGCCTAAGATCTTTACTTG807.L0228H12-36430628I05RikRIKEN cDNAL0228H12Mm.196533Chromosome 9TTGATTGGTGT6430658I05TTCTGAGCATTgeneCAGACTCCGCACCCTCATTTCTAATAAATGCAACATTG808.L0855B10-3BM117713ESTsL0855B10Mm.216997Chromosome 10CTAGTGAAATTBM117713TATGTCAGAATGACATATCTGAACTCTGAATTCATCTCTAGTTTCCACG809.H3075B10-32810404F18RikRIKEN cDNAH3075B10Mm.29476Chromosome 11TAGTTAATACT2810404F18TCTCTGAAATAgeneCATGGTAACAACTAGTAAGCAAGAGATACCGCAGATTG810.L0022G07-3L0033G07-3L0022G07No ChromosomeTGGATTATTCCNIA MouselocationCGCCAAAGCAE12.5 Femaleinfo availableCCCAAGTCGGCMesonephrosCTGTTTAATTGand GonadsGAGAAAGATGcDNA LibraryGAATTAAMus musculuscDNA cloneL0022G07 3′,MRNAsequence811.H3107C11-3Efemp2epidermalH3107C11Mm.471781Chromosome 19GATCCAGGCAgrowth factor-ACCTCTGTTTAcontainingCCCTGGGGCCTfibulin-likeACAATGCCTTTextracellularCAGATCCGTTCmatrix protein 2TGGAAA812.H3025H12-31200003O06RikRIKEN cDNAH3025H12Mm.142104Chromosome 3GTTCCATCTGA1200003O06CTTAAACAAAAgeneACCGTAGTTTCCAGCTCAGAATCATCCTAACATAGAAA813.J0040E05-3Stx3syntaxin 3J0040E05Mm.203928Chromosome 19GTAGGGGAATAACTAACCAAAGTAGAGGGAATTCTAAGTTTAGTAGTAAATGTGGCTTGG814.H3075F03-3ClscomplementH3075F03Mm.24128Chromosome 6GGTGTGGGACTcomponent 1, sTATGGGGTCTAsubcomponentCACAAAGGTAAAGAATTACGTGGACTGGATCCTGAAAA815.L0600G09-3BM125147ESTsL0600G09Mm.221784Chromosome 1AGGTATGACATBM125147TTTACATCCTTGAATCTTACTTACTATGTGCTAAACAATTGGCAGAAGG816.K0115H01-3KLHL6kelch-like 6K0115H01Mm.86699Chromosome 16TGCTTGTGTGAACTACCTCAGGATGAAGGGTAATGTTTAACATTCCATACATGCCTACTG817.H3015B10-2Gusbeta-H3015B10Mm.3317Chromosome 5CGATGGACCCAglucuronidaseAGATACCGACATGAGAGTAGTGTTGAGGATCAACAGTGCCCATTATTAT818.H3108A12-30910001A06RikRIKEN cDNAH3108A12Mm.22383Chromosome 15GCAGCCAAAA0910001A06TGGAAATGTTTgeneAAATTAACTGTGTTGTACAATGACCCAACACAAAACC819.H3108H90-5UNKNOWN:H3108H09Data not foundChromosome 13TTGACATGATASimilar toCATTACGCCTTHomo sapiensTGCAGTGAGCTKIAA1577AATAAGCTAACproteinATTTGTGCACA(KIAA1577),GATAAmRNA820.K0645H01-3FybFYN bindingK0645H01Mm.257567Chromosome 15TCTCAACTCATproteinCTCAGATTAGGAAGTATTTGGCAGTATTAGCATCATGTGTCCCTGTGA821.H3029A02-3ShycselectiveH3029A02Mm.12912Chromosome 7ATTTTCATGCChybridizingGAATATTCCAGcloneCAGCTATTATAAAATGCTAAATTCACTCATCCTGTACG822.K0410D10-3Cxcl12chemokine (C-K0410D10Mm.465Chromosome 6GAGAATTAATCX-C motif)ATAAACGGAAligand 12GTTTAAATGAGGATTTGGACTTTGGTAATTGTCCCTGAG823.H3118H11-3Snrpgsmall nuclearH3118H11Mm.21764Chromosome 18CATGAGCAAAribonucleoproteinGCCCACCCTCCpolypeptide GCGAGCTGAAGAAGTTTATGGACAAGAAGTTATCATTGAA824.K0517D08BM238427ESTsK0517D08Mm.222266Chromosome 19CTCTGTAAAGTBM238427CAAGTTGCATTGCATTTACAGTTAATTATGGAAAAGTCCTAAATCTGGC825.L0227G11-3Sh3d1BSH3 domainL0227G11Mm.40285Chromosome 12TTTTCAGGGCTprotein 1BATAAAAGTATTATGTGGAAATGAGGCATCAGACCACCGGACGTTACCAC826.H3134B10-36530409L22RikRIKEN cDNAH3134b10Mm.41940Chromosome MultipleAAGAAGCTGA6530409L22MappingsGGAAAAACAGgeneGAGAGTGAGAAACCGCTTTTGGAACTATGAGTTCTGCTCT827.H3115A08-3Ly6alymphocyteH3115A08Mm.263124Chromosome 15CCTGATGGAGTantigen 6CTGTGTTACTCcomplex, locusAGGAGGCAGCAAGTTATTGTGGATTCTCAAACAAGGAAA828.C0120G03-3Cskc-src tyrosinC0120G03Mm.21974Chromosome 9AGCAAATGGGkinaseCATTTTACAAGAAGTACGAATCTTATTTTTCCTGTCCTGCCCCTGGGGGT829.H3094G08-3Tigd2tiggerH3094G08Mm.25843Chromosome 6CTGCACTTGAAtransposableTGGACTGAAAelement derivedACTTGCTGGAT2TATCTAGAACAACAAGATGACATGCTAC830.NM_008362.1IIlr1interleukin 1NM_008362Mm.896Chromosome 1AGATTTCACCGreceptor, type 1TACTTTCTGATGGTGTTTTTAAAAGGCCAAGTGTTGCAAAAGTTTGCAC831.C0300E10-3Trps1trichorhinophalC0300E10Mm.30466Chromosome 15ATAAAACCACangealAAACTAGTATCsyndrome IATGCTTATAAG(human)TGCACAGTAGAAGTATAGAACTGATGGG832.L0274A03-3Ptpn2protein tyrosineL0274A03Mm.260433Chromosome 18ACCTAAATGTTphosphate,CATGACTTGAGnon-receptorACATTCTGCAtype 2GCTATAAAATTTGAACCTTTGATGTGC833.H3005H07-31810031K02RikRIKEN cDNAH3005H07Mm.145384Chromosome 4TTTATAGTTCT1810031K02AGGTTTACACCgeneAGAGAGGAGTTAATTTATCAACAGCCTAAAACTGTTGC834.H3109H12-31810009M01RikRIKEN cDNAH3109H12Mm.28385Chromosome MultipleTTCTTCCACGA1810009M01MappingsACAGATATTATgeneGTCATTTTATCCAATGCCCGATAAAGGAGAAACAACTTG835.J0008D01-3Enpp1ectonucleotideJ0008D01Mm.27254Chromosome 10TACGTGGTCTGpyrophosphatase/GGGACCTGATGphosphodiesteraseTTGGAATCCTA1TTGTTGTTAATAAAACTGAGTAAAGGA836.H3119HO5-3Mafbv-mafH3119H05Mm.233891Chromosome 10ACCAACTTCTGmusculoaponiuroticTCAAAGAACAfibrosarcomaGTAAAGAACTToncogeneGAGATACATCCfamily, proteinATCTTTGTCAAB (avian)ATAGTC837.H3048G11-3BlvrbbiliverdinH3048G11Mm.24021Chromosome 7TGACACAAATAreductase BGAGGGGTCAA(flavinTAAATTTTTAGreductaseCCAAAAGCTTC(NADPH))AAATTCTTTCAGGAAGC838.H3107D05-31110004C05RikRIKEN cDNAH3107D05Mm.14102Chromosome 7ATCACCATTGT1110004C05TAGTGTCATCAgeneTCATTGTTCTTAACGCTCAAAACCTTCACACTTAATAG839.H3006B01-3Cklfsf3chemokine-likeH3006B01Mm.292081Chromosome 8GCCGCTTTTTTfactor superGTAACCTAAAAfamily 3GGCCCCATGAATAAGGGCCCATGTTTTGGGCATTTGTA840.L0853H04-3transcribedL0853H04Mm.275315Chromosome 12CCAAGAACAAsequence withGTATAAACTTAweak similarityAGCTCTGTAGAto proteinACTGAAATTCTpir:A43932TTCAAGTCCTT(H. sapiens)TCGATCA43932 mucin2 precursor,intestinal-human(fragments)841.C0949G05-3BM221093ESTsC0949G05Mm.221696Chromosome 6AGGACATCTTGBM221093CAACTTCTATGCASATAATAAGGATTTCCATCTGACAAATAAGACAAGTG842.K0648D10-3tlr1toll-likeK0648D10Mm.33922Chromosome 5GGGGAGTTCTAreceptor 1ATAATAGTACCATTCATATCAGCAAGAACCTAAAAATGGTTCTGACTTT843.H3014E09-3BC016443cDNA sequenceH3014E09Mm.27182Chromsome 11TGCCACTAGTTBC017643CTGACTTGGGGAATATGGTCCCTTAAACATGCCAAAGTGAGCTTTTTAA844.H3022D06-3Il2rginterleukin 2H3022D06Mm.2923Chromosome XCATCAATCCTTreceptor,TGATGGAACCTgamma chainCAAAGTCCTATAGTCCTAAGTGACGCTAACCTCCCCTA845.L0201A03-32410004H05RikRIKEN cDNAL0201A03Mm.8766Chromosome 14CAGTTGGAAA241114H05AATGGATGAAgeneGCTCAATGTAGAAGAGGGATTATACAAGCAGAACTCTGGCA846.H3026E03-5Mus musculusH3026E03Mm.249306Chromosome Un: notTCAGTCAAATG2 days neonateplacedTGCATAACTGTthymus thymicAAATCAACACTcells cDNA,AAGAGCTCTGGRIKEN full-AAGGTTAAAAlength enrichedAGGTCAlibrary,clone:E430039C10product:unknownEST, fullinsert sequence.847.H3091E12-3Abhd2abhydrolaseH3091E12Mm.87337Chromosome 7AGCAGGTGTTTdomainCGGACTTGCAAcontaining 2TGAGCAATGCAATTTTTTCTAAATATGAGGATATTTAC848.H3003E01-3Cutl1cut-like 1H3003E01Mm.258225Chromosome 5CTTGCTTCTTT(Drosophila)AGCAAAATATTCTGGTTTCTAGAAGAGGAAGTCTGTCCAACAAGGCCCC849.H3016H08-5Crsp9cofactorH3016H08Mm.24159Chromosome 11TCTCAATTTTCrequired forAAGGTGTATTTSp1CCTATCAGGAAtranscriptionalACTTGAAGATAactivationATATGGTCTGAsubunit 9,ACCCA33kDa850.C0118E09-3Oas1a2′-5′C0118E09Mm.14301Chromosome 5ACTGGACAAAoligoadenylateGTATTATGACTsynthetase 1ATTCAACACCAGGAGGTCTCCAAATACCTGCACAGACAGC851.L0535B02-3Coll5a1procollagen,L0535B02Mm.233547Chromosome 4GGCTGTTGAGTtype XVGTAAAATGTGCTTTGTGTTTGCTTACAACATCAGCTTTTAGACACACAG852.L0500E02-3Sgcgsarcoglycan,L0500E02Mm.72173Chromosome 14TGAGTGCAATGgammaTGTCAGATTTC(dystrophin-ACCAAGAGATassociatedCTCCAAGGTTglycoprotein)GTAGGTAATTTGTGGTT853.H3077B08-35330431K02RikRIKEN cDNAH3077B08Mm.101992Chromosome MultipleGTCATTGTCCA5330431K02MappingsAGGTGACAGGgeneAGGAACTCAGTCGTTAAAATGACGAGCCTTATTTCATGA854.J0209G02-3Gnb4guanineJ0209G02Mm.9336Chromosome 3TCTTAGAATTnucleotideGGAATTGAGTGbinding protein,CCATATTTTCTbeta 4GTTCTCCAATGATACCTGGAGAAATCC855.C0661E01-3Lcn7lipocalin 7C0661E01Mm.15801Chromosome 4TGCTTTCTTATTCTTTAAAGATATTTATTTTTCTTCTCATTAAAATAAAACCAAAGTATT856.K0221E09-3Scml2sex, comb onK0221E09Mm.159173Chromosome XCTGCATGTTATmidleg-like 2AACTTTATATG(Drosophila)ATGGTGTAGTGCATATAAGCTATGAGAATCATTTATAC857.C0184F12-3D8Ertd594eDNA segment,C0184F12Mm.235074Chromosome 8CGTGCTGGAGGChr 8, ERATOACGAGAGATTCDoi 594,CAGAAGCTTCTexpressedGAAGCAAGCAGAGAAGCAGGCTGAACA858.L0602B03Myoz2myozenin 2L0602B03Mm.141157Chromosome 3TGGAGGCTTTGTACCCAAAACTTTTCAAGCCTGAAGGAAAAGCAGAACTGCGGGATTACA859.C0944F04-31110055E19RikRIKEN cDNAC0944F04Mm.39046Chromosome 6TGGAGGATCTG1110055E19TGTGAAAAAGgeneAAGTCACCCTCACAAACCGCCGTGCCTAAGGACTCTGTC860.L0004A03-3Gli2GL1-kruppelL0004A03Mm.12090Chromosome 1CTATTTTGTGTfamily memberAGACATCGTCTGL12TGCCTGAATAGACTGTGGGTGAATCCAAATTTGGTCCA861.L0860B03-3ESTsL0860B03Mm.221891Chromosome 5: notTAATTATCTACAV321020placedATTGGGGTAATTGAAGTAGAAAGATCCATCTTAACTACGGTAATCTCCG862.L0841F10-32310045A20RikRIKEN cDNAL0841F10Mm.235050Chromosome 5TTGGGTATCGT2310045A20TTATGTTTCCCAgeneTCATAACACATTCATAACACATGCAATAACATCTAGGAAATCTT863.L0008H10-3AgrnagrinL0008H10Mm.269006Chromosome 4TCTGATGTGGAAGTGCGGTCATTCCTGGTTTAACTCACAGCAACTTTTAATTGGTCTAAG864.C0128B02-3Casq1calsequestrin 1C0128B02Mm.12829Chromosome 1ATCTCCTGTTAATGTATTTGGGTCAAATGCAAGGCCTTAATAAAGAAATCTGGGGCAGAA865.C0645C09-3BM209340ESTsC0645C09Mm.222131No ChromosomeGCAGCAAGAGBM209340locationAAAAGAGCAAinfo availableGAGAGCCAAAGGCAAGAAATCTCTCTGTCACTCCCTTTTA866.H3082B03-3Mylkmyosin, lightH3082B03Mm.288200Chromosome 16TGAGGAAAAGpolypeptideCCCCATGTGAAkinaseACCTTATTTCTCTAAGACCATCCGTGATCTGGAAGTCGT867C0309D09-3transcribedC0309D09Mm.213420Chromosome 11ACCGGCTGTACsequence withCCAAATAGAAmoderateCGTCATTTTGAsimilarity toTATGAAGGATTproteinTCAGCCCCTGAsp:P00722 (E.AGATTTcoli)BGAL_ECOLIBeta-galatosidase(Lactase)868.H3157H09-3BG076287ESTsH3157H09Mm.131026Chromosome 2ATGGTTTCTTCBG076287CAGCAATTTAGCATTGCCTGAGGGGTCTAAAAGAATAAGTTGGTTCTTG869.H3061D03-3Pcsk5proproteinH3061D03Mm.3401Chromosome 19ACAATCTCTGTconvertaseCAGCGAAAAGsubtilisin/kexinTTCTACAACAGtype 5CTGTGCTGCAAAACATGTACATTCCAAG870.L0843D01-33732412D22RikRIKEN cDNAL0843D01Mm.18830No ChromosomeAACTGTTACTG3732412D22locationGATTGAAATTCgeneinfo availableCCATCCCCTTTCCCTAAAAATTGTGCCTTAGAAAACCC871.L0702H07-35830415L20RikRIKEN cDNAL0702H07Mm.46184Chromosome 5CGACTGAGGTT5830415L20ATGACATCCTTgeneAGACTTTGTTGTATGCTGCTTCGAATGAACCAGAGATA872.L0548G08-3XincardiacL0548G08Mm.10117Chromosome 9TGCCTCTTCATmorphogenesisCGCCAGTGGTCCAAAGGGCGCAGAGAGCGCACTAGCAGTCAATAGTGTT873.L0803E02-3Nkdlnaked cuticle 1L0803E02Mm.30219Chromosome 8CCACTAATATThomologTAGCCAGCCTT(Drosophila)CATGTAGAAGACACATGGAAACACAGAAGTAAACTTTT874.C0925G12-3Fbxo30F-box proteinC0925G12Mm.276229Chromosome 10AGAAATGAAC30ATACATTGTCAGCATTTAGAAGTAAGTTGTGAAGACAGGGACATTAAGTG875L0911A11-32010313D22RikRIKEN cDNAL0911A11Mm.260594Chromosome 5CAAACGGGAT2010313D22CCTGTCTTCTTgeneCTTTTCTAATAGAATTTTGTAAAGGAAATGAATGTAGCC876.AF084466.1rradRas-relatedAf084466Mm.29467Chromosome 8ACCGTTCTATCassociated withACTGTGGATGGdiabetesAGAAGAAGCGTCACTATTGGTCTATGACATTTGGGAAG877.H3073G09-31600029N02RikRIKEN cDNAH0373G09Mm.154121Chromosome 7CTATTTTTGGG1600029N02AGATGTCTATTgeneGCGGAGTACAGTAATATATACCCAGAGTATGTCTATAG878.L0815B08-31100001D19RikRIKEN cDNAL0815B08Mm.260515Chromosome XACCCAACTCCA110001D19GTGCTCTCTGTgeneCTTTTAGTACAGGATTTTCACCCATGTGCATGAAAAAT879.J1037H05-3D230016N13RikRIKEN cDNAJ1037H05Mm.21685Chromosome 13 TTACCATTTTTD230016N13GGTTAAATGGCgeneCAAATTCAGAAAATAACTCCATTTGAATCTCCAGCAGG880.K0421F09-3transcribedK0421F09Mm.222196Chromosome 6TCACCATACTTsequence withTGAAAGTGTAAweak similarityACTACCACATAto proteinTTAACATGTGTref:NP_081764.1GATTTAAGACC(M. musculus)CTCAGRIKEN cDNA5730493B19[Mus musculus]881.H3082E06-31110003B01RikRIKEN cDNAH3082E06Mm.275648Chromosome 13TGTTGCCCTCA1110003B01GATATGTCAGAgeneTCAACTTGGAAGGAAAGACCTTCTACTCCAAGAAGGAC882.C0935B04-3HhipHedgehog-C0935B04Mm.254493Chromosome 8TCTAACAAGTGinteractingTATTTGTGTTAproteinTCTTTAAAATAGAACAATTGTATCTTGAAATGGTAAAT883.H3116B02-31110007C05RikRIKEN cDNAH3116B02Mm.27571Chromosome 7CGACACTGGGT1110007C05GGCCCTGCGACgeneAGGTAGATGGCATCTACTATAATCTGGACTCAAAGCTG884.C0945G10-3Tp53il1tumor proteinC0945G10Mm.41033Chromosome 2TCTCAGAGGTGp53 inducibleTTGAAGATTTAprotein 11TCATCTTGAATCCTCCACAAATACAGATACAGTCCCAA885.K0440609-3Tgfb3transformingK0440G09Mm.3992Chromosome 12TCTTTTCACCTgrowth factorCGATCAGCATCbeta 3ATGAGTCATCACAGATCATGTAATTAGTTTCTGGGCCA886.L0916G12-3BM118833ESTsL0916G12Mm.221415Chromosome 6TGGGAATTGCABM118833TTTAGGATAGAATTGTATCTGATTTGCAAAATCCATAAGCTCTCATGCC887.L0505A04-3Dnajb5DnaJ (Hsp40)L0505A04Mm.20437Chromosome 4TACTCCCACAGhomologTTGTATAGAAGsubfamily B,TCGAATAGTGAmember 5AGGAGCTGGGAGAAAACTGCTTCAGCT888.L0542E08-3Usmg4unpregulatedL0542E08Mm.27881Chromosome 3CCGCACTTAGCduring skeletalCTAGACCTTTmuscle growth 4CTTACATGATCTCAAGTTGAACCGACTTCCTTAACTCT889.L0223E12-3SparcllSPARC-like 1L0223E12Mm.29027Chromosome 5GCTTTGGAATT(mast9, hevin)AAAGAGGAGGATATAGATGAAAACCCCCTCTTTTGAATTAAGATTTGAG890.K0349C07-34631423F02RikRIKEN cDNAK0349C07Mm.68617Chromosome 1AAATCAGATAT4631423F02GCAGGTCATCTgeneGATAAATGAGTTAATGTTTGATATTCGGGGTATCTCAC891.C0302A11-3EST B1988881C0302A11Mm.260261No ChromosomeGAACCATATGClocationTGGAATGAAAinfo availableCATAAGAGTTTTCAACAGTTATCCTCTCACCTCTGTATG892.C0930C11-3Fgfl3fibroblastC0930C11Mm.7995Chromosome XGTATCGTCAATgrowth factorCCCAGTCAGTA13AGATAAGTTGAAACAAGATTATCCTCAAGTGTAGATTT893.H3022A11-3Cald1caldesmon 1H3022A11Mm.130433Chromosome 6GTCAAAAACGCCTTCAGGAAGCCTTAGAGCGTCAGAAGGAGTTTGATCCGACCATAACAG894.C0660B06-3Csrp1cysteine andC0660B06Mm.196484Chromosome 1AATAGAATCTTglycine-richTTCACTTAGGAprotein1ATGGAGAACAAGCCAGTTCAGAGGACCCCAAAGTCTAG895.L0949F12-3Heylhairy/enhancer-L0949F12Mm.103615Chromosome 4CGTGGAGGATof-split relatedGGGCTAGCCTGwith YRPWAGCTCTGGGACmotif-likeTAATCTTTATTACATACTTGTTAATGAG896.K0225B06-3Unc5cunc-5 homologK0225B06Mm.24430Chromosome 3CTTATAGGGAGC (C. elgans)AATGTTCTATTCCTCAATCCATACTCATTCCTACAGTATGCGCTCTGGA897.K0541E04-3Herc3hect domainK0541E04Mm.33788Chromosome 6AGCAGGGGGAand RLD 3TTATGTTAAGTCAAATGCGTGTGTCTCAAAAGTGACATGTTTAACTGCTC898.C0151A03-3BC026744cDNA sequenceC0151A03Mm.4079Chromosome 5ACTCTGTACCCBC026744TACTGGAACCACTCTGTAAAGAGACAAAGCTGTATGTGCCACTTCAGTA899.L0045C07-36-Sepseptin 6L0045C07Mm.258618Chromosome XTTACAGGTCACTGTTTGTCACTTTTGTGTACCAGCTTCCCCATTAGAATTCAACCGATAC900.L0509E03-3Ryr2ryanodineL0509E03Mm.195900Chromosome 13ATGGAAGCGAreceptor 2,GGTCATTCTGCcardiacGAACATTGGAGATCTTTTATTACAAGTCTGCTTGTTAAT901.H3049B08-3Testetis derivedH3049B08Mm.271829Chromosome 6TAAAATTAGTGtranscriptTCCTGGGAGAGATGACCATTTTAACTTCTATGCTTATTTCACATGGGAA902.L0533C09-3BM123974ESTsL0533C09Mm.213265Chromosome 14TCGACGTCAABM123974CTTACCTCTCTAGGCAACATGTTATCCCCGGATGATCAGAAATTCCCAA903.H3108C01-34930444A02RikRIKEN cDNAH3108C01Mm.17631Chromosome 8ACCTGTGTTTT4930444A02GTTTTTGTTTTgeneAAGAAACCAAAGTGCACCAAGATAGCATGCTCTTGAGA904.C0110C06-3Epb4.111erythrocyteC0110C06Mm.20852Chromosome 2CTGCAGGTAACprotein bandTCTCATTGGAA4.1-like 1GAAAAAGAAACTACAAGAGCAAACAGAAGCCATGGGAA905.C032H08-3EnahenabledC0324H08Mm.87759Chromosome MultipleAAAGATTTCAThomologMappingsCCACGTCTGGC(Drosophila)GTAGTGGAAAACCCGAAGGGAATATGTAATGATCTTTC906.C0917A09-3ESTsC0917A09Mm.242207No ChromosomeGTGTTGTACCCBB231855locationTAATTTGAATTinfo availableTAAAGTAGGCAGTAGGTAGGGTTAATTGGTAGACTATC907.L0854B10-3Anks1ankyrin repeatL0854B10Mm.32556Chromosome 17CTTGGGTTTGAand SAMGCACTCAGAACdomainACATGGCTGCAcontaining 1ATCATCAAGACAGTTCACAGTTAGCTT908.K0326D08-3Ly75lymphocyteK0326D08Mm.2074Chromosome 2CCCTAAGACAAantigen 75TGAAACTCAGAACTCTGTGATTCCTGTGGAAATATTTAAAACTGAAATG909.H3074H01-3C430017H16hypotheticalH3074H01Mm.268854Chromosome 3ATTTATAGAGGproteinTATCCTTAACAC430017H16TGCTGACTTCAGTAACTGCCCTTGTTTCTAAGGAAGTC910.H3131D02-3Tnk2tyrosine kinase,H3131D02Mm.1483Chromosome 16ACCTGTAGCTTnon-receptorCACTGTGAACTTGTGGGCTTGGCTGGTCTTAGGAACTTGTACCTATAAA911.C0112B03-3Heylhairy/enhancer-C0112B03Mm.103615Chromosome 4TAATCCCTGGCof-split relatedAAAGTCAAGAwith YRPWCTGTGGGAAACmotif-likeTAGAACTGGTTACTCACTACTGCTGGTA912.L0514A09-36430511F03hypotheticalL0514A09Mm.19738Chromosome XTTAGTCCCATGproteinACCCCAAGGTT6430511F03AAGGTTCTGCCAACAAGCATTCTGCCTGACATCTACTT913.C0234D07-3Fbxo30F-box proteinC0234D07Mm.276229Chromosome 10AATAAAGGCC30CCTTAGAAGCTACTGTAAGCTCTTCAAAGTTTTCATGTAATCATAGGCA914.H3152A02-3St6ga11beta galactosideh3152A02Mm.149029Chromosome 16AGAGATGGAGalpha 2, 6ACTACACTGGGsialyltransferaseTAGATTCTAGT1TTTTAGTTCTTATTAATGTGGGGGAGTA915.H3075C04-3Ches1checkpointH3075C04Mm.268534Chromosome 12TATGGCCATTTsuppressor 1GGTTTCAGCATGTCAGGAGATTTCTAATGATTTGATGGCAATATCAGCAA916.L0600E02-3BM125123ESTsL0600E02Mm.221782Chromosome 19TGTGTCAAGATBM125123AATCCTGAGTCAACCTGGACACTTAATCCCTTTGGACCTCTATCTGGAG917.K0501F10-3BM237456ESTsK0501F10Mm.34527Chromosome XCCACCCATTAABM237456AATGACAGTACAAGTAGACCACAGTTTAAATAGTTAGTCTAATTCTAC918.K0301H08-3Oxct3-oxoacid CoAK0301H08Mm.13445Chromosome 15CATAGTGGAAtransferaseATATGCTCATCTTTTATGCTATATGTATTAAACCTCGACTTAGCCCTGAA919.L0229E07-3LuLutheran bloodL0229E07Mm.29236Chromosome 7GTTGAGGCTGAgroupCGACCTCCCAG(Auberger bAGGCAATCTCTantigenGGATCTGGAACincluded)TTTGGGCATCATCGGA920.H3077C06-34931430I01RikRIKEN cDNAH3077C06Mm.12454Chromosome 1ACCAACCAGG4931430I01GACTAGTTTGAgeneTGCTATCTTTGCCTGTCTCTTGGCTCTTAACAATGCCTA921.J0807D02-3Mus musculusJ0807D02Mm.125975Chromosome 7CCAGGGAAGG10 days neonateAACGATCCATTcerebellumCAGTGGTTTTAcDNA, RIKENAAATATCTCTTfull-lengthCCTCAACAGAAenrichedAAAGATlibrary,clone:B930022I23product:unclassifiable, fullinsert sequence.922.H3118G11-3C130068N17hypotheticalH3118G11Mm.138073Chromosome 2GGTGCAAGCTAproteinGTACTCACACTC130068N17GTCACACCTTTACGCATGCGAAAGGTAATGTGCTAAAT923.L0818F01-3Smarcd3SWI/SNFL0818F01Mm.140672ChromosomeAGATCAGTGCTrelated, matrixCTGGACAGTAAassociated,GATCCATGAGAactin dependentCGATTGAGTCCregulator ofATAAACCAGCTchromatin,CAAGAsubfamily d,member 3924.C0359A10-3BM198389ESTsC0359A10Mm.218312Chromosome 1ATACCCTGCTBM198389AACTTAACAGCAGTTAGTTTCCTTGTTATGAATAAAAATGACAGTCTGG925.G0108E12-31190009E20RikRIKEN cDNAG0108E12Mm.260102AAAGCAAATG1190009E20TTAGTAAAAAGgeneCTGGTGTGCATAGTCTTGTTACATTGATGCAGTTTTTCC926C0941C09-3Gja7gap, junctionC0941C09Mm.3096Chromosome 11CAACTTGCTGAmembraneATAATGACTTCchannel proteinCATTGAGTAAAalpha 7CATTTGGCTCTGGTTATCTTCAGGGAT927.H3111BO305UNKNOWNH3111B03Data not foundNo ChromosomeAGGAATTAGTAH3111B03locationACGTTTCATCCinfo availableAAGTAACCTTGTTACAGTGAACAAGTGTCAAGTGCTCA


The following Examples are intended to illustrate, but not limit, the invention.


EXAMPLES
Example 1
Signature Patterns of Gene Expression in Mouse Atherosclerosis and their Correlation to Human Coronary Disease

Mouse genetic models of atherosclerosis allow systematic analysis of gene expression, and provide a good representation of the human disease process (Breslow (1996) Science 272: 685-688). ApoE-deficient mice predictably develop spontaneous atherosclerotic plaques with numerous features similar to human lesions (Nakashima et al. (1994) Arterioscler Thromb 14: 133-140; Napoli et al. (2000) Nutr Metab Cardiovasc Dis 10: 209-215; Reddick et al. (1994) Arterioscler Thromb 14: 141-147. On a high-fat diet, the rate and extent of progression of lesions are accelerated. In addition to environmental influences such as diet, the genetic background of mice has also been found to have an important role in disease development and progression. Whereas C57B1/6 (C57) mice are susceptible to developing atherosclerosis, the C3H/HeJ (C3H) strain of mice is resistant (Grimsditch et al. (2000) Atherosclerosis 151:389-397. Previously, genetic-based diet and age induced transcriptional differences have been demonstrated between these two strains (Tabibiazar et L. (2005) Arterioscler Thromb Vasc Biol 25:302-308.


To more fully characterize the vascular wall gene expression patterns that are associated with atherosclerosis, a systematic large scale transcriptional profiling study was undertaken to take advantage of a longitudinal experimental design, and mouse genetic model and diet combinations that provide varying susceptibility to atherosclerosis. In this experiment, atherosclerosis-associated genes were studied independent of other variables. Primarily, these studies investigated differential gene expression over time in apoE-deficient mice on an atherogenic diet, with comparison to apoe-deficient mice (C57BL/6J-ApoetmlUnc) on normal diet as well as C57B1/6 and C3H/HeJ mice on both normal chow and atherogenic diet. Identification of atherosclerosis-associated genes was facilitated by development of permutation-based statistical tools for microarray analysis which takes advantage of the statistical power of time-course experimental design and multiple biological and technical replicates. Using these tools, hundreds of known and novel genes that are involved in all stages of atherosclerotic plaque, from fatty streak to end stage lesions, were identified. To further examine the expression of individual genes in the context of particular biological or molecular pathways, a pathway enrichment methodology with gene ontology (GO) terms for functional annotation was utilized. Using classification algorithms, a signature pattern of expression for a core group of mouse atherosclerosis genes was identified, and the significance of these classifier genes was validated with additional mouse and human atherosclerosis samples. These studies identified atherosclerosis related genes and molecular pathways.


Methods


Atherosclerotic Lesion Analysis


For select time points for various experimental groups, 5 to 7 female mice were used for histological lesion analysis. Atherosclerosis lesion area was determined as described previously (Tabibiazar et al. (2005), supra). Briefly, the arterial tree was perfused with PBS (pH 7.3) and then perfusion-fixed with phosphate-buffered paraformaldehyde (3%, pH 7.3). The heart and full length of the aorta to iliac bifurcation was exposed and dissected carefully from any surrounding tissues. Aortas were then opened along the ventral midline and dissected free of the animal and pinned out flat, intimal side up, onto black wax. Aortic images were captured with a Polaroid digital camera (DMC1) mounted on a Leica MZ6 stereo microscope, and analyzed using Fovea Pro (Reindeer Graphics, Inc. P. O. Box 2281, Asheville, N.C. 28802). Percent lesion area was calculated as total lesion area/total surface area.


Experimental Design, RNA Preparation and Hybridization to Microarrays


All experiments were performed following Stanford University animal care guidelines (Saadeddin et al. (2002) Med Sci Monit 8:RA5-12). Three week old female apoE knock-out mice (C57BL/6J-ApoetmlUnc), C57Bl/6J, and C3H/HeJ mice were purchased from Jackson Labs (Bar Harbor, Me.). At four weeks of age the mice were either continued on normal chow or were fed high fat diet which included 21% anhydrous milkfat and 0.15% cholesterol (Dyets #101511, Dyets Inc., Bethlehem, Pa.) for maximum period of 40 weeks. At each of the time-points, including 0 (baseline), 4, 10, 24 and 40 weeks, for each of the conditions (strain-diet combination), 15 mice (3 pools of 5) were harvested for RNA isolation (total of 405 mice). Additional mice were used for histology for quantification of atherosclerotic lesions as described above. A separate cohort of sixteen-week-old apoE-deficient mice on high fat diet for two weeks (4 pools of 3 aortas) was also used for classification purposes.


After perfusion of mice with saline, the aortas were carefully dissected in their entireties from the aortic root to the common iliac and subsequently were flash frozen in liquid nitrogen. Total RNA was isolated as described previously (Tabibiazar et al. (2003) Circ Res 93:1192-1201) using a modified two-step purification protocol. RNA integrity was also assessed using the Agilent 2100 Bioanalyzer System with RNA 6000 Pico LabChip Kit (Agilent).


First strand cDNA was synthesized from 10 μg of total RNA from each pool and from a whole 17.5-day embryo for reference RNA in the presence of Cy5 or Cy3 dCTP, respectively. Hybridization to a mouse 60mer oligo microarray (G4120A, Agilent Technologies, Palo Alto, Calif.) (Carter et al. (2003) Genome Res 13:1011-1021) was performed following manufacture's instructions, generating three biological replicates for each of the time points. The RNA from the group of sixteen-week-old mice was linearly amplified and hybridized to a different array (G4121A, Agilent Technologies). Technical validation of the microarray has been performed previously using quantitative real-time reverse transcriptase polymerase chain reaction (results reported in Tabibiazar et al. (2005), supra). Primers and probes for 10 representative differentially expressed genes were obtained from Applied Biosystems Assays-on-Demand. A total of 90 reactions, including triplicate assays on three pools of five aortas, was performed from representative RNA samples used for microarray experiments, demonstrating a high correlation between the two platforms (Pearson correlation of 0.82).


Data Processing


Image acquisition of the mouse oligo microarrays was performed on an Agilent G2565AA Microarray Scanner System and feature extraction was performed with Agilent feature extraction software (version A.6. 1.1, Agilent Technologies). Normalization was carried out using a LOWESS algorithm. Dye-normalized signals of Cy3 and Cy5 channels were used in calculating log ratios. Features with reference values of <2.5 standard deviation for the negative control features were regarded as missing values. Those features with values in at least ⅔ of the experiments and present in at least one of the replicates were retained for further analysis. Reproducibility of microarray results, as measured by the variation between arrays for signal intensities, was assessed using box plots (GeneData,Inc., South San Francisco, Calif.). For further statistical analysis of the data, a K-nearest-neighbor (KNN) algorithm was applied to impute missing values (Troyansakaya et al. (2001) Bioinformatics 17:520-525). Numerical raw data were then migrated into an Oracle relational database (CoBi) that has been designed specifically for microarray data analysis (GeneData, Inc.). Heat maps were generated using “HeatMap Builder” software (Blake and Ridker (2002) J Intern Med 252:283-294). All microarray data were submitted to the National Center for Biotechnology information's Gene Expression Omnibus (GEO GSE1560; www.ncbi.nlm.nih.gov/geo/).


Data Analysis




  • i) Principal components analysis



For each gene the average log expression values were computed at the four post-baseline observation times, 4, 10, 24, and 40 weeks. This was done separately for the six different (diet, strain) combinations, for example ApoE on high fat, presumably the most atherogenic combination. Differences of these vectors were taken for various interesting contrasts, e.g., for ApoE, high-fat minus C3H, normal chow, giving N=20280 vectors of length 4, one for each gene. Principal components analysis of the N vectors showed a consistent pattern, with the first principal vector indicating a roughly linear increase with observation time.

  • ii) Time course regression analysis


A standard ANACOVA model was fit separately to the log expression values for each gene, using a model incorporating strain, diet, and time period effects. A single important “z value” was extracted from each ANACOVA analysis, for example corresponding to the significance of the time slope difference between the ApoE, high-fat combination and the average of the other five combinations. The N z-values were then analyzed simultaneously, using empirical Bayes false discovery rate methods described previously (Efron (2004) J Amer Stat Assoc 99:82-95; Efron and Tibshirani (2002) Genetic Epidemiology 23:70-86; Efron et al. (2001) J Amer Stat Assoc 96:1151-1160. These analyses identified a set of several hundred genes clearly associated with atherosclerosis progression.

  • iii) Time course area under the curve analysis



Area under the curve (AUC) analysis was employed as described previously (Tabibiazar et al. (2005), supra). For each sequence of 4 triplicate gene expression measurements over time, the measurement at time 0 was subtracted from all values. The signed area under the curve was then computed. The area is a natural measure of change over time. These areas were then used to compute an F-statistic for the 6 groups (3 mouse strains and 2 diets) and 3 replicates (between sum of squares/within sum of squares). A permutation analysis, similar to that employed in Significance Analysis of Microarrays (SAM) (Tusher et al. Proc Natl Acad Sci 98:5116-5121), was carried out to estimate the false discovery rate (q-value or “FDR”) for different levels of the F-statistic.

  • iv) Enrichment analysis


For enrichment analysis, the Expressionist software (GeneData, Inc.), which employs the Fisher exact test to derive biological themes within particular gene sets defined by functional annotation with Gene Ontology (GO) terms (www.geneontology.org) and Biocarta pathways (www.biocarta.com/genes/allpathways.asp), was used. In this way, over-representation of a particular annotation term corresponding to a group of genes was quantified.

  • v) Support vector machine for gene selection


For supervised analyses, the Expressionist software (GeneData USA), which employs Support Vector Machine (SVM) algorithm (Burges (1998) Data Mining and Knowledge Discovery 2:121-167),was used to rank genes based on their utility for class discrimination between time points 0, 4, 10, 24, and 40 weeks in apoE mice on high-fat diet. SVM is a binary classifier, so in order to classify multiple categories, N classifiers were created that classify one group vs. a combination of the rest of the groups (“one vs. all” classifiers) (Ramaswamy et al. (2001) Proc Natl Acad Sci 98:15149-15154). The larger set of genes identified by the time-course analysis was used for this analysis. This method was then used to determine the optimal number of ranked genes to classify the experiments into their correct groups at minimal error rate. The optimal error rate or misclassification is calculated by cross-validation with 25% of the experiments as the test group and the rest as the training group. This is reiterated 1000 times (FIG. 5A). In this study, a linear Kernel was used, since a nonlinear Gaussian kernel yielded similar results. This minimal subset of classifier genes was then used for cross-validation as well as classification of other independent gene expression profiling datasets.

  • vi) Analysis of independent datasets.


The SVM algorithm was utilized for classification of independent groups of experiments (Yeang et al. (2001) Bioinformatics 17 Suppl 1:S316-322). In this analysis, the primary time-course experiments were used (corresponding to 5 time points mentioned above) as the training set and the independent set of experiments (different array and labeling methodology) as the test set. SVM output for each experiment based on one-versus-all comparisons was represented graphically in a heatmap format (FIG. 5B), which is the normalized margin value for each of the 5 SVM classifiers mentioned above. The SVM output permits classification of a new experiment according to the 5 SVM hyperplane. The SVM algorithm (Linear Kernel) was also utilized for external validation by classifying different sets of human expression data. In these analyses, a confusion matrix was generated using cross validation with repeated splits into 75% training and 25% test sets to determine the accuracy of classification based on the small subset of genes identified earlier. Results are represented in tabular fashion (Table 3).


Transcriptional Profiling of Human Atherosclerotic Tissue and Atherectomy Samples


For one set of samples, coronary arteries were dissected from explanted hearts of patients undergoing orthotopic heart transplantation. Arteries were divided into 1.5 cm segments, classified as lesion or non-lesion after inspection of the luminal surface under a dissecting microscope. RNA was isolated from each individual sample and hybridized to a microarray. A central portion (1-2mm) of each segment was removed and stored in OCT for later histological staining (hematoxylin and eosin, Masson's trichrome). Samples (n=40) were derived from 17 patients (male 13, female 4, mean age 43 years). Six patients had a diagnosis of ischemic cardiomyopathy, while 11 were classified as non-ischemic, although some vessel segments from the latter had microscopic evidence of coronary artery disease. Of 21 diseased segments, 7 were classified as grade I, 4 grade III and 9 grade V, according to the modified American Heart Association criteria (Virmani et al. (2000) Arterioscler Thromb Vasc Biol 20:1262-1275), and one sample had only macroscopic information available. For a second set of tissues, coronary atherectomy samples were obtained with a cutting atherectomy catheter system (Fox Hollow Inc., Redwood City, Calif.), for chronic atherosclerosis lesions (n=28) and in-stent restonsis lesions (n=14). Patient characteristics in both groups were similar (male 78% vs. 71%, mean age 64 vs. 67). RNA was isolated from each individual sample, labeled by direct or linear amplification methods, and hybridized as described above to a 22k feature custom cardiovascular oligonucleotide microarray designed in conjunction with Agilent Technologies (G2509A, Agilent Inc., Palo Alto, Calif.). Common reference RNA for all human hybridizations was a mixture of 80% HeLa cell RNA and 20% human umbilical vein endothelial cell RNA. Data processing and analysis were performed as described above. For 2-class comparison of gene expression, Significance Analysis of Microarrays (SAM) was used (www-stat.stanford.edu/tibs/SAM/; Tabibiazar et al. (2003), supra; Tusher et al. (2002), supra).


Results and Discussion


Atherosclerosis in the Genetic Models


To correlate the gene expression results with the extent of disease in each experimental group, the total atherosclerotic plaque burden in the aorta was determined by calculating a percent lesion area from the ratio of atherosclerotic area to total surface area. ApoE-deficient mice (C57BL/6J-ApoetmlUnc) (n=7) on high-fat diet were compared to other control mice (n=5-7 for each mouse-diet combination). Representative time-intervals were used for analysis, including baseline measurements in mice prior to initiation of high-fat diet at 4 weeks and end-point measurements corresponding to 40 weeks on either high-fat or normal diet (FIGS. 1, 2). Gross histological evaluation of these mice demonstrated increased atherosclerotic lesions in ApoE-deficient mice on high-fat diet involving about 50% of the entire aorta, and lesser area involved in ApoE-deficient mice on normal diet (FIG. 2). As expected, the control mice on either diet did not demonstrate evidence of atherosclerosis throughout the course of the experiment (Jawien et al. (2004) J Physiol Pharmacol 55:503-517; Nishina et al. (1990) J Lipid Res 31:859-869). Although some fatty infiltrates were noted on histological evaluation of the aortic root in C57 mice on high-fat diet, there were no obvious changes in inflammatory cell infiltrate (Tabibiazar et al. (2005), supra). The metabolic and lipid profiles of these mice were not obtained in this study, since they are well described in the literature (Grimsditch et al., supra; Nishina et al. (1990), supra; Nishina et al. (1993) Lipids 28:599-605).


Temporal Patterns of Gene Expression


Employing a number of mouse models with different propensity to develop atherosclerosis, two different diets, and a longitudinal experimental design, it was possible to factor out differentially regulated genes that are unlikely to be related to the vascular disease process in the apoE deficient model. For instance, age-related and diet-related gene expression patterns that are not linked to vascular disease were eliminated by virtue of their expression in the genetic models that did not develop atherosclerosis. However, the complexity of the experimental design provided significant difficulties related to statistical analysis. Although analytic methods have been proposed to address a single set of time-course microarray data (Luan and Li (2003) Bioinformatics 19:474-482; Park et al. (2003) Bioinformatics 19:694-703; Peddada et al. (2003) Bioinformatics 19:834-841; Xu and Li (2003) Bioinformatics 19:1284-1289), there was no accepted algorithm for comparing differences in patterns of gene expression across multiple longitudinal datasets.


Using principle component analysis, it was determined that the greatest variation in the data was between time points, correlating with the progression of disease described previously for the apoE knockout mouse on high fat diet (Nakashima et al. (1994) Arterioscler Thromb 14:133-140; Reddick et al. (1994) Arterioscler Thromb 14:141-147). Given this finding, a linear regression model was utilized to identify genes that were differentially expressed in ApoE-deficient mice on high-fat diet, compared with all other experimental groups across time. This comparison across strains and dietary groups was employed to focus the analysis on atherosclerosis-specific genes, taking into account gene expression changes in the vessel wall associated with aging, diet, and genetic background. Empirical Bayes and permutation methods were employed to derive a false discovery rate (FDR) and minimize false detection due to multiple testing. With high stringency limits, global FDR<0.05 and local FDR<0.3, 667 genes demonstrated a linear increase with time, whereas only 64 genes showed the opposite profile (FIG. 3).


Genes with Increased Expression in the Atherosclerotic Vessel Wall


The identification of known genes previously linked to atherosclerosis validated the methodology and analysis algorithm. Most striking in this regard were inflammatory genes, including chemokines and chemokine receptors, such as Ccl2, Ccl9, CCr2, CCr5, Cklfsf7, Cxcl1, Cxcl12, Cxcl16, and Cxcr4 (FIG. 3). Also upregulated were interleukin receptor genes, including IL1r, IL2rg, IL4ra, IL7r, IL10ra, IL13ra, and IL15ra, and major histocompatibility complex (MHC) molecules such as H2-EB1 and H2-Ab. The value of transcriptional profiling in this disease was demonstrated by the identification of numerous inflammatory genes not previously linked to atherosclerosis, including CD38, Fcer1g, oncostatin M (Osm) and its receptor (Osmr).


Oncostatin M (Osm) and its cognate receptor (Osmr) are likely to have significant roles in atherosclerosis, based on number of studies that suggest several important related functions for these genes (Mirshahi et al. (2002) Blood Coagul Fibrinolysis 13:449-455. Osm is a member of a cytokine family that regulates production of other cytokines by endothelial cells, including Il6, G-CSF and GM-CSF. Osm also induces Mmp3 and Timp3 gene expression via JAK/STAT signaling (Li et al. (2001) J Immunol 166:3491-3498). It induces cyclooxygenase-2 expression in human vascular smooth muscle cells (Bernard et al. (1999) Circ Res 85:1124-1131), as well as Abcal in HepG2 cells (Langmann et al. (2002) J Biol Chem 277:14443-14450). Interestingly, Stat1, Jak3, Cox2, and Abca1 were among the disease-associated upregulated genes. Additionally, Osm produced by macrophages may contribute to development of vascular calcification (Shioi et al. (2002) Circ Res91:9-16). This may occur via regulation of osteopontin or osteoprotegerin (Palmqvist et al. (2002) J Immunol 169:3353-3362, both of which have demonstrated significant changes in the dataset described herein. Osteopontin (Spp1) is thought to mediate type-1 immune responses (Ashkar et al. (2000) Science 287:860-864. While Spp1 has been extensively studied in atherosclerosis and other immune diseases, some of the osteopontin-related genes identified through these studies are novel and provide additional links between inflammation and calcification. Some of these include Cd44, Hgf; osteoprotegerin, Mglap, Il10ra, Infgr, Runx2, and Ccnd1. Ibsp, (sialoprotein II), was also noted to be upregulated in these studies. Despite its similar expression profile to Spp1 in various cancer types and its binding to the same alpha-v/beta-3 integrin, the role of Ibsp in atherosclerosis has not been elucidated.


Known and novel genes were identified for many other protein classes that have been studied in atherosclerosis. Genes encoding endothelial cell adhesion molecules were among these groups, including Alcam and Vcam1. Extracellular matrix and matrix remodeling proteins were found to be upregulated, including fibronectin, Col8al, Ibsp, Igsf4, Itga6, and thrombospondin-1. Matrix metalloproteinase genes such as Mmp2 and Mmp14 as well as those encoding tissue inhibitors of metalloproteinases, including Timp1, were also among the upregulated genes. Many transcription factors, lipid metabolism and vascular calcification genes, as well as macrophage and smooth muscle cell specific genes, were among those found to be upregulated. New genes were identified in each of these classes, for example, members of the ATP-binding-cassette family that were not previously associated with atherosclerosis were identified through these studies, including Abcc3 and Abcb1b.


Interesting genes linked to atherosclerosis for the first time through these studies encode a variety of functional classes of proteins. For example, genes encoding transcription factors Runx2 and Runx3 were linked to atherosclerosis in these studies. Cytoplasmic signaling molecules Vav1, Hras1, and Kras2 are factors that are well known to have critical signaling functions, but their role in atherosclerosis has not yet been defined. Wispl is a secreted wnt-stimulated cysteine-rich protein that is a member of a family of factors with oncogenic and angiogenic activity. Rgs10 is a member of a family of cytoplasmic factors that regulate signaling through Toll-like receptors and chemokine receptors in immune cells. Among the new classes of genes identified through these studies to be upregulated in atherosclerosis were those encoding histone deacetylases. Among those genes identified were Hdac7and Hdac2. Although there is significant evidence that HDACs have important functions regulating growth, differentiation and inflammation, these molecules have not been well studied in the context of atherosclerosis (Dressel et al. (2001) J Biol Chem 276:17007-17013); Ito et al. (2002) Proc Natl Acad Sci 99:8921-8926). Histone deacetylase inhibitors have been postulated to modulate inflammatory responses (Suuronen et al. (2003) Neurochem 87:407-416).


The data from the experiments described herein has also yielded numerous ESTs and uncharacterized genes. These genes may be attractive candidates for further characterization. One example of such ESTs is 2510004L01Rik, a gene termed “viral hemorrhagic septicemia virus induced gene” (VHSV), which was originally cloned from interferon-stimulated macrophages. This gene is enriched in bone marrow macrophages, is upregulated by CMV infection and is similar to human inflammatory response protein 6 (Chin and Cresswell (2001) Proc Natl Acad Sci 98:15125-15130). Several ESTs such as 5930412E23Rik and 2700094L05Rik have been cloned from hematopoietic stem cells (genome-www5.stanford.edu/cgi-bin/source/sourceSearch), consistent with data suggesting cells in the diseased vessel wall may emanate from the bone marrow (Rauscher et al. (2003) Circulation 108:457-463.


Genes with Decreased Expression in the Atherosclerotic Vessel Wall


The 64 genes that showed decreased expression during progression of atherosclerosis were of interest, given the lack of previous attention to such genes. Sparcl1 (Hevin) is an extracellular matrix protein which is downregulated in the dataset described herein, and may have antiadhesive (Girard and Springer (1996) J Biol Chem 271:4511-4517) and antiproliferative (Claeskens et al. (2000) Br J Cancer 82:1123-1130) properties. It has been shown to be downregulated in neointimal formation and suggested to have a possible protective effect in the vessel wall (Geary et al. (2002) Arterioscler Thromb Vasc Biol 22:2010-2016). Another gene with decreased expression, Tgfb3, may also have a protective effect. The factor encoded by this gene has been shown to decrease scar formation, and to exert an inhibitory effect on G-CSF, suggesting an anti-inflammatory role that would counter pro-inflammatory factors in the vascular wall (Hosokawa et al. (2003) J Dent Res 82:558-564); Jacobsen et al. (1993) JImmunol 151:4534-4544).


Interestingly, numerous genes characteristic of various muscle lineages were shown to be downregulated. For smooth muscle cells, this might reflect decreased expression of differentiation markers. For example, the smooth muscle cell gene caldesmon encodes a marker of differentiated smooth muscle cells (Sobue et al. (1999) Mol Cell Biochem 190:105-118), and previous studies have noted that the population of differentiated contractile smooth muscle cells that express caldesmon is relatively lower in atherosclerotic plaque (Glukhova et al. (1988) Proc Natl Acad Sci 85:9542-9546). Other potential smooth muscle cell marker genes with decreased expression included Csrp1 and Mylk. Other downregulated skeletal and cardiac muscle genes included calsequesterin, which is expressed in fast-twitch skeletal muscle, Usmg4, which is upregulated during skeletal muscle growth, Xin, which is related to cardiac and skeletal muscle development, and Sgcg, that is strongly expressed in skeletal and heart muscle as well as proliferating myoblasts. The possible association of these and other myocyte related genes identified in this study to normal vascular function is not known.


Pathways Analysis


To identify important biological themes represented by genes differentially expressed in the atherosclerotic lesions, the genes were functionally annotated using Gene Ontology (GO) terms (www.geneontology.org) and curated pathway information. Enrichment analysis with the Fisher Exact Test demonstrated several statistically significant ontologies (Table 3), including several associated with inflammation. Inflammatory processes such as immune response, chemotaxis, defense response, antigen processing, inflammatory response, as well as molecular functions such as interleukin receptor activity, cytokine activity, cytokine binding, chemokine and chemokine receptor activity, Tnf-receptor, and MHC I and II receptor activity were noted to be significantly over-represented in the group of genes upregulated with atherosclerosis. Subanalysis of the inflammatory response pathways revealed genes characteristic of the macrophage lineage, as well as both the TH-1 and TH-2 T-cell populations, to be over-represented. Biocarta terms further delineated novel genes that were associated with pathways within the inflammation category, including classical complement, Rac-CyclinD, Egf, and Mrp pathways, as well as those known to be differentially regulated in atherosclerosis, such as Il2, Il7, Il22, Cxcr4, CCr3, Ccr5, Fcer1, and Infg pathways.


In addition to inflammation, other biological processes and molecular functions were over-represented in the group of differentially upregulated genes. These included expected pathways such as wound healing, ossification, proteo- and peptidolysis, apoptosis, nitric oxide mediated signal transduction, cell adhesion and migration, and scavenger receptor activity. However, several pathways that are less known for their role in atherosclerosis were also identified, including carbohydrate metabolism, complement activation, calcium ion hemostasis, collagen catabolism, glycosyl bonds and hydrolase activity, taurine transporter activity, heparin activity, etc. The lack of oxygen radical metabolism among the significant processes was surprising, but consistent with up-regulation of genes related to oxygen radical metabolism in all groups with aging.


Taken together, these pathway analyses support prior observations regarding the importance of inflammatory molecular pathways in atherosclerosis, but additionally, expand the repertoire of molecular pathways that are involved in this disease process.


Identification of Other Time-related Patterns of Gene Expression in Atherosclerosis


The above analysis examined in detail genes with increased expression levels which correlate with atherosclerotic plaque development. However, additional patterns of gene expression were also identified in these longitudinal studies, to identify classes of genes and pathways not previously identified. For these analyses, the AUC algorithm was employed, which measured expression changes over time, made comparisons between the different strain/diet longitudinal datasets to identify gene expression changes specific for the apoE knockout model, and employed permutation to estimate the FDR (Tabibiazar et al. (2005), supra). Using this methodology several distinct gene expression patterns and pathways that reflect particular biological processes were identified (FIG. 4). For instance, some disease-related pathways were upregulated very early in the disease process and downregulated thereafter (Pattern 6). Others were upregulated early and maintained at relative high expression throughout the time course of the disease (Pattern 8). Whereas the earlier pattern is enriched in pathways representing biological processes such as extracellular matrix and collagen metabolism, as well as DNA replication and response to stress, the later pattern is enriched in pathways representing biological processes such as fatty acid metabolism, oxidoreductase activity and heat-shock protein activity. Some disease related pathways were upregulated in both early and late phases of disease development (Pattern 3), including those associated with metabolism, such as glycolysis and gluconeogenesis. Other patterns (Pattern 4) are represented by key pathways regulating plaque development, including growth factor, cytokine, and cell adhesion activity. Interestingly, inflammation is represented in almost all of the patterns described herein.


Identification of Stage Specific Gene Expression Signature Patterns


Classification approaches to human cancer have provided significant insights regarding the clinical features of the tumor, including propensity to metastasis, drug responsiveness, and long term prognosis (Golub et al. (1999) Science 286:531-537; Lapointe et al. (2004) Proc Natl Acad Sci 101:811-816; Paik et al. (2004) N Engl JMed (“Multigene Assay to Predict Recurrence of Tamoxifen-Treated, Node-Negative Breast Cancer”); Sorlie et al. (2001) Proc Natl Acad Sci 98:10869-10874). For atherosclerosis, the clinical utility of classification algorithms will include prediction of future events. To establish a panel of genes whose expression in the vessel wall can accurately classify disease stage, and which may thus be useful for clinical genomic and biomarker applications, the support vector machines algorithm was employed on this comprehensive mouse model disease data set. Employing the SVM classification algorithm, 38 genes were identified that were able to accurately classify each experiment with one of five defined stages of atherosclerosis in mice (FIG. 5A). The results demonstrated that these genes can distinguish normal from severe lesions with 100% accuracy. The intermediate stages of the disease are also distinguished from the other stages with a high degree of accuracy (88-97%) (Table 3).


To validate the classifier genes, their ability to accurately categorize an independent group of 16 week old apoE knockout mice, which were evaluated with a different array and labeling methodology, was evaluated. The microarray utilized different probes for some of the same genes. Moreover, the labeling methodology used a linear amplification step which may introduce further variability in the data. Using the SVM classification algorithm, each of the 4 replicate experiments was accurately classified with the correct stage of the disease process (FIG. 5B). As indicated by the greater correlation between gene expression in this independent group of mice and gene expression patterns in the original experimental group aged 24 weeks, the classifier genes accurately matched this validation dataset to the closest timepoint in the database.


Identification of Mouse Disease Gene Expression Patterns in Human Coronary Atherosclerosis


The expression profile of differentially regulated mouse genes was investigated in human coronary artery atherosclerosis. For transcriptional profiling of human atherosclerotic plaque, 40 coronary artery samples, dissected from explanted hearts of 17 patients undergoing orthotopic heart transplantation, were used. Of the 21 diseased segments, lesions ranged in severity from grade I to V (modified American Heart Association criteria based on morphological description (Virmani et al., supra)). For the purpose of this analysis, human artery segments were classified as non-lesion or lesion (combined all grades). Atherosclerosis related mouse genes were matched to human orthologs by gene symbol or by known homology (www.ncbi.nlm.nih.gov/entrez/query.fcgi?DB=homologene). Comparison of expression of the mouse genes between lesion and non-lesion human samples using the significance analysis of microarrays algorithm (FDR<0.025) revealed more than 100 mouse genes with higher expression in the diseased human tissue (FIG. 6). In view of the differences between the tissue samples used in these gene expression experiments, these constitute an important common set of disease relevant genes.


To further test the relevance of our findings in mouse atherosclerosis, the accuracy of the mouse classifier genes was assessed in human atherosclerotic disease, employing established statistical methods. The mouse classifier genes were first used to predict various stages of coronary artery disease in the human arterial samples. The results demonstrated a high degree of accuracy in predicting atherosclerotic disease severity (71.2 to 84.7% accuracy) (Table 3).


Additionally, the mouse classifier genes were used to categorize human atherectomy tissue obtained from coronary vessels treated for chronic atherosclerosis or in-stent restenosis. The pathophysiological basis of restenosis is quite distinct from that of chronic coronary atherosclerosis, and it was of interest to demonstrate that the classifier genes could distinguish the disease processes (Rajagopal and Rockson (2003) Am J Med 115:547-553). The results (Table 3) demonstrated significant accuracy in distinguishing the two types of lesions (85.4 to 93.7% accuracy), further validating the significance of the mouse atherosclerosis gene expression patterns in human disease. The greater accuracy of classification with these samples compared to the arterial segments likely reflects less variation in the clinical profile of the patients, which have much less complex medication and comorbid features than the pre-cardiac transplant patients in the above analysis.

TABLE 2Biological themes in atherosclerosis. Enrichment analysis of atherosclerosis-related genesannotated with Gene Ontology and Biocarta terms demonstrates involvement of multiplemolecular pathways and biological processes. Probabilities (p-values) were derived usingFisher exact test. 8478 of the entire microarray and 513 of genes in our set (includingadditional 183 genes which demonstrated Pearson correlation >0.8 with the upregulatedpattern) were annotated with GO, Biocarta, or other terms.List gene #Total gene #p-valueBiological Process (GO annotation)immune response1978<0.0001chemotaxis1023<0.0001cell surface receptor linked signal transduction1238<0.0001defense response1560<0.0001carbohydrate metabolism1467<0.0001antigen processing59<0.0001locomotory behavior46<0.0001inflammatory response830<0.0001complement activation512<0.0001proteolysis and peptidolysis252040.001antigen presentation4100.002intracellular signaling cascade282690.003zinc ion homeostasis220.004transmembrane receptor protein220.004tyrosine kinase activatiotext missing or illegible when filedhormone metabolism220.004hair cell differentiation220.004cell death220.004exogenous antigen via MHC class II370.006ossification4140.008collagen catabolism380.010classical pathway380.010vesicle transport along actin filament230.011taurine transport230.011nitric oxide mediated signal transduction230.011negative regulation of angiogenesis230.011endogenous antigen via MHC class I230.011endogenous antigen230.011cellular defense response (sensu Vertebrsta)230.011beta-alanine transport230.011lymph gland development4170.017perception of pain240.020myeloid blood cell differentiation240.020female gamete generation240.020cytolysis240.020ATP biosynthesis4190.025regulation of peptidyl-tyrosine phosphorylation3110.025neurotransmitter transport3120.032sex differentiation250.032exogenous antigen250.032call adhesion202170.039regulation of cell migration3130.040wound healing260.047ureteric bud branching260.047cellular defense response260.047acute-phase response260.047regulation of transcription from Pot II promoter6440.048hydrogen transport3140.049calcium ion homeostesis3140.049Molecular Function (GO annotation)acting on glycosyl bonds1231<0.0001interleukin receptor activity813<0.0001hydrolase activity67641<0.0001cytokine activity1357<0.0001hematopoietin932<0.0001complement activity59<0.0001cytokine binding33<0.0001C-C chemokine receptor activity33<0.0001chemokine activity47<0.0001cysteine-type endopeptidase activity11630.001tumor necrosis factor receptor activity350.002platelet-derived growth factor receptor binding220.004cathepsin D activity220.004beta-N-acetylhexosaminidase activity220.004antimicrobial peptide activity220.004scavenger receptor activity360.004cysteine-type peptidase activity9560.006mannosyl-oligosaccharide370.0061,2-alpha-mannosidase activitext missing or illegible when filedrecepter activity424790.009taurine:sodium symporter activity230.011taurine transporter activity230.011myosin ATPase activity230.011MHC class I receptor activity230.011cathepsin B activity230.011calcium channel regulator activity230.011beta-alanine transporter activity230.011catalytic activity232300.012solute:hydrogen antiporter activity240.020protein kinase C activity240.020tumor necrosis factor receptor binding3110.025hydrogen-exporting ATPase activity5290.028neurotransmitter:sodium symporter activity250.032MHC class II receptor activity250.32heparin binding5310.037endopeptidase inhibitor activity4220.041protein-tyrosine-phosphatase activity7540.043hydrogen ion transporter activity5330.046sulfuric ester hydrolase activity260.047Cellular Component (GO annotation)extracellular space1391148<0.0001lysosome2666<0.0001extracellular23117<0.0001integral to membrane1381637<0.0001membrane77862<0.0001integral to plasma membrane222050.006extracellular matrix141140.009external side of plasma membrane390.014Biocarta PathwaysclassicPathway33<0.0001il22bppathway47<0.0001nktPathway512<0.0001Ccr5Pathway5130.001reckPathway480.001compPathway340.001il7Pathway4100.002TPOPathway5170.003cxcr4Pathway5170.003blymphocytePathway220.004il10Pathway370.006pdgfPathway5220.009ionPathway230.011egfPathway5230.011biopeptidesPathway5230.011bcrPathway5250.015ghPathway4170.017fcer1Pathway5260.018spryPathway3100.019neutrophilPathway240.020mrpPathway240.020trkaPathway3110.025pmlPathway3110.025srcRPTPPathway3120.032plcdPathway250.032itngPathway250.032il2Pathway3130.040RacCycDPathway4220.041lymphocytePathway260.047nuclearRsPathway3140.049cdMacPathway3140.049CCR3Pathway3140.049Summary annotation for Inflammatory genesdefense1554<0.0001chemokine922<0.0001interleukin938<0.0001cytokine181440.003TNF4130.006TH24150.011TH14160.013macrophage3130.040









TABLE 3










Classification of mouse and human atherosclerotic tissues employing mouse classifier genes.


To validate the accuracy of mouse classifier genes in predicting disease severity we utilized


various mouse and human expression datasets. The SVM algorithm was utilized for cross


validation of mouse experiments grouped on the basis of (A) stage of disease (no disease-


apoE time 0, mild disease-apoE at 4 and 10 weeks on normal diet, mild-moderate disease-


apoE at 4 and 10 weeks on highfat diet, moderate disease-apoE at 24 and 40 weeks on normal


diet, and severe disease-apoE at 24 and 40 weeks on high fat diet); (B) 3 different time points


(apoE at 0 vs. 10, vs. 40 weeks); (C) Human coronary artery with lesion vs. no lesion; and (D)


atherectomy samples derived from in-stent restenosis vs. native atherosclerotic lesions.


For each analysis, the accuracy of classification is represented in tabular fashion with the


confusion matrix generated using N-fold cross validation methods.
















A
TRUE
TRUE
TRUE
TRUE
TRUE



PREDICTED
No dz
Mild_dz
Mild_mod dz
Mod_dz
Severe_dz
Correct [%]





No dz
64
0
1
0
0
98.5


Mild_dz
2
140
0
0
0
98.6


Mild_mod dz
0
0
148
20
0
88.1


Mod_dz
0
0
3
149
0
98.0


Severe_dz
0
0
0
0
173
100.0


Correct [%]
97.0
100.0
97.4
88.2
100.0














B
TRUE
TRUE
TRUE



PREDICTED
ApoE_T00_NC
ApoE_T10_HF
ApoE_T40_HF
Correct [%]





ApoE_T00_NC
68
0
0
100


ApoE_T10_HF
0
56
0
100


ApoE_T40_HF
0
0
76
100


Correct [%]
100
100
100















C
TRUE
TRUE




PREDICTED
Lesion
No lesion
Correct [%]







Lesion
183
33
84.7



No lesion
53
131
71.2



Correct [%]
77.5
79.9







D
TRUE
TRUE



PREDICTED
ISR
De novo
Correct [%]







ISR
345
44
88.7



De novo
59
652
91.7



Correct [%]
85.4
93.7










Example 2
Mouse Strain—Specific Differences in Vascular Wall Gene Expression and Their Relationship to Vascular Disease

Methods


RNA Preparation and Hybridization to the Microarray


Three-week old female C3H/HeJ, C57B1/6J, and apoE knock-out mice (C57BL/6J-ApoetmlUnc) were purchased from Jackson Labs (JAX® Mice and Services, Bar Harbor, Me.). At four weeks of age the mice were either continued on normal chow or switched to non-cholate containing high-fat diet which included 21% anhydrous milkfat and 0.15% cholesterol (Dyets #101511, Dyets Inc., Bethlehem, Pa.) for a maximum period of 40 weeks. At each of the time-points, including 0 (baseline), 4, 10, 24 and 40 weeks, for each of the conditions (strain-diet combination), 15 mice were harvested for RNA isolation, for a total of 450 mice. Following Stanford University animal care guidelines, the mice were anesthetized with Avertin and perfused with normal saline. The aortas from the root to the common iliacs were carefully dissected, flash frozen in liquid nitrogen, and divided into three pools of five aortas for further RNA isolation. Total RNA was isolated as described in Tabibiazar et al. (2003) Circ Res 93:1193-1201. First strand cDNA was synthesized from 10 μg of total RNA from each pool and from whole 17.5-day embryo for reference RNA in the presence of Cy5 or Cy3 dCTP, respectively, and hybridized to a mouse 60mer oligo microarray (G4120A, Agilent Technologies, Palo Alto, Calif.), generating three biological replicates for each time point.


Data Processing


Array image acquisition and feature extraction was performed using the Agilent G2565AA Microarray Scanner and feature extraction software version A.6.1.1. Normalization was carried out using a LOWESS algorithm, and Dye-normalized signals were used in calculating log ratios. Features with reference values of<2.5 standard deviations above background for the negative control features were regarded as missing values. Those features with values in at least ⅔ of the experiments and present in at least one of the replicates were retained for further analysis. For SAM analyses, a K-nearest-neighbor (KNN) algorithm was applied to impute for missing values. (Tabibiazar et al. (2003), supra.)


Data Analysis


Experimental design and analysis flow chart is depicted in FIG. 7. Significance Analysis of Microarrays (SAM) was employed to identify genes with statistically different expression between the C3H and C57 mice at baseline. (Tabibiazar et al. (2003), supra; Tusher et al. (2001) PNAS 98:5116-5121; Chen et al. (2003) Circulation 108:1432-1439.) For partitioning clustering of the genes with K-Means and self-organizing-maps (SOM), we used positive correlation for distance determination and required complete linkage, which uses the greatest distance between genes to ascribe similarity. SOM and K-Means analyses were performed using Expressionist software (GeneData, Inc., USA). Heatmaps were generated using HeatMap Builder. For enrichment analysis we used the EASE analysis software which employs Gene Ontology (GO) annotation and the Fisher's exact test to derive biological themes within particular gene sets. (Hosack et al. (2003) Genome Biol. 4:R70.) For time-course study, a new statistical algorithm, the Area-Under-Curve (AUC) analysis was devised. For each sequence of 4 triplicate gene expression measurements over time, we first subtracted the measurement at time 0 from all values. We then computed the signed area under the curve. The area is a natural measure of change over time. These areas were then used to compute an F-statistic for comparing C57 and C3H mice across the different diets. A permutation analysis, similar to that employed in SAM, was carried out to estimate the false discovery rate (q-value or “FDR”) for different levels of the F-statistic. For ease of presentation, genes which meet our FDR cutoffs will be referred to as “significant” throughout the remainder of the article. All microarray data were submitted to the NCBI Gene Expression Omnibus (GEO GSE1560; http://www.ncbi.nlm.nih.gov/geo/).


Aortic Lesion Analysis


For select time points within various experimental groups, 5 to 7 female mice were used for histological lesion analysis. Atherosclerosis lesion area was determined as described in Tangirala et al. (1995) 36:2320-2328.


Quantitative Real-Time Reverse Transcriptase-Polymerase Chain Reaction


Primers and probes for 10 representative differentially expressed genes were obtained from Applied Biosystems Assays-on-Demand. A Total of 90 reactions were performed from representative RNA samples used for microarray experiments. These included triplicate assay on three pools of five aortas. cDNA was synthesized and Taqman was performed as described in Tabibiazar et al. (2003), supra.


Results


Baseline Differences in Gene Expression Patterns between the Mouse Strains


Differences in gene expression levels between the two strains at baseline, before effects of aging or diet become apparent, may identify genes that play a role in determining vascular wall disease susceptibility. To identify such genes SAM was used to compare the vascular wall gene expression of C3H vs. C57 mice at 4 weeks of age, with all animals on normal chow diet. SAM identified 311 genes as being significantly differentially expressed (FDR<0.1 with>1.5 fold difference), and expression patterns of these genes provided a clear partition between C3H and C57 mice (FIG. 8). A separate 2-class comparison (SAM, FDR<0.1) between C57 and apoE-deficient mice with a C57B1/6 genetic background revealed only a few genes, including Apo-E, which were differentially expressed in the 2 groups of mice (data not shown).


Comparison of C3H and C57 vascular wall gene expression at baseline provided a list of compelling candidate genes which reflected differences in biological processes such as growth, differentiation, and inflammation as well as molecular functions such as cathecholamine synthesis, phosphatase activity, peroxisome function, insulin like growth factor activity, and antigen presentation (FIG. 8). These processes were exemplified by higher expression of genes such as Cdknla, Pparbp, protein tyrosine phosphatase-4a2, and Socs5 in C3H mice, compared with genes such as ABCC1, H2-D1, Bat5, IGFBP1, SCD1, and Serpine6b which demonstrated higher expression in C57 mice. These fundamental baseline gene expression differences may determine disease susceptibility as the mice are exposed to age-related stimuli or dietary challenges.


Age-related Differences in Gene Expression Patterns between the Mouse Strains


To further examine the vascular wall gene expression differences between C57 and C3H mice, an analysis was performed to identify genes differentially expressed in response to aging (FIG. 9). Data was collected at five time points over a 40 week period. To identify such genes, we developed the Area Under the Curve (AUC) analysis. The AUC analysis relies on a permutation procedure to reduce the number of potential false positives generated due to multiple testing, but still utilizes the increase in statistical power of time-course experimental design. Comparing C57 vs. C3H time-course differences on normal diet with a rigid cutoff (FDR<0.05) did not identify any genes. However, relaxing the AUC stringency (f-statistic>10, FDR <0.45) allowed a large number of genes (413) to be included for pathway over-representation analysis using GO annotation. Functional annotation and group over-representation analysis (Fisher test p-value <0.02) of the resultant differentially expressed genes revealed differences in a number of biological processes, including growth and development, as well as a number of molecular fimctions such as cell cycle control, regulation of mitosis, and metabolism (FIG. 9b). Some of these processes are exemplified by genes with higher expression in C57 mice, such as Aocl (pro-oxidative stress), Bub1 (cell cycle check point), Cyclin B2, as well as genes with higher expression in C3H, including INHBA and INHBB.


Temporally variable genes identified by AUC analysis were further characterized with K-Means clustering to identify dynamic patterns of expression during the aging process (FIG. 3c). Clusters 1, 4, and 9 revealed either higher overall expression or temporally increasing levels of expression in C3H mice compared with C57 mice. In contrast, clusters 2, 6, and 14 revealed the opposite pattern. Of the genes which were noted to be differentially expressed in the two strains during aging, 51 genes were also differentially expressed at baseline, suggesting that baseline differences of certain genes can further be affected with aging.


Diet-related Differences in Gene Expression Patterns between the Mouse Strains


Differential vascular wall response to atherogenic stimuli was determined by comparing temporal gene expression patterns in C57 vs. C3H mice on high-fat diet (FIG. 10A). Comparing C57 vs. C3H time-course differences on high-fat diet with a rigid cutoff (FDR<0.05) identified 35 genes, including Hgfl and Tgf4, which were down regulated in C57 on high-fat diet. Additional known genes, as well as a number of ESTs were also identified. Employing a less stringent AUC cutoff allowed identification of a larger number of genes, which could be evaluated with pathway over-representation analysis using GO annotation. At this level of stringency (f-statistic>10, FDR<0.35), a total of 650 genes with temporally variable expression were identified. Genes that were also differentially regulated by the aging process (141 of 650 genes) were excluded from further analysis of this group. 38 of the remaining 509 genes were among those differentially expressed at baseline. Functional annotation and group over-representation analysis (Fisher test p-value<0.02) of these differentially expressed genes revealed differences in biological processes such as catabolism, oxygen reactive species and superoxide metabolism, and proteo- and peptidolysis as well as molecular functions such as fatty acid metabolism, oxidoreductase and methyltransferase activities (FIG. 10B). Interestingly, this analysis suggested important differences between the two mouse strains with respect to the activity of the peroxisome, microbody and lysosome. Some of these processes were exemplified by genes with higher expression in C3H mice, such as Ccs, Ephx2, Gpx4, Prdx6 (anti-oxidants), Sirt3 (transcriptional repressor), PPARa, and Mcd, as well as genes with higher expression in C57 mice, such as Lysyl oxidase and Cdkn1a. K-means clustering of these genes identified a small number of distinct expression patterns (FIG. 10C), with clusters 3 and 9 revealing increased gene expression in C3H mice and clusters 8 and 10 showing the opposite pattern.


Evaluation of Strain-specific Differentially Regulated Genes in the ApoE Model


Using these techniques, a significant number of genes have been identified that are differentially expressed in the atherosclerosis resistant C3H and susceptible C57 mice, some of which are likely involved in atherogenesis and some of which are likely irrelevant to the process. To further select genes most likely to be involved in atherogenesis, expression in apoE-deficient mice fed normal or high-fat diet over a period of 40 weeks was investigated (FIG. 11). We utilized SOM analysis to visualize the expression profiles of these subsets of genes throughout the development and progression of atherosclerosis in the ApoE-deficient mice. The analysis revealed several patterns of gene expression. For example, SOM cluster 8 demonstrated a consistently increasing pattern of expression which correlated with disease progression in the apoE-deficient mice (FIG. 11). As evidenced by the pie chart, this cluster is enriched with genes that were identified as more highly expressed in C57 versus C3H mice at baseline (i.e., potentially atherogenic). In contrast, clusters 4, 5, and 6 showed decreasing expression with disease progression. The decreased expression of genes in cluster 4 was somewhat attenuated with high-fat challenge of the ApoE-deficient mice. This cluster is particularly enriched with genes that had revealed a higher expression in C3H mice (i.e., potentially atheroprotective) with atherogenic stimuli and with aging.


Given C3H resistance and C57 susceptibility to atherosclerosis, as an initial hypothesis it was postulated that genes with higher expression in C3H mice confer resistance, whereas genes with higher expression in C57 mice may have a pro-atherogenic role. With this point of reference, gene clusters were further examined. For example, limiting the list of genes in SOM cluster 8 (genes with increased expression with atherosclerosis) to those that also had higher baseline expression in C57 mice yielded an interesting set of genes that may be atherogenic. This group included inflammation related genes such as H2-D1, Pdgfc, Paf, and Cd47. Other compelling genes included Agpt2, Mglap, Xdh, Th, and Ctsc. Conversely, limiting the list of genes in clusters 4 and 5 to those with higher expression in C3H mice identified a group of genes with potential athero-protective function. Some of those genes included Pparα, Pparbp, as well as Ptp4a1, and Mcd.


Lesion Analysis in the Genetic Models


To address whether some of the gene expression differences are related to presence of atherosclerotic lesion in C57 mice, the total atherosclerotic burden was determined in the aorta by calculating a percent lesion area in aortas of C57 (n=5) and C3H (n=5) mice. Comparisons were made at time 0 and 40 weeks on normal or high-fat diet. Non-cholate containing high-fat diet was used to prevent caustic effects on the vascular wall. As expected, C57 and C3H mice on either diet did not demonstrate evidence of atherosclerosis throughout the course of the experiment, suggesting that observed gene expression changes cannot be explained by different cellular composition of the vessel wall. Although minimal fatty infiltrates were noted on histological evaluation of the aortic root in C57 mice on high-fat diet, there were no obvious changes in inflammatory cell infiltrate.


Quantitative RT-PCR Validation of Expression Differences


To validate the array results with quantitative RT-PCR and assure that the statistical analyses were identifying truly differentially expressed genes, ten representative genes were assayed by quantitative RT-PCR. Several genes were used from each group of significant genes. There is high degree of correlation between the two methodologies (Pearson correlation of 0.86), validating the results of the microarray analyses.


Although the foregoing invention has been described in some detail by way of illustration and examples for purposes of clarity of understanding, it will be apparent to those skilled in the art that certain changes and modifications may be practiced without departing from the spirit and scope of the invention. Therefore, the description should not be construed as limiting the scope of the invention.


All publications, patents and patent applications cited herein are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual publication, patent or patent application were specifically and individually indicated to be so incorporated by reference.

Claims
  • 1. A system for detecting gene expression, comprising at least two isolated polynucleotide molecules, wherein each of said at least two isolated polynucleotide molecules detects an expressed gene product from a gene that is differentially expressed in atherosclerotic disease in a mammal, wherein said gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927.
  • 2. A system for detecting gene expression, comprising at least two isolated polynucleotide sequences, wherein each of said at least two isolated polynucleotide molecules detects an expressed gene product from a gene that is differentially expressed in atherosclerotic disease in a mammal, wherein said gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 3. A system for detecting gene expression according to claim 1, wherein at least one of said isolated polynucleotide molecules detects a expressed gene product from a gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 4. A system according to claim 1, wherein the isolated polynucleotide molecules are immobilized on an array.
  • 5. A system according to claim 4, wherein the array is selected from the group consisting of a chip array, a plate array, a bead array, a pin array, a membrane array, a solid surface array, a liquid array, an oligonucleotide array, polynucleotide array or a cDNA array, a microtiter plate, a membrane, and a chip.
  • 6. A system according to claim 1, wherein the isolated polynucleotides are selected from the group consisting of synthetic DNA, genomic DNA, cDNA, RNA, or PNA.
  • 7. A kit comprising the system of claim 1.
  • 8. A kit comprising the system of claim 4.
  • 9. A method of monitoring atherosclerotic disease in an individual, comprising detecting the expression level of at least one gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927.
  • 10. The method of claim 9, wherein said at least one gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 11. The method of claim 9, comprising detecting the expression level of at least two of said genes.
  • 12. The method of claim 11, wherein at least one of said at least two genes is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 13. The method of claim 9, comprising detecting the expression level of at least ten of said genes.
  • 14. The method of claim 13, wherein at least one of said at least ten genes is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 15. The method of claim 9, comprising detecting the expression level of at least one hundred of said genes.
  • 16. The method of claim 15, wherein at least one of said at least one hundred genes is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 17. The method of claim 9, wherein said atherosclerotic disease comprises coronary artery disease.
  • 18. The method of claim 9, wherein said atherosclerotic disease comprises carotid atherosclerosis.
  • 19. The method of claim 9, wherein said atherosclerotic disease comprises peripheral vascular disease.
  • 20. The method of claim 9, wherein said expression level is detected by measuring the RNA level expressed by said one or more genes.
  • 21. The method of claim 20, comprising isolating RNA from said individual prior to detecting the RNA expression level.
  • 22. The method of claim 20, wherein said detection of said RNA expression level comprises amplifying RNA from said individual.
  • 23. The method of claim 22, wherein amplification of RNA comprises polymerase chain reaction (PCR).
  • 24. The method of claim 20, wherein detection of said RNA expression level comprises hybridization of RNA from said individual to a polynucleotide corresponding to said at least one gene selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 1-927.
  • 25. The method of claim 20, wherein said expression level is detected by measuring the protein level expressed by said one or more genes.
  • 26. The method of claim 9, further comprising selecting an appropriate therapy for said atherosclerotic disease.
  • 27. The method of claim 9, comprising detecting the expression of said at least one gene in serum from said individual.
  • 28. The method of claim 20, comprising measuring said RNA level in serum from said individual.
  • 29. The method of claim 25, comprising measuring said protein level in serum from said individual.
  • 30. A method of monitoring atherosclerotic disease in an individual, comprising detecting RNA expressed from at least one gene selected from the group of genes corresponding to at least one polynucleotide sequence depicted in SEQ ID NOs: 1-927.
  • 31. The method of claim 30, wherein said at least one gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 32. The method of claim 30, comprising measuring said RNA in serum from said individual.
  • 33. A method of monitoring atherosclerotic disease in an individual, comprising detecting protein expressed from at least one gene selected from the group of genes corresponding to at least one polynucleotide sequence depicted in SEQ ID NOs: 1-927.
  • 34. The method of claim 33, wherein said at least one gene is selected from the group of genes corresponding to the polynucleotide sequences depicted in SEQ ID NOs: 8, 14, 26, 32, 50, 64, 83, 99, 142, 154, 159, 161, 177, 181, 200, 390, 430, 434, 439, 440, 476, 491, 508, 530, 534, 565, 567, 572, 624, 647, 657, 690, 733, 745, 806, 824, 886, 882, 901, 905, 913, and 927.
  • 35. The method of claim 33, comprising measuring said protein in serum from said individual.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 60/664,550, filed Mar. 22, 2005, which is incorporated by reference herein in its entirety.

Provisional Applications (1)
Number Date Country
60664550 Mar 2005 US