METHODS AND COMPOSITIONS FOR DIAGNOSIS, MONITORING AND DEVELOPMENT OF THERAPEUTICS FOR TREATMENT OF ATHEROSCLEROTIC DISEASE

Information

  • Patent Application
  • 20090305903
  • Publication Number
    20090305903
  • Date Filed
    September 05, 2008
    15 years ago
  • Date Published
    December 10, 2009
    14 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-Apoe5m1Unc), 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 C57B1/6 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 TOO, 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. 10B: 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. 10C: 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-ApoetmlUnc) 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, Calif. (1990); Arnheim 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. Pat. 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 60mer sequences represented in Table 1 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(1):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® 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™ 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™ 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, 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 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 0.1×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, RNA 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, et al. (1981) J. Mol. Biol. 150:1; Santerre, et al. (1984) Gene 30:147; Janknecht, et al. (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 1





Polynucleotide sequences which detect differentially



expressed genes in atherosclerotic disease




















SEQ







ID

GENE
GENE
CLONE


NO:
CLONE ID
SYMBOL
NAME
NAME





  1.
C0267B04-3

C0267B04-5N
C0267B04





NIA Mouse





7.5-dpc Whole





Embryo cDNA





Library (Long)






Mus musculus






cDNA clone





NIA: C0267B04





IMAGE: 30017007





5′, MRNA





sequence





  2.
M29697.1
Il7r
interleukin 7
M29697





receptor





  3.
L0304D03-3
Wnt4
wingless-
L0304D03





related MMTV





integration site 4





  4.
L0237D12-3
Ctsd
cathepsin D
L0237D12





  5.
C0266B08-3
BM204200
ESTs
C0266B08





BM204200





  6.
J0537C05-3
Pfdn2
prefoldin 2
J0537C05





  7.
L0216F02-3
C430008C19Rik
RIKEN cDNA
L0216F02





C430008C19





gene





  8.
NM_017372.1
Lyzs
lysozyme
NM_017372





  9.
C0271B02-3
4732437J24Rik
RIKEN cDNA
C0271B02





4732437J24





gene





 10.
H3022C10-3
AA408868
expreexpressed
H3022C10





sequence





AA408868





 11.
L0806E05-3
Gtl2
GTL2,
L0806E05





imprinted





maternally





expressed





untranslated





mRNA





 12.
H3111E06-5
Acas2l
acetyl-
H3111E06





Coenzyme A





synthetase 2





(AMP





forming)-like





 13.
H3091H05-3
Hras1
Harvey rat
H3091H05





sarcoma virus





oncogene 1





 14.
K0324B10-3
Timp1
tissue inhibitor
K0324B10





of





metalloproteinase 1





 15.
K0508B06-3

transcribed
K0508B06





sequence with





moderate





similarity to





protein





ref: NP_077285.1





(H. sapiens)





A20-binding





inhibitor of NF-





kappaB





activation 2;





LKB1-





interacting





protein [Homo






sapiens]






 16.
C0176A01-3
Syngr1
synaptogyrin 1
C0176A01





 17.
J0748G02-3

AU018093
J0748G02





Mouse two-cell





stage embryo





cDNA Mus






musculus






cDNA clone





J0748G02 3′,





MRNA





sequence





 18.
J0035G10-3
C77672
ESTs C77672
J0035G10





 19.
C0630C02-3
Cxcl16
chemokine
C0630C02





(C—X—C motif)





ligand 16





 20.
K0313A10-3
5430435G22Rik
RIKEN cDNA
K0313A10





5430435G22





gene





 21.
L0070E11-3
Cbfa2t1h
CBFA2T1
L0070E11





identified gene





homolog





(human)





 22.
H3072E02-3
BG069076
ESTs
H3072E02





BG069076





 23.
H3079B06-3


Mus musculus

H3079B06





unknown





mRNA





 24.
H3002D08-3
4833412N02Rik
RIKEN cDNA
H3002D08





4833412N02





gene





 25.
H3159A08-3
Gp49b
glycoprotein 49 B
H3159A08





 26.
C0612F12-3
BM207436
ESTs
C0612F12





BM207436





 27.
H3108A03-3
Apobec1
apolipoprotein
H3108A03





B editing





complex 1





 28.
C0180G01-3
BI076556
ESTs BI076556
C0180G01





 29.
C0938A03-3
Sf3a1
splicing factor
C0938A03





3a, subunit 1





 30.
J0703E02-3
Ogdh
oxoglutarate
J0703E02





dehydrogenase





(lipoamide)





 31.
C0274D12-3

transcribed
C0274D12





sequence with





moderate





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





 32.
H3097H03-3
Expi
extracellular
H3097H03





proteinase





inhibitor





 33.
H3074D10-3

transcribed
H3074D10





sequence with





weak similarity





to protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





 34.
M14222.1
Ctsb
cathepsin B
M14222





 35.
C0176G01-3
2400006H24Rik
RIKEN cDNA
C0176G01





2400006H24





gene





 36.
H3092F08-5

UNKNOWN:
H3092F08





Similar to Mus






musculus






immediate-





early antigen





(E-beta) gene,





partial intron 2





sequence





 37.
H3054F02-3
1200003C15Rik
RIKEN cDNA
H3054F02





1200003C15





gene





 38.
C0012F07-3
3010021M21Rik
RIKEN cDNA
C0012F07





3010021M21





gene





 39.
L0955A10-3
9030409G11Rik
RIKEN cDNA
L0955A10





9030409G11





gene





 40.
L0045B05-3

transcribed
L0045B05





sequence with





moderate





similarity to





protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





 41.
H3049A10-3
BG066966
ESTs
H3049A10





BG066966





 42.
X70298.1
Sox4
SRY-box
X70298





containing gene 4





 43.
L0001C09-3

transcribed
L0001C09





sequence with





weak similarity





to protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





 44.
H3010D12-5

UNKNOWN:
H3010D12





Similar to Mus






musculus






RIKEN cDNA





8430421I07





gene





(8430421I07Rik),





mRNA





 45.
C0923E12-3
Ptpns1
protein tyrosine
C0923E12





phosphatase,





non-receptor





type substrate 1





 46.
C0941E09-3
D330001F17Rik
RIKEN cDNA
C0941E09





D330001F17





gene





 47.
K0534C04-3
Tce1
T-complex
K0534C04





expressed gene 1





 48.
H3064E11-3
BG068354
ESTs
H3064E11





BG068354





 49.
L0957C02-3
E130319B15Rik
RIKEN cDNA
L0957C02





E130319B15





gene





 50.
L0240C12-3
C1qa
complement
L0240C12





component 1, q





subcomponent,





alpha





polypeptide





 51.
J0018H07-3
Rnf149
ring finger
J0018H07





protein 149





 52.
K0508E12-3
Rin3
Ras and Rab
K0508E12





interactor 3





 53.
L0208A01-3
4933437K13Rik
RIKEN cDNA
L0208A01





4933437K13





gene





 54.
C0239G03-3
BM202478
EST
C0239G03





BM202478





 55.
L0518C11-3
1700016K05Rik
RIKEN cDNA
L0518C11





1700016K05





gene





 56.
H3054C09-3
Oas1c
2′-5′
H3054C09





oligoadenylate





synthetase 1C





 57.
L0811E07-3
3110057O12Rik
RIKEN cDNA
L0811E07





3110057O12





gene





 58.
J0948A06-3


Mus musculus

J0948A06





mRNA similar





to RIKEN





cDNA





4930503E14





gene (cDNA





clone





MGC: 58418





IMAGE: 6708114),





complete





cds





 59.
C0931B05-3

transcribed
C0931B05





sequence with





weak similarity





to protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





 60.
H3022A09-3
Eps8l2
EPS8-like 2
H3022A09





 61.
G0118B03-3
Usf2
upstream
G0118B03





transcription





factor 2





 62.
H3156C12-3
Ms4a6d
membrane-
H3156C12





spanning 4-





domains,





subfamily A,





member 6D






 63.
H3074G06-3
9530020G05Rik
RIKEN cDNA
H3074G06





9530020G05





gene





 64.
NM_003254.1
TIMP1
tissue inhibitor
NM_003254





of





metalloproteinase





1 (erythroid





potentiating





activity,





collagenase





inhibitor)





 65.
K0647H07-3
Il7r
interleukin 7
K0647H07





receptor





 66.
J0257F12-3
Rnf25
ring finger
J0257F12





protein 25





 67.
H3083G02-3
Lcn2
lipocalin 2
H3083G02





 68.
M64086.1
Serpina3n
serine (or
M64086





cysteine)





proteinase





inhibitor, clade





A, member 3N





 69.
C0906B05-3
Cenpc
centromere
C0906B05





autoantigen C





 70.
H3094B08-3
BG071051
ESTs
H3094B08





BG071051





 71.
K0110F02-3
Pstpip1
proline-serine-
K0110F02





threonine





phosphatase-





interacting





protein 1





 72.
L0072G08-3
Renbp
renin binding
L0072G08





protein





 73.
J0088G06-3
4930472G13Rik
RIKEN cDNA
J0088G06





4930472G13





gene





 74.
K0121F05-3
Fcgr2b
Fc receptor,
K0121F05





IgG, low





affinity IIb





 75.
K0124E12-3
Wbscr5
Williams-
K0124E12





Beuren





syndrome





chromosome





region 5





homolog





(human)





 76.
K0649H05-3
F730038I15Rik
RIKEN cDNA
K0649H05





F730038I15





gene





 77.
K0154C05-3
D230024O04
hypothetical
K0154C05





protein





D230024O04





 78.
C0182E05-3
Hmox1
heme
C0182E05





oxygenase





(decycling) 1





 79.
L0823E04-3

transcribed
L0823E04





sequence with





weak similarity





to protein





pir: T26134





(C. elegans)





T26134





hypothetical





protein





W04A4.5 -






Caenorhabditis







elegans






 80.
K0130E05-3
9830126M18
hypothetical
K0130E05





protein





9830126M18





 81.
C0908B11-3
P2ry6
pyrimidinergic
C0908B11





receptor P2Y,





G-protein





coupled, 6





 82.
K0438A08-3
Ccl2
chemokine (C-
K0438A08





C motif) ligand 2





 83.
H3082C12-3
Spp1
secreted
H3082C12





phosphoprotein 1





 84.
H3014A12-3
Capg
capping protein
H3014A12





(actin filament),





 85.
H3089C11-3
BG070621
ESTs
H3089C11





BG070621





 86.
X67783.1
Vcam1
vascular cell
X67783





adhesion





molecule 1





 87.
J0509D03-3

AU018874
J0509D03





Mouse eight-





cell stage





embryo cDNA






Mus musculus






cDNA clone





J0509D03 3′,





MRNA





sequence





 88.
H3055A11-5

UNKNOWN:
H3055A11





Similar to






Homo sapiens






KIAA1363





protein





(KIAA1363),





mRNA





 89.
C0455A05-3
AW413625
expressed
C0455A05





sequence





AW413625





 90.
NM_019732.1
Runx3
runt related
NM_019732





transcription





factor 3





 91.
L0008A03-3
AW546412
ESTs
L0008A03





AW546412





 92.
K0329C10-3
Thbs1
thrombospondin 1
K0329C10





 93.
H3115H03-3
BC019206
cDNA sequence
H3115H03





BC019206





 94.
C0643F09-3
Usp18
ubiquitin
C0643F09





specific





protease 18





 95.
X84046.1
Hgf
hepatocyte
X84046





growth factor





 96.
L0236C05-3
Aldh1b1
aldehyde
L0236C05





dehydrogenase





1 family,





member B1





 97.
H3055E08-3
Mcoln2
mucolipin 2
H3055E08





 98.
H3009F12-3
BG063639
ESTs
H3009F12





BG063639





 99.
J0208G12-3
Cxcl1
chemokine
J0208G12





(C—X—C motif)





ligand 1





100.
K0300C11-3
9130025P16Rik
RIKEN cDNA
K0300C11





9130025P16





gene





101.
H3104F03-5
Krt1-18
keratin complex
H3104F03





1, acidic, gene





18





102.
L0858D08-3
Trim2
tripartite motif
L0858D08





protein 2





103.
L0508H09-3
BY564994
EST BY564994
L0508H09





104.
L0701G07-3
BM194833
ESTs
L0701G07





BM194833





105.
K0102A10-3
E430025L02Rik
RIKEN cDNA
K0102A10





E430025L02





gene





106.
C0190H11-3
Spn
sialophorin
C0190H11





107.
L0514A11-3
2810457I06Rik
RIKEN cDNA
L0514A11





2810457I06





gene





108.
J0911E11-3
Nefl
neurofilament,
J0911E11





light





polypeptide





109.
K0647E02-3
Def6
differentially
K0647E02





expressed in





FDCP 6





110.
H3091E09-3
Eif1a
eukaryotic
H3091E09





translation





initiation factor





1A





111.
AF286725.1
Pdgfc
platelet-derived
AF286725





growth factor,





C polypeptide





112.
D31942.1
Osm
oncostatin M
D31942





113.
L0046B04-3
Alcam
activated
L0046B04





leukocyte cell





adhesion





molecule





114.
K0131D09-3
LOC217304
similar to
K0131D09





triggering





receptor





expressed on





myeloid cells 5





(LOC217304),





mRNA





115.
H3024C07-3
Hexa
hexosaminidase A
H3024C07





116.
L0251A07-3
B4galt1
UDP-
L0251A07





Gal:betaGlcNAc





beta 1,4-





galactosyltransferase,





polypeptide 1





117.
C0612G04-3
Grip1
glutamate
C0612G04





receptor





interacting





protein 1





118.
C0357B04-3

C0357B04-3
C0357B04





NIA Mouse





Undifferentiated





ES Cell





cDNA Library





(Short) Mus






musculus






cDNA clone





C0357B04 3′,





MRNA





sequence





119.
L0529E02-3
Egfl3
EGF-like-
L0529E02





domain,





multiple 3





120.
L0218E05-3
Dnase2a
deoxyribonuclease
L0218E05





II alpha





121.
H3074C12-3
Dutp
deoxyuridine
H3074C12





triphosphatase





122.
H3072F09-3
Icsbp1
interferon
H3072F09





consensus





sequence





binding protein 1





123.
C0829F05-3
4632404H22Rik
RIKEN cDNA
C0829F05





4632404H22





gene





124.
L0063A12-3

similar to
L0063A12





ubiquitin-





conjugating





enzyme UBCi





(LOC245350),





mRNA





125.
C0143E09-3
6330548O06Rik
RIKEN cDNA
C0143E09





6330548O06





gene





126.
K0127G03-3

transcribed
K0127G03





sequence with





weak similarity





to protein





ref: NP_000072.1





(H. sapiens)





beige protein





homolog;





Lysosomal





trafficking





regulator





[Homo sapiens]





127.
H3109D03-3
Lamp2
lysosomal
H3109D03





membrane





glycoprotein 2





128.
J0034B02-3
Dhx16
DEAH (Asp-
J0034B02





Glu-Ala-His)





box polypeptide





16





129.
K0428C07-3
Plcb3
phospholipase
K0428C07





C, beta 3





130.
K0119F10-3
Ccl9
chemokine (C-
K0119F10





C motif) ligand 9





131.
J0046B07-3
Tuba4
tubulin, alpha 4
J0046B07





132.
C0117E11-3
Neu1
neuraminidase 1
C0117E11





133.
C0101C01-3
Sdc1
syndecan 1
C0101C01





134.
K0245A03-3
9130012B15Rik
RIKEN cDNA
K0245A03





9130012B15





gene





135.
H3109A02-3
Fcer1g
Fc receptor,
H3109A02





IgE, high





affinity I,





gamma





polypeptide





136.
L0819C05-3
Mapk8ip
mitogen
L0819C05





activated





protein kinase 8





interacting





protein





137.
U77083.1
Anpep
alanyl
U77083





(membrane)





aminopeptidase





138.
C0164B01-3
Tnfaip2
tumor necrosis
C0164B01





factor, alpha-





induced protein 2





139.
H3085G03-3
Cyba
cytochrome b-
H3085G03





245, alpha





polypeptide





140.
H3074F04-3
Abcc3
ATP-binding
H3074F04





cassette, sub-





family C





(CFTR/MRP),





member 3





141.
H3145E02-3
Wbp1
WW domain
H3145E02





binding protein 1





142.
K0609F07-3
Cd53
CD53 antigen
K0609F07





143.
K0205H04-3
9830148O20Rik
RIKEN cDNA
K0205H04





9830148O20





gene





144.
H3095H04-3
2410002I16Rik
RIKEN cDNA
H3095H04





2410002I16





gene





145.
C0623H08-3
Tm7sf1
transmembrane
C0623H08





7 superfamily





member 1





146.
L0242F05-3
2700088M22Rik
RIKEN cDNA
L0242F05





2700088M22





gene





147.
C0177F02-3
Sdc3
syndecan 3
C0177F02





148.
L0803B02-3
Ppp1r9a
protein
L0803B02





phosphatase 1,





regulatory





(inhibitor)





subunit 9A





149.
H3061D01-3
BB172728
ESTs
H3061D01





BB172728





150.
L0259D11-3
C1qb
complement
L0259D11





component 1, q





subcomponent,





beta





polypeptide





151.
H3011D10-3
Lcp1
lymphocyte
H3011D10





cytosolic





protein 1





152.
H3052B11-3
Pctk3
PCTAIRE-
H3052B11





motif protein





kinase 3





153.
K0413H04-3
Anxa8
annexin A8
K0413H04





154.
H3054F05-3
Lyzs
lysozyme
H3054F05





155.
H3060F11-3
Cybb
cytochrome b-
H3060F11





245, beta





polypeptide





156.
H3012F08-3
9430068N19Rik
RIKEN cDNA
H3012F08





9430068N19





gene





157.
G0106B08-3
Abr
active BCR-
G0106B08





related gene





158.
L0287A12-3
Tdrkh
tudor and KH
L0287A12





domain





containing





protein





159.
H3083D01-3
AY007814
hypothetical
H3083D01





protein,





12H19.01.T7





160.
H3131F02-3
BG074151
ESTs
H3131F02





BG074151





161.
C0172H02-3
Lgals3
lectin, galactose
C0172H02





binding, soluble 3





162.
K0542E07-3
Cd44
CD44 antigen
K0542E07





163.
C0450H11-3
E430019N21Rik
RIKEN cDNA
C0450H11





E430019N21





gene





164.
K0216A08-3
Orc51
origin
K0216A08





recognition





complex,





subunit 5-like





(S. cerevisiae)





165.
H3122D03-3
Pdgfc
platelet-derived
H3122D03





growth factor,





C polypeptide





166.
C0037H07-3
Il13ra1
interleukin 13
C0037H07





receptor, alpha 1





167.
H3054F04-3
2610318I15Rik
RIKEN cDNA
H3054F04





2610318I15





gene





168.
L0908A12-3
Blnk
B-cell linker
L0908A12





169.
G0111E06-3
Car7
carbonic
G0111E06





anhydrase 7





170.
L0284B06-3
Ngfrap1
nerve growth
L0284B06





factor receptor





(TNFRSF16)





associated





protein 1





171.
K0145G06-3
Tcfec
transcription
K0145G06





factor EC





172.
H3001B08-3
Lyn
Yamaguchi
H3001B08





sarcoma viral





(v-yes-1)





oncogene





homolog





173.
G0117F12-3
Prkcsh
protein kinase
G0117F12





C substrate





80K-H





174.
C0903A11-3
2510004L01Rik
RIKEN cDNA
C0903A11





2510004L01





gene





175.
L0062C10-3
Rasa3
RAS p21
L0062C10





protein





activator 3





176.
L0939G09-3
Cd38
CD38 antigen
L0939G09





177.
H3115B07-3
S100a9
S100 calcium
H3115B07





binding protein





A9 (calgranulin





B)





178.
K0608H07-3
Fyb
FYN binding
K0608H07





protein





179.
C0104E07-3
Tcirg1
T-cell, immune
C0104E07





regulator 1





180.
K0431D02-3
Wisp1
WNT1
K0431D02





inducible





signaling





pathway protein 1





181.
L0837H10-3
Igfbp2
insulin-like
L0837H10





growth factor





binding protein 2





182.
C0159A08-3
Mta3
metastasis
C0159A08





associated 3





183.
K0649D06-3
Ms4a6b
membrane-
K0649D06





spanning 4-





domains,





subfamily A,





member 6B





184.
K0609D11-3
Man1a
mannosidase 1,
K0609D11





alpha





185.
C0907B04-3
Mcoln3
mucolipin 3
C0907B04





186.
H3020D08-3
Edem1
ER degradation
H3020D08





enhancer,





mannosidase





alpha-like 1





187.
J0039F05-3
Gdf3
growth
J0039F05





differentiation





factor 3





188.
C0906C11-3
BM218094
ESTs
C0906C11





BM218094





189.
L0266E10-3
B930060C03
hypothetical
L0266E10





protein





B930060C03





190.
H3060D11-3
Mll5
myeloid/lymphoid
H3060D11





or mixed-





lineage





leukemia 5





191.
L0062E01-3
Tnc
tenascin C
L0062E01





192.
K0132G08-3
AI662270
expressed
K0132G08





sequence





AI662270





193.
H3114D08-3
Arpc3
actin related
H3114D08





protein 2/3





complex,





subunit 3





194.
C0649E02-3
Unc93b
unc-93
C0649E02





homolog B (C. elegans)





195.
L0293H10-3
2510048K03Rik
RIKEN cDNA
L0293H10





2510048K03





gene





196.
H3024C03-3
1110008B24Rik
RIKEN cDNA
H3024C03





1110008B24





gene





197.
H3055G02-3
Ctsc
cathepsin C
H3055G02





198.
K0518A04-3
BM238476
ESTs
K0518A04





BM238476





199.
K0128H01-3
Parvg
parvin, gamma
K0128H01





200.
K0649F04-3
Ccr2
chemokine (C-
K0649F04





C) receptor 2





201.
K0603E03-3
Vav1
vav 1 oncogene
K0603E03





202.
K0649A02-3
Stat1
signal
K0649A02





transducer and





activator of





transcription 1





203.
H3013D11-3
Mt2
metallothionein 2
H3013D11





204.
H3013B02-3
Atp6v1b2
ATPase, H+
H3013B02





transporting,





V1 subunit B,





isoform 2





205.
L0541H09-3

transcribed
L0541H09





sequence with





weak similarity





to protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





206.
K0516E03-3


Mus musculus

K0516E03





12 days embryo





embryonic





body between





diaphragm





region and neck





cDNA, RIKEN





full-length





enriched





library,





clone: 9430012B12





product: unknown





EST, full





insert sequence.





207.
H3034A10-3
Plaur
urokinase
H3034A10





plasminogen





activator





receptor





208.
C0910G05-3
BM218419
ESTs
C0910G05





BM218419





209.
C0262H12-3
Msh2
mutS homolog
C0262H12





2 (E. coli)





210.
H3078C11-3
BG069620
ESTs
H3078C11





BG069620





211.
L0926H09-3
6030440G05Rik
RIKEN cDNA
L0926H09





6030440G05





gene





212.
J0076H03-3

C80125 Mouse
J0076H03





3.5-dpc





blastocyst





cDNA Mus






musculus






cDNA clone





J0076H03 3′,





MRNA





sequence





213.
L0817B08-3

transcribed
L0817B08





sequence with





strong





similarity to





protein





sp: P00722 (E. coli)





BGAL_ECOLI





Beta-





galactosidase





(Lactase)





214.
H3065D11-3
Crnkl1
Crn, crooked
H3065D11





neck-like 1





(Drosophila)





215.
H3157E02-3
5630401J11Rik
RIKEN cDNA
H3157E02





5630401J11





gene





216.
H3007C11-3
BG063444
ESTs
H3007C11





BG063444





217.
K0517E07-3
C530050H10Rik
RIKEN cDNA
K0517E07





C530050H10





gene





218.
H3150B11-5
Ptpn2
protein tyrosine
H3150B11





phosphatase,





non-receptor





type 2





219.
C0199C01-3
9930104E21Rik
RIKEN cDNA
C0199C01





9930104E21





gene





220.
H3063A09-3
Rassf5
Ras association
H3063A09





(RalGDS/AF-6)





domain family 5





221.
K0445A07-3
Hfe
hemochromatosis
K0445A07





222.
H3123G07-3
C630007C17Rik
RIKEN cDNA
H3123G07





C630007C17





gene





223.
H3094C03-3
Baz1a
bromodomain
H3094C03





adjacent to zinc





finger domain





1A





224.
L0845H04-3
BM117070
ESTs
L0845H04





BM117070





225.
C0161F01-3
BC010311
cDNA sequence
C0161F01





BC010311





226.
H3034E07-3
BG065726
ESTs
H3034E07





BG065726





227.
J0419G11-3
Cldn8
claudin 8
J0419G11





228.
C0040C08-3
Cxcr4
chemokine
C0040C08





(C—X—C motif)





receptor 4





229.
K0612H02-3
BM241159
ESTs
K0612H02





BM241159





230.
J0460B09-3

AU024759
J0460B09





Mouse





unfertilized egg





cDNA Mus






musculus






cDNA clone





J0460B09 3′,





MRNA





sequence





231.
H3103F07-3


Mus musculus

H3103F07





transcribed





sequence with





weak similarity





to protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





232.
H3079H09-3
BG069769
ESTs
H3079H09





BG069769





233.
H3130D06-3
BG074061
ESTs
H3130D06





BG074061





234.
H3071D08-3
Lcp2
lymphocyte
H3071D08





cytosolic





protein 2





235.
K0218E07-3


Mus musculus

K0218E07





10 days neonate





olfactory brain





cDNA, RIKEN





full-length





enriched





library,





clone: E530016P10





product: weakly





similar to





ONCOGENE





TLM [Mus






musculus], full






insert sequence.





236.
C0907H07-3
BM218221
ESTs
C0907H07





BM218221





237.
K0605B09-3
BM240642
ESTs
K0605B09





BM240642





238.
C0322F05-3
Eya3
eyes absent 3
C0322F05





homolog





(Drosophila)





239.
J0004A01-3
C76123
ESTs C76123
J0004A01





240.
K0139H06-3
BM223668
ESTs
K0139H06





BM223668





241.
L0941F06-3
BM120591
ESTs
L0941F06





BM120591





242.
C0300G03-3
3021401C12Rik
RIKEN cDNA
C0300G03





3021401C12





gene





243.
C0925E03-3

transcribed
C0925E03





sequence with





moderate





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





244.
H3083B07-5
BG082983
ESTs
H3083B07





BG082983





245.
H3056F01-3
Gdf9
growth
H3056F01





differentiation





factor 9





246.
J0259A06-3
C88243
EST C88243
J0259A06





247.
C0124B09-3
BC042513
cDNA sequence
C0124B09





BC042513





248.
L0933E02-3

L0933E02-3
L0933E02





NIA Mouse





Newborn





Kidney cDNA





Library (Long)






Mus musculus






cDNA clone





L0933E02 3′,





MRNA





sequence





249.
H3072B12-3
BG069052
ESTs
H3072B12





BG069052





250.
L0266C03-3
D930020B18Rik
RIKEN cDNA
L0266C03





D930020B18





gene





251.
K0423B04-3
Zfp91
zinc finger
K0423B04





protein 91





252.
J0403C04-3

AU021859
J0403C04





Mouse





unfertilized egg





cDNA Mus






musculus






cDNA clone





J0403C04 3′,





MRNA





sequence





253.
J0248E12-3
1700011I03Rik
RIKEN cDNA
J0248E12





1700011I03





gene





254.
J0908H04-3
Rpl24
ribosomal
J0908H04





protein L24





255.
K0205H10-3
Madd
MAP-kinase
K0205H10





activating death





domain





256.
C0507E09-3
Gpr22
G protein-
C0507E09





coupled





receptor 22





257.
J0005B11-3


Mus musculus

J0005B11





transcribed





sequence with





weak similarity





to protein





ref: NP_083358.1





(M. musculus)





RIKEN cDNA





5830411J07





[Mus musculus]





258.
L0201E08-3
AW551705
ESTs
L0201E08





AW551705





259.
J0426H03-3
AU023164
ESTs
J0426H03





AU023164





260.
C0649D06-3
Cdkn2b
cyclin-
C0649D06





dependent





kinase inhibitor





2B (p15,





inhibits CDK4)





261.
J0421D03-3
Rpl24
ribosomal
J0421D03





protein L24





262.
K0643F07-3

ESTs
K0643F07





BQ563001





263.
H3103C12-3
Slamf1
signaling
H3103C12





lymphocytic





activation





molecule





family member 1





264.
J0416H11-3
Pscdbp
pleckstrin
J0416H11





homology, Sec7





and coiled-coil





domains,





binding protein





265.
AF015770.1
Rfng
radical fringe
AF015770





gene homolog





(Drosophila)





266.
C0933C05-3

ESTs
C0933C05





BQ551952





267.
C0931A05-3
E130304F04Rik
RIKEN cDNA
C0931A05





E130304F04





gene





268.
J0030C02-3
C77383
ESTs C77383
J0030C02





269.
H3061A07-3
Srpk2
serine/arginine-
H3061A07





rich protein





specific kinase 2





270.
J0823B08-3

AU041035
J0823B08





Mouse four-





cell-embryo





cDNA Mus






musculus






cDNA clone





J0823B08 3′,





MRNA





sequence





271.
L0942H08-3


Mus musculus

L0942H08





transcribed





sequence with





moderate





similarity to





protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





272.
C0280H06-3
Mrp150
mitochondrial
C0280H06





ribosomal





protein L50





273.
L0534E07-3
4632417D23
hypothetical
L0534E07





protein





4632417D23





274.
U22339.1
Il15ra
interleukin 15
U22339





receptor, alpha





chain





275.
L0533C12-3

L0533C12-3
L0533C12





NIA Mouse





Newborn Heart





cDNA Library






Mus musculus






cDNA clone





L0533C12 3′,





MRNA





sequence





276.
C0909E04-3
Mvk
mevalonate
C0909E04





kinase





277.
J0093B09-3
Bhmt2
betaine-
J0093B09





homocysteine





methyltransferase 2





278.
H3066D09-3
BG068517
ESTs
H3066D09





BG068517





279.
C0346F01-3
BM197260
ESTs
C0346F01





BM197260





280.
K0125A06-3
Hdac7a
histone
K0125A06





deacetylase 7A





281.
J0214H07-3

C85807 Mouse
J0214H07





fertilized one-





cell-embryo





cDNA Mus






musculus






cDNA clone





J0214H07 3′,





MRNA





sequence





282.
C0309H10-3
5930412E23Rik
RIKEN cDNA
C0309H10





5930412E23





gene





283.
C0351C04-3
2610034E13Rik
RIKEN cDNA
C0351C04





2610034E13





gene





284.
K0204G07-3
Arf3
ADP-
K0204G07





ribosylation





factor 3





285.
L0928B09-3

transcribed
L0928B09





sequence with





strong





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





286.
H3059A09-3
C430004E15Rik
RIKEN cDNA
H3059A09





C430004E15





gene





287.
C0949D03-3

UNKNOWN
C0949D03





C0949D03





288.
K0118A04-3
Rgs1
regulator of G-
K0118A04





protein





signaling 1





289.
H3123F11-3

transcribed
H3123F11





sequence with





moderate





similarity to





protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





290.
H3154A06-3
Gng13
guanine
H3154A06





nucleotide





binding protein





13, gamma





291.
L0534E01-3

L0534E01-3
L0534E01





NIA Mouse





Newborn Heart





cDNA Library






Mus musculus






cDNA clone





L0534E01 3′,





MRNA





sequence





292.
L0250B10-3
Ap4m1
adaptor-related
L0250B10





protein





complex AP-4,





mu 1





293.
L0518G04-3
BM123045
ESTs
L0518G04





BM123045





294.
J1020E03-3

transcribed
J1020E03





sequence with





moderate





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





295.
X12616.1
Fes
feline sarcoma
X12616





oncogene





296.
J0026H02-3
C77164
expressed
J0026H02





sequence





C77164





297.
H3154D11-5
Taf7l
TAF7-like
H3154D11





RNA





polymerase II,





TATA box





binding protein





(TBP)-





associated





factor





298.
H3054H04-3
Kcnn4
potassium
H3054H04





intermediate/small





conductance





calcium-





activated





channel,





subfamily N,





member 4





299.
J0425B03-3
R75183
expressed
J0425B03





sequence





R75183





300.
C0930C02-3
0610037D15Rik
RIKEN cDNA
C0930C02





0610037D15





gene





301.
L0812A11-3

ESTs BI793430
L0812A11





302.
J0243F04-3
9530020D24Rik
RIKEN cDNA
J0243F04





9530020D24





gene





303.
C0335A03-3
1110035O14Rik
RIKEN cDNA
C0335A03





1110035O14





gene





304.
H3003B10-3
BG063111
ESTs
H3003B10





BG063111





305.
U97073.1
Prtn3
proteinase 3
U97073





306.
K0300D08-3
Afmid
arylformamidase
K0300D08





307.
H3029H06-3
Sf3b2
splicing factor
H3029H06





3b, subunit 2





308.
H3074D09-3
Drg2
developmentally
H3074D09





regulated





GTP binding





protein 2





309.
K0647G12-3
Plek
pleckstrin
K0647G12





310.
H3137A08-3


Mus musculus

H3137A08





transcribed





sequence with





moderate





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





311.
C0166D06-3
Slc38a3
solute carrier
C0166D06





family 38,





member 3





312.
K0406B07-3
Sirt7
sirtuin 7 (silent
K0406B07





mating type





information





regulation 2,





homolog) 7 (S. cerevisiae)





313.
H3085D10-3
Gda
guanine
H3085D10





deaminase





314.
H3099C09-3
Igf1
insulin-like
H3099C09





growth factor 1





315.
H3099B07-5
2610028H24Rik
RIKEN cDNA
H3099B07





2610028H24





gene





316.
H3114H10-3
Rec8L1
REC8-like 1
H3114H10





(yeast)





317.
L0703E03-3
Lipc
lipase, hepatic
L0703E03





318.
H3074H08-3
BG069302
ESTs
H3074H08





BG069302





319.
K0443D01-3
Baz1b
bromodomain
K0443D01





adjacent to zinc





finger domain,





1B





320.
J0409E10-3
AU022163
ESTs
J0409E10





AU022163





321.
L0528E01-3
BM123655
EST
L0528E01





BM123655





322.
L0031B11-3
Alcam
activated
L0031B11





leukocyte cell





adhesion





molecule





323.
G0115A06-3
Fem1a
feminization 1
G0115A06





homolog a (C. elegans)





324.
L0947C07-3
Mal
myelin and
L0947C07





lymphocyte





protein, T-cell





differentiation





protein





325.
H3101A05-3
AU040576
expressed
H3101A05





sequence





AU040576





326.
H3064E10-3
BG068353
ESTs
H3064E10





BG068353





327.
K0505H05-3
Ian6
immune
K0505H05





associated





nucleotide 6





328.
H3082E12-3
Ptpre
protein tyrosine
H3082E12





phosphatase,





receptor type, E





329.
H3088A06-3
2310047N01Rik
RIKEN cDNA
H3088A06





2310047N01





gene





330.
K0635B07-3
Ccr5
chemokine (C-
K0635B07





C motif)





receptor 5





331.
C0153A12-3
1110025F24Rik
RIKEN cDNA
C0153A12





1110025F24





gene





332.
C0143E02-3
BC022145
cDNA sequence
C0143E02





BC022145





333.
L0863F12-3
Nr2c2
nuclear receptor
L0863F12





subfamily 2,





group C,





member 2





334.
H3045F02-3
LOC214424
hypothetical
H3045F02





protein





LOC214424





335.
H3035G05-3
BG065832
ESTs
H3035G05





BG065832





336.
H3137D02-3
Hnrpl
heterogeneous
H3137D02





nuclear





ribonucleoprotein L





337.
H3097F07-3
AU040829
expressed
H3097F07





sequence





AU040829





338.
J0029C02-3
Frag1-pending
FGF receptor
J0029C02





activating





protein 1





339.
BB416014.1


Mus musculus

BB416014





B6-derived





CD11 +ve





dendritic cells





cDNA, RIKEN





full-length





enriched





library,





clone: F730035A01





product: similar





to SWI/SNF





COMPLEX





170 KDA





SUBUNIT





[Homo






sapiens], full






insert sequence.





340.
H3087E01-3
Anxa4
annexin A4
H3087E01





341.
H3088E08-3
BG070548
ESTs
H3088E08





BG070548





342.
AF179424.1


Mus musculus

AF179424





13 days embryo





male testis





cDNA, RIKEN





full-length





enriched





library,





clone: 6030408M17





product: GATA





binding protein





4, full insert





sequence





343.
J0258C01-3


Mus musculus

J0258C01





mRNA for





mKIAA1335





protein





344.
K0507B09-3

ESTs
K0507B09





BM238095





345.
L0846F07-3
BM117131
ESTs
L0846F07





BM117131





346.
U48866.1
CEBPE
CCAAT/enhancer
U48866





binding





protein





(C/EBP),





epsilon





347.
K0301B06-3
Fech
ferrochelatase
K0301B06





348.
NM_009756.1
Bmp10
bone
NM_009756





morphogenetic





protein 10





349.
NM_010100.1
Edar
ectodysplasin-A
NM_010100





receptor





350.
G0115E06-3
C430014D17Rik
RIKEN cDNA
G0115E06





C430014D17





gene





351.
L0266D11-3
Ppp3ca
protein
L0266D11





phosphatase 3,





catalytic





subunit, alpha





isoform





352.
L0526F10-3


Mus musculus

L0526F10





10 days neonate





cortex cDNA,





RIKEN full-





length enriched





library,





clone: A830020C21





product: unknown





EST, full





insert sequence.





353.
H3047C10-3
Slc6a6
solute carrier
H3047C10





family 6





(neurotransmitter





transporter,





taurine),





member 6





354.
K0322G06-3
BC042620
cDNA sequence
K0322G06





BC042620





355.
NM_009580.1
Zp1
zona pellucida
NM_009580





glycoprotein 1





356.
H3150E08-3
Map4k5
mitogen-
H3150E08





activated





protein kinase





kinase kinase





kinase 5





357.
J0059G03-3
C79059
ESTs C79059
J0059G03





358.
U93191.1
Hdac2
histone
U93191





deacetylase 2





359.
H3033C04-5

H3033C04-5
H3033C04





NIA Mouse





15K cDNA





Clone Set Mus






musculus






cDNA clone





H3033C04 5′,





MRNA





sequence





360.
H3085C01-3
2700038N03Rik
RIKEN cDNA
H3085C01





2700038N03





gene





361.
J0412G02-3
BB336629
ESTs
J0412G02





BB336629





362.
K0527H09-3
BM239048
ESTs
K0527H09





BM239048





363.
H3009C10-3
Serpinb9b
serine (or
H3009C10





cysteine)





proteinase





inhibitor, clade





B, member 9b





364.
H3142D11-3


Mus musculus

H3142D11





mRNA similar





to hypothetical





protein





FLJ20811





(cDNA clone





MGC: 27863





IMAGE: 3492516),





complete





cds





365.
H3094B07-3


Mus musculus

H3094B07





transcribed





sequence with





weak similarity





to protein





sp: P11369





(M. musculus)





POL2_MOUSE





Retrovirus-





related POL





polyprotein





[Contains:





Reverse





transcriptase;





Endonuclease]





366.
J0068F09-3
C79588
ESTs C79588
J0068F09





367.
H3039B03-5
E030024M05Rik
RIKEN cDNA
H3039B03





E030024M05





gene





368.
H3068B03-3
BG068673
ESTs
H3068B03





BG068673





369.
C0250F05-3
BM203195
ESTs
C0250F05





BM203195





370.
H3110C11-3
Mlph
melanophilin
H3110C11





371.
H3121F01-3
Wnt4
wingless-
H3121F01





related MMTV





integration site 4





372.
J1012G09-3
Brd3
bromodomain
J1012G09





containing 3





373.
L0952B09-3
Usp49
ubiquitin
L0952B09





specific





protease 49





374.
K0131B12-3
Il4ra
interleukin 4
K0131B12





receptor, alpha





375.
H3046E09-3
Nfatc2ip
nuclear factor
H3046E09





of activated T-





cells,





cytoplasmic 2





interacting





protein





376.
K0520B05-3

transcribed
K0520B05





sequence with





weak similarity





to protein





pir: I58401





(M. musculus)





I58401 protein-





tyrosine kinase





(EC 2.7.1.112)





JAK3 - mouse





377.
K0315G05-3
Stat5a
signal
K0315G05





transducer and





activator of





transcription





5A





378.
H3086F07-3
BC003332
cDNA sequence
H3086F07





BC003332





379.
H3156A10-5
Ctsd
cathepsin D
H3156A10





380.
C0890D02-3

C0890D02-3
C0890D02





NIA Mouse





Blastocyst





cDNA Library





(Long) Mus






musculus






cDNA clone





C0890D02 3′,





MRNA





sequence





381.
L0245G03-3
6430519N07Rik
RIKEN cDNA
L0245G03





6430519N07





gene





382.
J0447A10-3


Mus musculus

J0447A10





cDNA clone





IMAGE: 12820





81, partial cds





383.
J1031A09-3


Mus musculus

J1031A09





transcribed





sequence with





weak similarity





to protein





pir: I58401





(M. musculus)





I58401 protein-





tyrosine kinase





(EC 2.7.1.112)





JAK3 - mouse





384.
L0072H04-3
A630084M22Rik
RIKEN cDNA
L0072H04





A630084M22





gene





385.
J0050E03-3

transcribed
J0050E03





sequence with





weak similarity





to protein





ref: NP_081764.





1 (M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





386.
H3039C11-3
Tyro3
TYRO3 protein
H3039C11





tyrosine kinase 3





387.
C0324F11-3
6720458F09Rik
RIKEN cDNA
C0324F11





6720458F09





gene





388.
L0018F11-3
AW547199
ESTs
L0018F11





AW547199





389.
X69902.1
Itga6
integrin alpha 6
X69902





390.
H3105A09-3

transcribed
H3105A09





sequence with





weak similarity





to protein





ref: NP_416488.





1 (E. coli)





putative





transport





protein,





shikimate





[Escherichia






coli K12]






391.
H3159F01-5

UNKNOWN
H3159F01





H3159F01





392.
K0522B04-3
F5
coagulation
K0522B04





factor V





393.
C0123F08-3
AI843918
expressed
C0123F08





sequence





AI843918





394.
H3067G08-3
BG068642
ESTs
H3067G08





BG068642





395.
K0349B03-3
Stam2
signal
K0349B03





transducing





adaptor





molecule (SH3





domain and





ITAM motif) 2





396.
C0620D11-3
Bid
BH3 interacting
C0620D11





domain death





agonist





397.
C0189H10-3
4930486L24Rik
RIKEN cDNA
C0189H10





4930486L24





gene





398.
H3140A02-3
Slc9a1
solute carrier
H3140A02





family 9





(sodium/hydrogen





exchanger),





member 1





399.
K0645B04-3
Smc4l1
SMC4
K0645B04





structural





maintenance of





chromosomes





4-like 1 (yeast)





400.
C0300G08-3
6720460I06Rik
RIKEN cDNA
C0300G08





6720460I06





gene





401.
M59378.1
Tnfrsf1b
tumor necrosis
M59378





factor receptor





superfamily,





member 1b





402.
NM_009399.1
Tnfrsf11a
tumor necrosis
NM_009399





factor receptor





superfamily,





member 11a





403.
C0168E12-3
2810442I22Rik
RIKEN cDNA
C0168E12





2810442I22





gene





404.
L0228H10-3
C1r
complement
L0228H10





component 1, r





subcomponent





405.
H3088B10-3
BG070515
ESTs
H3088B10





BG070515





406.
K0409D10-3
Lrrc5
leucine-rich
K0409D10





repeat-





containing 5





407.
H3056D02-3

transcribed
H3056D02





sequence with





moderate





similarity to





protein





ref: NP_079108.1





(H. sapiens)





hypothetical





protein





FLJ22439





[Homo sapiens]





408.
J0430F08-3
AU023357
ESTs
J0430F08





AU023357





409.
H3158C06-3
2810457I06Rik
RIKEN cDNA
H3158C06





2810457I06





gene





410.
M85078.1
Csf2ra
colony
M85078





stimulating





factor 2





receptor, alpha,





low-affinity





(granulocyte-





macrophage)





411.
C0145E06-3
Satb1
special AT-rich
C0145E06





sequence





binding protein 1





412.
H3015B08-3
BG064069
ESTs
H3015B08





BG064069





413.
C0842H05-3
Fbln1
fibulin 1
C0842H05





414.
G0117D07-3
Otx2
orthodenticle
G0117D07





homolog 2





(Drosophila)





415.
L0806E03-3
Stmn4
stathmin-like 4
L0806E03





416.
H3073B06-3
BG069137
ESTs
H3073B06





BG069137





417.
H3082G08-3
Myo10
myosin X
H3082G08





418.
C0141F07-3
C3ar1
complement
C0141F07





component 3a





receptor 1





419.
K0525G09-3
5830411I20
hypothetical
K0525G09





protein





5830411I20





420.
H3064D01-3

transcribed
H3064D01





sequence with





weak similarity





to protein





ref: NP_001362.1





(H. sapiens)





dynein,





axonemal,





heavy





polypeptide 8





[Homo sapiens]





421.
C0120F08-3
6330406L22Rik
RIKEN cDNA
C0120F08





6330406L22





gene





422.
H3105G04-3
Map4k4
mitogen-
H3105G04





activated





protein kinase





kinase kinase





kinase 4





423.
J0800D09-3
2310004L02Rik
RIKEN cDNA
J0800D09





2310004L02





gene





424.
L0226H02-3
5830411I20
hypothetical
L0226H02





protein





5830411I20





425.
L0529D10-3
BM123730
ESTs
L0529D10





BM123730





426.
H3088E05-3
Gla
galactosidase,
H3088E05





alpha





427.
K0621H11-3

K0621H11-3
K0621H11





NIA Mouse





Hematopoietic





Stem Cell (Lin-/





c-Kit-/Sca-1+)





cDNA Library





(Long) Mus






musculus






cDNA clone





NIA: K0621H11





IMAGE: 30070846





3′, MRNA





sequence





428.
C0846H03-3
D330025I23Rik
RIKEN cDNA
C0846H03





D330025I23





gene





429.
J0058E06-3
C78984
ESTs C78984
J0058E06





430.
K0325E09-3
Ibsp
integrin binding
K0325E09





sialoprotein





431.
K0336F07-3
Pycs
pyrroline-5-
K0336F07





carboxylate





synthetase





(glutamate





gamma-





semialdehyde





synthetase)





432.
H3013B04-3
B230106I24Rik
RIKEN cDNA
H3013B04





B230106I24





gene





433.
L0238A07-3
Midn
midnolin
L0238A07





434.
L0929C04-3
Tnfrsf11b
tumor necrosis
L0929C04





factor receptor





superfamily,





member 11b





(osteoprotegerin)





435.
L0020F05-3
6330583M11Rik
RIKEN cDNA
L0020F05





6330583M11





gene





436.
H3012H07-3
Cd44
CD44 antigen
H3012H07





437.
K0240E11-3
Myo5a
myosin Va
K0240E11





438.
K0401C06-3
Col8a1
procollagen,
K0401C06





type VIII, alpha 1





439.
C0917F02-3
Frzb
frizzled-related
C0917F02





protein





440.
H3104C03-3
1500015O10Rik
RIKEN cDNA
H3104C03





1500015O10





gene





441.
K0438D09-3
Col8a1
procollagen,
K0438D09





type VIII, alpha 1





442.
H3152C04-3
Usp16
ubiquitin
H3152C04





specific





protease 16





443.
H3079D12-3
Pld3
phospholipase
H3079D12





D3





444.
L0020E08-3
C1qg
complement
L0020E08





component 1, q





subcomponent,





gamma





polypeptide





445.
J0025G01-3
Yars
tyrosyl-tRNA
J0025G01





synthetase





446.
L0832H09-3
Mafb
v-maf
L0832H09





musculoaponeurotic





fibrosarcoma





oncogene





family, protein





B (avian)





447.
C0451C02-3
2700094L05Rik
RIKEN cDNA
C0451C02





2700094L05





gene





448.
H3063A08-3
Lgmn
legumain
H3063A08





449.
K0629D05-3
Evi2a
ecotropic viral
K0629D05





integration site





2a





450.
G0111D11-3
Ctsl
cathepsin L
G0111D11





451.
H3077D05-3
Npc2
Niemann Pick
H3077D05





type C2





452.
G0104C04-3
Dab2
disabled
G0104C04





homolog 2





(Drosophila)





453.
L0502D10-3
Pla1a
phospholipase
L0502D10





A1 member A





454.
H3126B08-3
Pla2g7
phospholipase
H3126B08





A2, group VII





(platelet-





activating factor





acetylhydrolase,





plasma)





455.
J0034A07-3
Creg
cellular
J0034A07





repressor of





E1A-stimulated





genes





456.
H3114B07-3
Slc12a4
solute carrier
H3114B07





family 12,





member 4





457.
K0339H12-3
Thbs1
thrombospondin 1
K0339H12





458.
H3028C09-3
Adk
adenosine
H3028C09





kinase





459.
L0277B06-3
Psap
prosaposin
L0277B06





460.
H3013F05-3
Sdc1
syndecan 1
H3013F05





461.
H3084A06-3
Spin
spindlin
H3084A06





462.
H3077F04-3
Osbpl8
oxysterol
H3077F04





binding protein-





like 8





463.
K0324A06-3
Itga11
integrin, alpha
K0324A06





11





464.
C0115E05-3
2010110K16Rik
RIKEN cDNA
C0115E05





2010110K16





gene





465.
C0668G11-3
Fabp5
fatty acid
C0668G11





binding protein





5, epidermal





466.
L0030A03-3
Alox5ap
arachidonate 5-
L0030A03





lipoxygenase





activating





protein





467.
H3009E11-3
Socs3
suppressor of
H3009E11





cytokine





signaling 3





468.
L0010B01-3
Abca1
ATP-binding
L0010B01





cassette, sub-





family A





(ABC1),





member 1





469.
G0116C07-3
Ctsb
cathepsin B
G0116C07





470.
K0426E09-3
Eps8
epidermal
K0426E09





growth factor





receptor





pathway





substrate 8





471.
H3102F08-3
Asah1
N-
H3102F08





acylsphingosine





amidohydrolase 1





472.
L0825G08-3
Dcamkl1
double cortin
L0825G08





and





calcium/calmodulin-





dependent





protein kinase-





like 1





473.
K0306B10-3
Fgf7
fibroblast
K0306B10





growth factor 7





474.
H3127F04-3
Chst11
carbohydrate
H3127F04





sulfotransferase





11





475.
L0208A08-3
1200013B22Rik
RIKEN cDNA
L0208A08





1200013B22





gene





476.
H3026G09-3
Col2a1
procollagen,
H3026G09





type II, alpha 1





477.
C0218D02-3
Madh1
MAD homolog
C0218D02





1 (Drosophila)





478.
J1031F04-3
Dfna5h
deafness,
J1031F04





autosomal





dominant 5





homolog





(human)





479.
L0276A08-3
Rai14
retinoic acid
L0276A08





induced 14





480.
C0508H08-3
Sptlc2
serine
C0508H08





palmitoyltransferase,





long





chain base





subunit 2





481.
J0042D09-3
C78076
ESTs C78076
J0042D09





482.
J0013B06-3
Akr1b8
aldo-keto
J0013B06





reductase





family 1,





member B8





483.
H3158D11-3
Mmp2
matrix
H3158D11





metalloproteinase 2





484.
H3001D04-3
Hist2h3c2
histone 2, H3c2
H3001D04





485.
C0664G04-3
Ppicap
peptidylprolyl
C0664G04





isomerase C-





associated





protein





486.
H3091E10-3
Nupr1
nuclear protein 1
H3091E10





487.
X98792.1
Ptgs2
prostaglandin-
X98792





endoperoxide





synthase 2





488.
L0908B12-3
Ptpn1
protein tyrosine
L0908B12





phosphatase,





non-receptor





type 1





489.
H3081D02-3
Bok
Bcl-2-related
H3081D02





ovarian killer





protein





490.
C0127E12-3
Cln5
ceroid-
C0127E12





lipofuscinosis,





neuronal 5





491.
K0310G10-3
Col5a2
procollagen,
K0310G10





type V, alpha 2





492.
H3023H09-3
Ftl1
ferritin light
H3023H09





chain 1





493.
G0104B11-3
Slc7a7
solute carrier
G0104B11





family 7





(cationic amino





acid transporter,





y+ system),





member 7





494.
C0123F05-3
B4galt5
UDP-
C0123F05





Gal:betaGlcNAc





beta 1,4-





galactosyltransferase,





polypeptide 5





495.
H3082D01-3
1810015C04Rik
RIKEN cDNA
H3082D01





1810015C04





gene





496.
C0121E07-3
AW539579
EST
C0121E07





AW539579





497.
H3153H08-3
Hs6st2
heparan sulfate
H3153H08





6-O-





sulfotransferase 2





498.
J0238C08-3
4930579A11Rik
RIKEN cDNA
J0238C08





4930579A11





gene





499.
L0942B10-3
Msr2
macrophage
L0942B10





scavenger





receptor 2





500.
J0915B05-3
Cdca1
cell division
J0915B05





cycle associated 1





501.
H3058B09-3
Lypla3
lysophospholipase 3
H3058B09





502.
C0197E01-3
D630023B12
hypothetical
C0197E01





protein





D630023B12





503.
J0802G04-3
0610011I04Rik
RIKEN cDNA
J0802G04





0610011I04





gene





504.
H3039E08-3
Sh3d3
SH3 domain
H3039E08





protein 3





505.
L0210A08-3
B130023O14Rik
RIKEN cDNA
L0210A08





B130023O14





gene





506.
H3114C10-3
Ppgb
protective
H3114C10





protein for beta-





galactosidase





507.
C0322A01-3
2810441C07Rik
RIKEN cDNA
C0322A01





2810441C07





gene





508.
L0256F11-3
Adfp
adipose
L0256F11





differentiation





related protein





509.
L0939H06-3
Mgat5
mannoside
L0939H06





acetylglucosaminyltransferase 5





510.
C0503B05-3
Dcamkl1
double cortin
C0503B05





and





calcium/calmodulin-





dependent





protein kinase-





like 1





511.
H3136H11-3
Map4k5
mitogen-
H3136H11





activated





protein kinase





kinase kinase





kinase 5





512.
K0349A04-3
Fn1
fibronectin 1
K0349A04





513.
C0177C04-3
Ctsz
cathepsin Z
C0177C04





514.
C0668D08-3
Grn
granulin
C0668D08





515.
C0106D12-3
Anxa1
annexin A1
C0106D12





516.
H3078E09-3
Hexb
hexosaminidase B
H3078E09





517.
L0033F05-3
2810442I22Rik
RIKEN cDNA
L0033F05





2810442I22





gene





518.
K0144G04-3
Ifi203
interferon
K0144G04





activated gene





203





519.
H3144E05-3
4933426M11Rik
RIKEN cDNA
H3144E05





4933426M11





gene





520.
K0336D02-3
Ifi16
interferon,
K0336D02





gamma-





inducible





protein 16





521.
H3004B12-3
Hpn
hepsin
H3004B12





522.
K0617G07-3
Atp6v1b2
ATPase, H+
K0617G07





transporting,





V1 subunit B,





isoform 2





523.
L0849B10-3
Pltp
phospholipid
L0849B10





transfer protein





524.
L0019H03-3
Fn1
fibronectin 1
L0019H03





525.
J0099E12-3
Slc6a6
solute carrier
J0099E12





family 6





(neurotransmitter





transporter,





taurine),





member 6





526.
J0023G04-3
BC004044
cDNA sequence
J0023G04





BC004044





527.
C0913D04-3
4933433D23Rik
RIKEN cDNA
C0913D04





4933433D23





gene





528.
H3020C02-3
Mt1
metallothionein 1
H3020C02





529.
C0217B11-3
Sema4d
sema domain,
C0217B11





immunoglobulin





domain (Ig),





transmembrane





domain (TM)





and short





cytoplasmic





domain,





(semaphorin)





4D





530.
C0917E01-3
Bhlhb2
basic helix-
C0917E01





loop-helix





domain





containing,





class B2





531.
H3132B12-5
Deaf1
deformed
H3132B12





epidermal





autoregulatory





factor 1





(Drosophila)





532.
L0270C04-3
Mpp1
membrane
L0270C04





protein,





palmitoylated





533.
J0709H10-3

transcribed
J0709H10





sequence with





moderate





similarity to





protein





pir: A38712





(H. sapiens)





A38712





fibrillarin





[validated] -





human





534.
C0166A10-3
Car2
carbonic
C0166A10





anhydrase 2





535.
L0511A03-3
BM122519
ESTs
L0511A03





BM122519





536.
H3029F09-3
Atp6v1e1
ATPase, H+
H3029F09





transporting,





V1 subunit E





isoform 1





537.
J0716H11-3
Kdt1
kidney cell line
J0716H11





derived





transcript 1





538.
C0102C01-3
Acp5
acid
C0102C01





phosphatase 5,





tartrate resistant





539.
C0641C07-3
Pdgfb
platelet derived
C0641C07





growth factor,





B polypeptide





540.
C0147C09-3
Ttc7
tetratricopeptide
C0147C09





repeat domain 7





541.
K0301G02-3
9430025M21Rik
RIKEN cDNA
K0301G02





9430025M21





gene





542.
H3022D05-3
Tpbpb
trophoblast
H3022D05





specific protein





beta





543.
H3007C09-3
Sh3bgrl3
SH3 domain
H3007C09





binding





glutamic acid-





rich protein-like 3





544.
L0820G02-3
Igsf4
immunoglobulin
L0820G02





superfamily,





member 4





545.
C0120H11-3
4933433D23Rik
RIKEN cDNA
C0120H11





4933433D23





gene





546.
J1016E08-3
1810046J19Rik
RIKEN cDNA
J1016E08





1810046J19





gene





547.
L0822D10-3
Prkcb
protein kinase
L0822D10





C, beta





548.
H3050H09-3
Ppp2r5c
protein
H3050H09





phosphatase 2,





regulatory





subunit B





(B56), gamma





isoform





549.
J0442H09-3


Mus musculus

J0442H09





hypothetical





LOC237436





(LOC237436),





mRNA





550.
H3141E06-3
Sra1
steroid receptor
H3141E06





RNA activator 1





551.
C0170H06-3
Adss2
adenylosuccinate
C0170H06





synthetase 2,





non muscle





552.
K0344C08-3
Emp1
epithelial
K0344C08





membrane





protein 1





553.
J0907F03-3
Npl
N-
J0907F03





acetylneuraminate





pyruvate





lyase





554.
J1008C10-3
Ptpn1
protein tyrosine
J1008C10





phosphatase,





non-receptor





type 1





555.
K0103F09-3
2500002K03Rik
RIKEN cDNA
K0103F09





2500002K03





gene





556.
C0837H01-3
Adam9
a disintegrin
C0837H01





and





metalloproteinase





domain 9





(meltrin





gamma)





557.
J0207H07-3
Runx2
runt related
J0207H07





transcription





factor 2





558.
J0246C10-3
Tpd52
tumor protein
J0246C10





D52





559.
H3158E12-3
BC003324
cDNA sequence
H3158E12





BC003324





560.
H3094A04-3
Dnajc3
DnaJ (Hsp40)
H3094A04





homolog,





subfamily C,





member 3





561.
L0231F01-3
Evl
Ena-vasodilator
L0231F01





stimulated





phosphoprotein





562.
K0512E10-3
Myo5a
myosin Va
K0512E10





563.
K0608H09-3
Ptprc
protein tyrosine
K0608H09





phosphatase,





receptor type, C





564.
L0842E04-3
Prkcb
protein kinase
L0842E04





C, beta





565.
H3121G01-3
BG073361
ESTs
H3121G01





BG073361





566.
C0947F04-3
5830411K21Rik
RIKEN cDNA
C0947F04





5830411K21





gene





567.
H3009D03-5
Plac8
placenta-
H3009D03





specific 8





568.
H3132E07-3
Lxn
latexin
H3132E07





569.
H3054C01-3
Nr2e3
nuclear receptor
H3054C01





subfamily 2,





group E,





member 3





570.
H3013H03-3
Man1a
mannosidase 1,
H3013H03





alpha





571.
J0058F02-3
ank
progressive
J0058F02





ankylosis





572.
L0829D10-3
Snca
synuclein, alpha
L0829D10





573.
H3037H02-3
1110018O12Rik
RIKEN cDNA
H3037H02





1110018O12





gene





574.
K0105H12-3
Cdk6
cyclin-
K0105H12





dependent





kinase 6





575.
C0105D10-3

C0105D10-3
C0105D10





NIA Mouse





E7.5





Extraembryonic





Portion cDNA





Library Mus






musculus






cDNA clone





C0105D10 3′,





MRNA





sequence





576.
L0229E05-3
Prkx
putative
L0229E05





serine/threonine





kinase





577.
L0931H07-3

ESTs
L0931H07





BQ557106





578.
K0138B11-3
Trim25
tripartite motif
K0138B11





protein 25





579.
H3019H03-3
Lass6
longevity
H3019H03





assurance





homolog 6 (S. cerevisiae)





580.
J0051F04-3
Ifi30
interferon
J0051F04





gamma





inducible





protein 30





581.
H3106G04-3
Cacna1d
calcium
H3106G04





channel,





voltage-





dependent, L





type, alpha 1D





subunit





582.
L0701D10-3
Arhgdib
Rho, GDP
L0701D10





dissociation





inhibitor (GDI)





beta





583.
H3137A02-3


Mus musculus

H3137A02





10 days neonate





cerebellum





cDNA, RIKEN





full-length





enriched





library,





clone: B930053





B19





product: unknown





EST, full





insert sequence.





584.
L0043D10-3
A530090O15Rik
RIKEN cDNA
L0043D10





A530090O15





gene





585.
H3087D06-3
Etf1
eukaryotic
H3087D06





translation





termination





factor 1





586.
C0827E01-3


Mus musculus

C0827E01





15 days embryo





head cDNA,





RIKEN full-





length enriched





library,





clone: D930031H08





product: unknown





EST, full





insert sequence.





587.
H3053E01-3
B130024B19Rik
RIKEN cDNA
H3053E01





B130024B19





gene





588.
K0117C08-3
BM222243
ESTs
K0117C08





BM222243





589.
H3056D11-3
Ptgfrn
prostaglandin
H3056D11





F2 receptor





negative





regulator





590.
C0228C02-3
2510004L01Rik
RIKEN cDNA
C0228C02





2510004L01





gene





591.
H3144F09-3
Rab7l1
RAB7, member
H3144F09





RAS oncogene





family-like 1





592.
H3052B06-3
Abcb1b
ATP-binding
H3052B06





cassette, sub-





family B





(MDR/TAP),





member 1B





593.
L0273B08-3
Tgif
TG interacting
L0273B08





factor





594.
K0406A08-3
Siat4c
sialyltransferase
K0406A08





4C (beta-





galactoside





alpha-2,3-





sialytransferase)





595.
AF075136.1
Sap30
sin3 associated
AF075136





polypeptide





596.
K0644H12-3
Prkch
protein kinase
K0644H12





C, eta





597.
H3108A04-3
Clu
clusterin
H3108A04





598.
H3020F06-3
Snx10
sorting nexin 10
H3020F06





599.
L0066C05-3
Uxs1
UDP-
L0066C05





glucuronate





decarboxylase 1





600.
L0025F08-3
Rgs19
regulator of G-
L0025F08





protein





signaling 19





601.
H3076F06-3
Siat4a
sialyltransferase
H3076F06





4A (beta-





galactoside





alpha-2,3-





sialytransferase)





602.
C0354G01-3


Mus musculus,

C0354G01





Similar to IQ





motif





containing





GTPase





activating





protein 2, clone





IMAGE: 3596508,





mRNA,





partial cds





603.
C0191H09-3
Atp6v1a1
ATPase, H+
C0191H09





transporting,





V1 subunit A,





isoform 1





604.
H3050G04-3
Dpp7
dipeptidylpeptidase 7
H3050G04





605.
L0219A09-3
Gatm
glycine
L0219A09





amidinotransferase





(L-





arginine:glycine





amidinotransferase)





606.
J0821E02-3
AU040950
expressed
J0821E02





sequence





AU040950





607.
H3080A02-3
Cbfb
core binding
H3080A02





factor beta





608.
C0276B08-3
Plscr1
phospholipid
C0276B08





scramblase 1





609.
C0279E04-3
Srd5a2l
steroid 5 alpha-
C0279E04





reductase 2-like





610.
K0434D04-3
Pgd
phosphogluconate
K0434D04





dehydrogenase





611.
C0174H01-3
Ddx21
DEAD (Asp-
C0174H01





Glu-Ala-Asp)





box polypeptide





21





612.
H3085A07-3
BG070224
ESTs
H3085A07





BG070224





613.
K0208E10-3
Mmab
methylmalonic
K0208E10





aciduria





(cobalamin





deficiency) type





B homolog





(human)





614.
H3006F10-3
Cops2
COP9
H3006F10





(constitutive





photomorphogenic)





homolog,





subunit 2





(Arabidopsis






thaliana)






615.
C0108A10-3
Nek6
NIMA (never in
C0108A10





mitosis gene a)-





related





expressed





kinase 6





616.
H3028H10-3
Ppic
peptidylprolyl
H3028H10





isomerase C





617.
H3121E08-3
Ralgds
ral guanine
H3121E08





nucleotide





dissociation





stimulator





618.
L0266H12-3
Opa1
optic atrophy 1
L0266H12





homolog





(human)





619.
K0635G02-3
2310046K10Rik
RIKEN cDNA
K0635G02





2310046K10





gene





620.
L0704C05-3
2610318G18Rik
RIKEN cDNA
L0704C05





2610318G18





gene





621.
C0303D10-3

UNKNOWN
C0303D10





C0303D10





622.
K0605C04-3
BM240648
ESTs
K0605C04





BM240648





623.
H3071G06-3
BG069012
ESTs
H3071G06





BG069012





624.
C0600A01-3
Coro2a
coronin, actin
C0600A01





binding protein





2A





625.
NM_007679.1
Cebpd
CCAAT/enhancer
NM_007679





binding





protein





(C/EBP), delta





626.
H3048A01-3
Kras2
Kirsten rat
H3048A01





sarcoma





oncogene 2,





expressed





627.
C0267D12-3
Tpp2
tripeptidyl
C0267D12





peptidase II





628.
J1012C06-3
AU041997
ESTs
J1012C06





AU041997





629.
L0072F04-3
Vav2
Vav2 oncogene
L0072F04





630.
L0836H04-3
C030038J10Rik
RIKEN cDNA
L0836H04





C030038J10





gene





631.
K0614A10-3
Sh3kbp1
SH3-domain
K0614A10





kinase binding





protein 1





632.
H3156B08-3
6620401D04Rik
RIKEN cDNA
H3156B08





6620401D04





gene





633.
C0334C11-3
B230339H12Rik
RIKEN cDNA
C0334C11





B230339H12





gene





634.
H3103G05-3
BG071839
ESTs
H3103G05





BG071839





635.
C0205H05-3
1600010D10Rik
RIKEN cDNA
C0205H05





1600010D10





gene





636.
L0513G12-3
Qk
quaking
L0513G12





637.
C0100E08-3
Pdap1
PDGFA
C0100E08





associated





protein 1





638.
J0055B04-3

transcribed
J0055B04





sequence with





strong





similarity to





protein





pir: S12207





(M. musculus)





S12207





hypothetical





protein (B2





element) -





mouse





639.
J0008D10-3
Mbp
myelin basic
J0008D10





protein





640.
K0319D09-3
Mtml
X-linked
K0319D09





myotubular





myopathy gene 1





641.
C0243H05-3
Galnt7
UDP-N-acetyl-
C0243H05





alpha-D-





galactosamine:





polypeptide N-





acetylgalactosaminyltransferase 7





642.
L0841H10-3
BM116846
ESTs
L0841H10





BM116846





643.
K0334D05-3
Ccnd1
cyclin D1
K0334D05





644.
L0209B01-3

L0209B01-3
L0209B01





NIA Mouse





Newborn Ovary





cDNA Library






Mus musculus






cDNA clone





L0209B01 3′,





MRNA





sequence





645.
K0151H10-3
BB129550
EST BB129550
K0151H10





646.
L0505B11-3
Ammecr1
Alport
L0505B11





syndrome,





mental





retardation,





midface





hypoplasia and





elliptocytosis





chromosomal





region gene 1





homolog





(human)





647.
L0944C06-3
BM120800
ESTs
L0944C06





BM120800





648.
J0027C07-3
Mrps25
mitochondrial
J0027C07





ribosomal





protein S25





649.
L0855B04-3
Wdr26
WD repeat
L0855B04





domain 26





650.
H3060H05-3


Mus musculus

H3060H05





cDNA clone





MGC: 28609





IMAGE: 42185





51, complete





cds





651.
K0330G09-3
5830461H18Rik
RIKEN cDNA
K0330G09





5830461H18





gene





652.
L0803E07-3
Dpysl4
dihydropyrimidinase-
L0803E07





like 4





653.
L0283B01-3
Ivnslabp
influenza virus
L0283B01





NS1A binding





protein





654.
L0065G02-3
6530401D17Rik
RIKEN cDNA
L0065G02





6530401D17





gene





655.
C0949A06-3


Mus musculus

C0949A06





0 day neonate





skin cDNA,





RIKEN full-





length enriched





library,





clone: 4632424N07





product: unknown





EST, full





insert sequence.





656.
H3100C11-3
BG071548
ESTs
H3100C11





BG071548





657.
C0142H08-3
3110020O18Rik
RIKEN cDNA
C0142H08





3110020O18





gene





658.
L0945G09-3
Bcl2l11
BCL2-like 11
L0945G09





(apoptosis





facilitator)





659.
L0848H06-3
E130318E12Rik
RIKEN cDNA
L0848H06





E130318E12





gene





660.
K0617B02-3
Bmp2k
BMP2
K0617B02





inducible





kinase





661.
C0203D07-3
Pftk1
PFTAIRE
C0203D07





protein kinase 1





662.
L0267A02-3
2210409B22Rik
RIKEN cDNA
L0267A02





2210409B22





gene





663.
J0086F05-3

transcribed
J0086F05





sequence with





moderate





similarity to





protein





sp: P00722 (E. coli)





BGAL_ECOLI





Beta-





galactosidase





(Lactase)





664.
C0606A03-3
Rps23
ribosomal
C0606A03





protein S23





665.
L0902D02-3
Ncoa6ip
nuclear receptor
L0902D02





coactivator 6





interacting





protein





666.
H3060C12-3
BG067974
ESTs
H3060C12





BG067974





667.
C0611E01-3
Tor3a
torsin family 3,
C0611E01





member A





668.
U54984.1
Mmp14
matrix
U54984





metalloproteinase





14





(membrane-





inserted)





669.
H3089F08-3
0610013E23Rik
RIKEN cDNA
H3089F08





0610013E23





gene





670.
K0633C04-3
Ebi2
Epstein-Barr
K0633C04





virus induced





gene 2





671.
J0943E09-3
Nup62
nucleoporin 62
J0943E09





672.
L0267D03-3
Dcn
decorin
L0267D03





673.
L0250B09-3
1110031E24Rik
RIKEN cDNA
L0250B09





1110031E24





gene





674.
L0915B12-3
Etv3
ets variant gene 3
L0915B12





675.
NM_009403.1
Tnfsf8
tumor necrosis
NM_009403





factor (ligand)





superfamily,





member 8





676.
C0308F04-3
2700064H14Rik
RIKEN cDNA
C0308F04





2700064H14





gene





677.
C0288G12-3
6030400A10Rik
RIKEN cDNA
C0288G12





6030400A10





gene





678.
H3005A11-3
Fancd2
Fanconi
H3005A11





anemia,





complementation





group D2





679.
H3121H07-3
2810405I11Rik
RIKEN cDNA
H3121H07





2810405I11





gene





680.
K0124A06-3
BM222608
ESTs
K0124A06





BM222608





681.
NM_010835.1
Msx1
homeo box,
NM_010835





msh-like 1





682.
K0134C07-3
Falz
fetal Alzheimer
K0134C07





antigen





683.
K0424H02-3
Pfkp
phosphofructokinase,
K0424H02





platelet





684.
H3153G06-3
8030446C20Rik
RIKEN cDNA
H3153G06





8030446C20





gene





685.
H3071C09-3
BG068971
ESTs
H3071C09





BG068971





686.
L0243B07-3

Possibly
L0243B07





intronic in





U008124-





L0243B07





687.
C0143D11-3
Ii
Ia-associated
C0143D11





invariant chain





688.
L0512A02-3
Snx5
sorting nexin 5
L0512A02





689.
K0112C06-3
Atp8a1
ATPase,
K0112C06





aminophospholipid





transporter





(APLT), class I,





type 8A,





member 1





690.
H3053A01-3
Tnfsf13b
tumor necrosis
H3053A01





factor (ligand)





superfamily,





member 13b





691.
C0668F08-3
Atp6ap2
ATPase, H+
C0668F08





transporting,





lysosomal





accessory





protein 2





692.
K0417E05-3
Osmr
oncostatin M
K0417E05





receptor





693.
NM_010872.1
Birc1b
baculoviral IAP
NM_010872





repeat-





containing 1b





694.
L0262G06-3
Cfh
complement
L0262G06





component





factor h





695.
J0249F06-3
2210023K21Rik
RIKEN cDNA
J0249F06





2210023K21





gene





696.
C0170A02-3
Serpinb9
serine (or
C0170A02





cysteine)





proteinase





inhibitor, clade





B, member 9





697.
H3076C12-3
Facl4
fatty acid-
H3076C12





Coenzyme A





ligase, long





chain 4





698.
H3155C07-3
1810036L03Rik
RIKEN cDNA
H3155C07





1810036L03





gene





699.
K0331C04-3
Sdccag8
serologically
K0331C04





defined colon





cancer antigen 8





700.
J0538B04-3
Laptm5
lysosomal-
J0538B04





associated





protein





transmembrane 5





701.
H3014E07-3
1810029G24Rik
RIKEN cDNA
H3014E07





1810029G24





gene





702.
K0515H12-3
2900064A13Rik
RIKEN cDNA
K0515H12





2900064A13





gene





703.
H3159D10-3
BG076403
ESTs
H3159D10





BG076403





704.
K0127F02-3
Prg
proteoglycan,
K0127F02





secretory





granule





705.
L0919B08-3
Bnip3l
BCL2/adenovirus
L0919B08





E1B 19 kDa-





interacting





protein 3-like





706.
J0904A09-3
1110060F11Rik
RIKEN cDNA
J0904A09





1110060F11





gene





707.
L0270B06-3
D11Ertd759e
DNA segment,
L0270B06





Chr 11,





ERATO Doi





759, expressed





708.
K0230D06-3
Eaf1
ELL associated
K0230D06





factor 1





709.
K0611A03-3
AI447904
expressed
K0611A03





sequence





AI447904





710.
H3155A07-3
BG076050
ESTs
H3155A07





BG076050





711.
H3028H11-3
Ctsh
cathepsin H
H3028H11





712.
L0001D12-3
4833422F06Rik
RIKEN cDNA
L0001D12





4833422F06





gene





713.
L0951G01-3
BG061831
ESTs
L0951G01





BG061831





714.
H3035G02-3
AI314180
expressed
H3035G02





sequence





AI314180





715.
C0925G02-3
Fer1l3
fer-1-like 3,
C0925G02





myoferlin (C. elegans)





716.
C0103H10-3
Il17r
interleukin 17
C0103H10





receptor





717.
H3129F05-3
Mrpl16
mitochondrial
H3129F05





ribosomal





protein L16





718.
L0942B12-3


Mus musculus

L0942B12





12 days embryo





spinal ganglion





cDNA, RIKEN





full-length





enriched





library,





clone: D130046C24





product: unknown





EST, full





insert sequence.





719.
L0009B09-3
Plcg2
phospholipase
L0009B09





C, gamma 2





720.
C0665B08-3
Sh3bp1
SH3-domain
C0665B08





binding protein 1





721.
H3102F04-3
Rgs10
regulator of G-
H3102F04





protein





signalling 10





722.
K0547F06-3

transcribed
K0547F06





sequence with





moderate





similarity to





protein





sp: P00722 (E. coli)





BGAL_ECOLI





Beta-





galactosidase





(Lactase)





723.
H3087C07-3
Glb1
galactosidase,
H3087C07





beta 1





724.
J0437D05-3
AU023716
ESTs
J0437D05





AU023716





725.
H3156A09-3
Pex12
peroxisomal
H3156A09





biogenesis





factor 12





726.
G0108H12-3
Ly6e
lymphocyte
G0108H12





antigen 6





complex, locus E





727.
H3098D12-5
Map2k1
mitogen
H3098D12





activated





protein kinase





kinase 1





728.
C0637C02-3
Zmpste24
zinc
C0637C02





metalloproteinase,





STE24





homolog (S. cerevisiae)





729.
H3119B06-3
Atp1b3
ATPase,
H3119B06





Na+/K+





transporting,





beta 3





polypeptide





730.
C0176B06-3
Ubl1
ubiquitin-like 1
C0176B06





731.
C0626D04-3
9130404D14Rik
RIKEN cDNA
C0626D04





9130404D14





gene





732.
H3155E07-3
Dock4
dedicator of
H3155E07





cytokinesis 4





733.
C0106A05-3
H2-Eb1
histocompatibility
C0106A05





2, class II





antigen E beta





734.
H3037B09-3


Mus musculus

H3037B09





12 days embryo





spinal cord





cDNA, RIKEN





full-length





enriched





library,





clone: C530028D16





product: 231000





8H09RIK





PROTEIN





homolog [Mus






musculus], full






insert sequence.





735.
H3003B09-3
F730017H24Rik
RIKEN cDNA
H3003B09





F730017H24





gene





736.
C0909E10-3
Pign
phosphatidylinositol
C0909E10





glycan,





class N





737.
H3045G01-3
BG066588
ESTs
H3045G01





BG066588





738.
H3006E10-3

transcribed
H3006E10





sequence with





weak similarity





to protein





sp: Q9H321





(H. sapiens)





VCXC_HUMAN





VCX-C





protein





(Variably





charged protein





X-C)





739.
H3098H09-3
2310016E02Rik
RIKEN cDNA
H3098H09





2310016E02





gene





740.
J0540D09-3
Adam9
a disintegrin
J0540D09





and





metalloproteinase





domain 9





(meltrin





gamma)





741.
L0208C06-3
Pknox1
Pbx/knotted 1
L0208C06





homeobox





742.
H3154G05-3
Napg
N-
H3154G05





ethylmaleimide





sensitive fusion





protein





attachment





protein gamma





743.
L0854E11-3
1500032M01Rik
RIKEN cDNA
L0854E11





1500032M01





gene





744.
H3014C06-3
B2m
beta-2
H3014C06





microglobulin





745.
K0538G12-3
Ccr2
chemokine (C-
K0538G12





C) receptor 2





746.
J0819C09-3
C030002B11Rik
RIKEN cDNA
J0819C09





C030002B11





gene





747.
C0175B11-3
Hist1h2bc
histone 1, H2bc
C0175B11





748.
H3009B11-3
Nufip1
nuclear fragile
H3009B11





X mental





retardation





protein





interacting





protein





749.
H3135D02-3
Lamp2
lysosomal
H3135D02





membrane





glycoprotein 2





750.
K0540G08-3
1200013B08Rik
RIKEN cDNA
K0540G08





1200013B08





gene





751.
H3089H05-3
Lnx2
ligand of numb-
H3089H05





protein X 2





752.
J0203A08-3
C85149
ESTs C85149
J0203A08





753.
H3119F01-3
Mcfd2
multiple
H3119F01





coagulation





factor





deficiency 2





754.
H3134C05-3
Mglap
matrix gamma-
H3134C05





carboxyglutamate





(gla) protein





755.
C0147D11-3
B230215M10Rik
RIKEN cDNA
C0147D11





B230215M10





gene





756.
C0949H10-3
Sulf1
sulfatase 1
C0949H10





757.
K0114E04-3
BM222075
ESTs
K0114E04





BM222075





758.
H3012C03-3
Cappa1
capping protein
H3012C03





alpha 1





759.
C0507E11-3
BE824970
ESTs
C0507E11





BE824970





760.
H3158D06-3
Lnk
linker of T-cell
H3158D06





receptor





pathways





761.
C0174C02-3
Pold3
polymerase
C0174C02





(DNA-





directed), delta





3, accessory





subunit





762.
C0130G10-3
Cklfsf7
chemokine-like
C0130G10





factor super





family 7





763.
C0137F07-3
Pik3cb
phosphatidylinositol
C0137F07





3-kinase,





catalytic, beta





polypeptide





764.
H3115F01-3
2610027O18Rik
RIKEN cDNA
H3115F01





2610027O18





gene





765.
H3097F03-3


Mus musculus,

H3097F03





clone





IMAGE: 53723





38, mRNA





766.
H3059A05-3
Mad2l1
MAD2 (mitotic
H3059A05





arrest deficient,





homolog)-like 1





(yeast)





767.
L0935E02-3
Syk
spleen tyrosine
L0935E02





kinase





768.
C0946F08-3
1110014L17Rik
RIKEN cDNA
C0946F08





1110014L17





gene





769.
H3079F02-5

Possibly
H3079F02





intronic in





U011488-





H3079F02





770.
H3137E07-3
Il10ra
interleukin 10
H3137E07





receptor, alpha





771.
C0143H12-3
Galns
galactosamine
C0143H12





(N-acetyl)-6-





sulfate sulfatase





772.
H3114D03-3
Man2a1
mannosidase 2,
H3114D03





alpha 1





773.
H3041H09-3
BG066348
ESTs
H3041H09





BG066348





774.
C0628H04-3
Slc2a12
solute carrier
C0628H04





family 2,





member 12





775.
K0125E07-3
Ifngr
interferon
K0125E07





gamma receptor





776.
G0115E02-3
Sdcbp
syndecan
G0115E02





binding protein





777.
C0032B05-3
Rap2b
RAP2B,
C0032B05





member of





RAS oncogene





family





778.
H3141C08-3
Ofd1
oral-facial-
H3141C08





digital





syndrome 1





gene homolog





(human)





779.
H3157C05-3
BG076236
ESTs
H3157C05





BG076236





780.
H3076A01-3
5031439G07Rik
RIKEN cDNA
H3076A01





5031439G07





gene





781.
H3080D06-3
BC018507
cDNA sequence
H3080D06





BC018507





782.
L0518D04-3
Uap1
UDP-N-
L0518D04





acetylglucosamine





pyrophosphorylase 1





783.
K0542B11-3
BM239901
ESTs
K0542B11





BM239901





784.
L0959D03-3
Tnfrsf1a
tumor necrosis
L0959D03





factor receptor





superfamily,





member 1a





785.
H3035C07-3
BG065787
ESTs
H3035C07





BG065787





786.
M29855.1
Csf2rb2
colony
M29855





stimulating





factor 2





receptor, beta 2,





low-affinity





(granulocyte-





macrophage)





787.
C0352C11-3
BM197981
ESTs
C0352C11





BM197981





788.
L0846B10-3
BM117093
ESTs
L0846B10





BM117093





789.
L0227C06-3
Serpinb6a
serine (or
L0227C06





cysteine)





proteinase





inhibitor, clade





B, member 6a





790.
J0214H09-3
Serpina3g
serine (or
J0214H09





cysteine)





proteinase





inhibitor, clade





A, member 3G





791.
H3077F12-3
Arhh
ras homolog
H3077F12





gene family,





member H





792.
C0341D05-3
BM196992
ESTs
C0341D05





BM196992





793.
H3043H11-3
BG066522
ESTs
H3043H11





BG066522





794.
K0507D06-3


Mus musculus,

K0507D06





clone





IMAGE: 1263252,





mRNA





795.
J0535D11-3
AU020606
ESTs
J0535D11





AU020606





796.
H3152F04-3
Sepp1
selenoprotein P,
H3152F04





plasma, 1





797.
L0701F07-3
H2-Ab1
histocompatibility
L0701F07





2, class II





antigen A, beta 1





798.
L0227H07-3
Clca1
chloride
L0227H07





channel calcium





activated 1





799.
J1014C11-3
2900036G02Rik
RIKEN cDNA
J1014C11





2900036G02





gene





800.
H3134H09-3
BG074421
ESTs
H3134H09





BG074421





801.
G0116A07-3
Atp6v1c1
ATPase, H+
G0116A07





transporting,





V1 subunit C,





isoform 1





802.
L0942F05-3
Ostm1
osteopetrosis
L0942F05





associated





transmembrane





protein 1





803.
C0912H10-3
0610041E09Rik
RIKEN cDNA
C0912H10





0610041E09





gene





804.
C0304E12-3
Pde1b
phosphodiesterase
C0304E12





1B, Ca2+-





calmodulin





dependent





805.
L0605C12-3
4930579K19Rik
RIKEN cDNA
L0605C12





4930579K19





gene





806.
K0539A07-3
Cd53
CD53 antigen
K0539A07





807.
L0228H12-3
6430628I05Rik
RIKEN cDNA
L0228H12





6430628I05





gene





808.
L0855B10-3
BM117713
ESTs
L0855B10





BM117713





809.
H3075B10-3
2810404F18Rik
RIKEN cDNA
H3075B10





2810404F18





gene





810.
L0022G07-3

L0022G07-3
L0022G07





NIA Mouse





E12.5 Female





Mesonephros





and Gonads





cDNA Library






Mus musculus






cDNA clone





L0022G07 3′,





MRNA





sequence





811.
H3107C11-3
Efemp2
epidermal
H3107C11





growth factor-





containing





fibulin-like





extracellular





matrix protein 2





812.
H3025H12-3
1200003O06Rik
RIKEN cDNA
H3025H12





1200003O06





gene





813.
J0040E05-3
Stx3
syntaxin 3
J0040E05





814.
H3075F03-3
C1s
complement
H3075F03





component 1, s





subcomponent





815.
L0600G09-3
BM125147
ESTs
L0600G09





BM125147





816.
K0115H01-3
KLHL6
kelch-like 6
K0115H01





817.
H3015B10-3
Gus
beta-
H3015B10





glucuronidase





818.
H3108A12-3
0910001A06Rik
RIKEN cDNA
H3108A12





0910001A06





gene





819.
H3108H09-5

UNKNOWN:
H3108H09





Similar to






Homo sapiens






KIAA1577





protein





(KIAA1577),





mRNA





820.
K0645H01-3
Fyb
FYN binding
K0645H01





protein





821.
H3029A02-3
Shyc
selective
H3029A02





hybridizing





clone





822.
K0410D10-3
Cxcl12
chemokine
K0410D10





(C—X—C motif)





ligand 12





823.
H3118H11-3
Snrpg
small nuclear
H3118H11





ribonucleoprote





in polypeptide G





824.
K0517D08-3
BM238427
ESTs
K0517D08





BM238427





825.
L0227G11-3
Sh3d1B
SH3 domain
L0227G11





protein 1B





826.
H3134B10-3
6530409L22Rik
RIKEN cDNA
H3134B10





6530409L22





gene





827.
H3115A08-3
Ly6a
lymphocyte
H3115A08





antigen 6





complex, locus A





828.
C0120G03-3
Csk
c-src tyrosine
C0120G03





kinase





829.
H3094G08-3
Tigd2
tigger
H3094G08





transposable





element derived 2





830.
NM_008362.1
Il1r1
interleukin 1
NM_008362





receptor, type I





831.
C0300E10-3
Trps1
trichorhinophal
C0300E10





angeal





syndrome I





(human)





832.
L0274A03-3
Ptpn2
protein tyrosine
L0274A03





phosphatase,





non-receptor





type 2





833.
H3005H07-3
1810031K02Rik
RIKEN cDNA
H3005H07





1810031K02





gene





834.
H3109H12-3
1810009M01Rik
RIKEN cDNA
H3109H12





1810009M01





gene





835.
J0008D01-3
Enpp1
ectonucleotide
J0008D01





pyrophosphatase/





phosphodiesterase 1





836.
H3119H05-3
Mafb
v-maf
H3119H05





musculoaponeurotic





fibrosarcoma





oncogene





family, protein





B (avian)





837.
H3048G11-3
Blvrb
biliverdin
H3048G11





reductase B





(flavin





reductase





(NADPH))





838.
H3107D05-3
1110004C05Rik
RIKEN cDNA
H3107D05





1110004C05





gene





839.
H3006B01-3
Cklfsf3
chemokine-like
H3006B01





factor super





family 3





840.
L0853H04-3

transcribed
L0853H04





sequence with





weak similarity





to protein





pir: A43932





(H. sapiens)





A43932 mucin





2 precursor,





intestinal -





human





(fragments)





841.
C0949G05-3
BM221093
ESTs
C0949G05





BM221093





842.
K0648D10-3
Tlr1
toll-like
K0648D10





receptor 1





843.
H3014E09-3
BC017643
cDNA sequence
H3014E09





BC017643





844.
H3022D06-3
Il2rg
interleukin 2
H3022D06





receptor,





gamma chain





845.
L0201A03-3
2410004H05Rik
RIKEN cDNA
L0201A03





2410004H05





gene





846.
H3026E03-5


Mus musculus

H3026E03





2 days neonate





thymus thymic





cells cDNA,





RIKEN full-





length enriched





library,





clone: E430039





C10





product: unknown





EST, full





insert sequence





847.
H3091E12-3
Abhd2
abhydrolase
H3091E12





domain





containing 2





848.
H3003E01-3
Cutl1
cut-like 1
H3003E01





(Drosophila)





849.
H3016H08-5
Crsp9
cofactor
H3016H08





required for





Sp1





transcriptional





activation,





subunit 9,





33 kDa





850.
C0118E09-3
Oas1a
2′-5′
C0118E09





oligoadenylate





synthetase 1A





851.
L0535B02-3
Col15a1
procollagen,
L0535B02





type XV





852.
L0500E02-3
Sgcg
sarcoglycan,
L0500E02





gamma





(dystrophin-





associated





glycoprotein)





853.
H3077B08-3
5330431K02Rik
RIKEN cDNA
H3077B08





5330431K02





gene





854.
J0209G02-3
Gnb4
guanine
J0209G02





nucleotide





binding protein,





beta 4





855.
C0661E01-3
Lcn7
lipocalin 7
C0661E01





856.
K0221E09-3
Scml2
sex comb on
K0221E09





midleg-like 2





(Drosophila)





857.
C0184F12-3
D8Ertd594e
DNA segment,
C0184F12





Chr 8, ERATO





Doi 594,





expressed





858.
L0602B03-3
Myoz2
myozenin 2
L0602B03





859.
C0944F04-3
1110055E19Rik
RIKEN cDNA
C0944F04





1110055E19





gene





860.
L0004A03-3
Gli2
GLI-Kruppel
L0004A03





family member





GLI2





861.
L0860B03-3

ESTs
L0860B03





AV321020





862.
L0841F10-3
2310045A20Rik
RIKEN cDNA
L0841F10





2310045A20





gene





863.
L0008H10-3
Agrn
agrin
L0008H10





864.
C0128B02-3
Casq1
calsequestrin 1
C0128B02





865.
C0645C09-3
BM209340
ESTs
C0645C09





BM209340





866.
H3082B03-3
Mylk
myosin, light
H3082B03





polypeptide





kinase





867.
C0309D09-3

transcribed
C0309D09





sequence with





moderate





similarity to





protein





sp: P00722 (E. coli)





BGAL_ECOLI





Beta-





galactosidase





(Lactase)





868.
H3157H09-3
BG076287
ESTs
H3157H09





BG076287





869.
H3061D03-3
Pcsk5
proprotein
H3061D03





convertase





subtilisin/kexin





type 5





870.
L0843D01-3
3732412D22Rik
RIKEN cDNA
L0843D01





3732412D22





gene





871.
L0702H07-3
5830415L20Rik
RIKEN cDNA
L0702H07





5830415L20





gene





872.
L0548G08-3
Xin
cardiac
L0548G08





morphogenesis





873.
L0803E02-3
Nkd1
naked cuticle 1
L0803E02





homolog





(Drosophila)





874.
C0925G12-3
Fbxo30
F-box protein
C0925G12





30





875.
L0911A11-3
2010313D22Rik
RIKEN cDNA
L0911A11





2010313D22





gene





876.
AF084466.1
Rrad
Ras-related
AF084466





associated with





diabetes





877.
H3073G09-3
1600029N02Rik
RIKEN cDNA
H3073G09





1600029N02





gene





878.
L0815B08-3
1100001D19Rik
RIKEN cDNA
L0815B08





1100001D19





gene





879.
J1037H05-3
D230016N13Rik
RIKEN cDNA
J1037H05





D230016N13





gene





880.
K0421F09-3

transcribed
K0421F09





sequence with





weak similarity





to protein





ref: NP_081764.1





(M. musculus)





RIKEN cDNA





5730493B19





[Mus musculus]





881.
H3082E06-3
1110003B01Rik
RIKEN cDNA
H3082E06





1110003B01





gene





882.
C0935B04-3
Hhip
Hedgehog-
C0935B04





interacting





protein





883.
H3116B02-3
1110007C05Rik
RIKEN cDNA
H3116B02





1110007C05





gene





884.
C0945G10-3
Tp53i11
tumor protein
C0945G10





p53 inducible





protein 11





885.
K0440G09-3
Tgfb3
transforming
K0440G09





growth factor,





beta 3





886.
L0916G12-3
BM118833
ESTs
L0916G12





BM118833





887.
L0505A04-3
Dnajb5
DnaJ (Hsp40)
L0505A04





homolog,





subfamily B,





member 5





888.
L0542E08-3
Usmg4
upregulated
L0542E08





during skeletal





muscle growth 4





889.
L0223E12-3
Sparcl1
SPARC-like 1
L0223E12





(mast9, hevin)





890.
K0349C07-3
4631423F02Rik
RIKEN cDNA
K0349C07





4631423F02





gene





891.
C0302A11-3

EST BI988881
C0302A11





892.
C0930C11-3
Fgf13
fibroblast
C0930C11





growth factor





13





893.
H3022A11-3
Cald1
caldesmon 1
H3022A11





894.
C0660B06-3
Csrp1
cysteine and
C0660B06





glycine-rich





protein 1





895.
L0949F12-3
Heyl
hairy/enhancer-
L0949F12





of-split related





with YRPW





motif-like





896.
K0225B06-3
Unc5c
unc-5 homolog
K0225B06





C (C. elegans)





897.
K0541E04-3
Herc3
hect domain
K0541E04





and RLD 3





898.
C0151A03-3
BC026744
cDNA sequence
C0151A03





BC026744





899.
L0045C07-3
6-Sep
septin 6
L0045C07





900.
L0509E03-3
Ryr2
ryanodine
L0509E03





receptor 2,





cardiac





901.
H3049B08-3
Tes
testis derived
H3049B08





transcript





902.
L0533C09-3
BM123974
ESTs
L0533C09





BM123974





903.
H3108C01-3
4930444A02Rik
RIKEN cDNA
H3108C01





4930444A02





gene





904.
C0110C06-3
Epb4.1l1
erythrocyte
C0110C06





protein band





4.1-like 1





905.
C0324H08-3
Enah
enabled
C0324H08





homolog





(Drosophila)





906.
C0917A09-3

ESTs
C0917A09





BB231855





907.
L0854B10-3
Anks1
ankyrin repeat
L0854B10





and SAM





domain





containing 1





908.
K0326D08-3
Ly75
lymphocyte
K0326D08





antigen 75





909.
H3074H01-3
C430017H16
hypothetical
H3074H01





protein





C430017H16





910.
H3131D02-3
Tnk2
tyrosine kinase,
H3131D02





non-receptor, 2





911.
C0112B03-3
Heyl
hairy/enhancer-
C0112B03





of-split related





with YRPW





motif-like





912.
L0514A09-3
6430511F03
hypothetical
L0514A09





protein





6430511F03





913.
C0234D07-3
Fbxo30
F-box protein
C0234D07





30





914.
H3152A02-3
St6gal1
beta galactoside
H3152A02





alpha 2,6





sialyltransferase 1





915.
H3075C04-3
Ches1
checkpoint
H3075C04





suppressor 1





916.
L0600E02-3
BM125123
ESTs
L0600E02





BM125123





917.
K0501F10-3
BM237456
ESTs
K0501F10





BM237456





918.
K0301H08-3
Oxct
3-oxoacid CoA
K0301H08





transferase





919.
L0229E07-3
Lu
Lutheran blood
L0229E07





group





(Auberger b





antigen





included)





920.
H3077C06-3
4931430I01Rik
RIKEN cDNA
H3077C06





4931430I01





gene





921.
J0807D02-3


Mus musculus

J0807D02





10 days neonate





cerebellum





cDNA, RIKEN





full-length





enriched





library,





clone: B930022I23





product: unclassifiable,





full





insert sequence.





922.
H3118G11-3
C130068N17
hypothetical
H3118G11





protein





C130068N17





923.
L0818F01-3
Smarcd3
SWI/SNF
L0818F01





related, matrix





associated,





actin dependent





regulator of





chromatin,





subfamily d,





member 3





924.
C0359A10-3
BM198389
ESTs
C0359A10





BM198389





925.
G0108E12-3
1190009E20Rik
RIKEN cDNA
G0108E12





1190009E20





gene





926.
C0941C09-3
Gja7
gap junction
C0941C09





membrane





channel protein





alpha 7





927.
H3111B03-5

UNKNOWN
H3111B03





H3111B03
















SEQ







ID
UG
CHR_LOCATION
60mer



NO:
CLUSTER
PENG [A]
SEQUENCE







  1.

No Chromosome location
ATGAGCCTAGA





info available
ACTCACATGCA






TTTTCCTGACT






TCTATCATTAG






AATAAGTTCAT






CAAGA







  2.
Mm.389
Chromosome 15
CCTATTGTTGA






GTGTCAAACAT






CACCACTAAGT






GGATGGTTATG






TAGTCCATTAT






CCAAA







  3.
Mm.103301
Chromosome 4
TACCTGAACCA






CTCTCTACTGT






TGTTGTCACAA






GGCAAAAGTG






GCATTCCTTCC






TCCAAG







  4.
Mm.231395
Chromosome 7
CCCTTTGCTGT






GTGGGCAGTAC






TCTGAAGCAGG






CAAATGGGTCT






TAGGATCCCTC






CCAGA







  5.
Mm.222000
Chromosome 6
TCCAAAGATAA






AATGAGCAAC






CGCACTGGCTT






AGCCATAGATG






ACTGACAGTGA






TTGGAA







  6.
Mm.10756
Chromosome 1
TGCCTTGGAGG






GCAACAAGGA






GCAGATACAG






AAGATCATTGA






GACACTGTTCA






CAGCAGC







  7.
Mm.268474
Chromosome 10
CATGAATTCCA






AACCAGTTATT






ATTAACATGAA






CCTGAACCTGA






ACAATTATGAC






TGTGC







  8.
Mm.45436
Chromosome 10
TTTCTGTCACT






GCTCAGGCCAA






GGTCTATGAAC






GTTGTGAGTTT






GCCAGAACTCT






GAAAA







  9.
Mm.39102
Chromosome 4
TTCATACCAAG






GAACCTGACCT






CTCTGACAATT






GCATTTTGAAC






ATTGTTGTCCC






CAAAG







 10.
Mm.247272
Chromosome 16
CATTGGAAACA






GACACGTTTGT






AGGCATTTGCG






TATTCTTGAAG






AGACTGTTTTA






TGAAT







 11.
Mm.200506
Chromosome 12
GTAATGGAGA






ATGTATCTGAA






CCCATATCAAG






CCATCTCTCTT






CCTTAACATGT






TAAGCA







 12.
Mm.7044
Chromosome 2
ACACCTCTAAC






TCCCAAGAAG






ACGGAGTGAA






TGTCCTCTCCT






TTACTTGTGAA






ATCATTT







 13.
Mm.6793
Chromosome 7
GTGAGATTCGG






CAGCATAAATT






GCGGAAACTG






AACCCACCCGA






TGAGAGTGGTC






CTGGCT







 14.
Mm.8245
Chromosome X
TCATAAGGGCT






AAATTCATGGG






TTCCCCAGAAA






TCAACGAGACC






ACCTTATACCA






GCGTT







 15.
Mm.217235
Chromosome 5
AAAGACTGAG






AGGAGTCATG






AACCAGGGTA






AAACTTATTGG






TGCTTTGAGAC






TTCCAGCA







 16.
Mm.230301
Chromosome 15
GCAGCATCGCT






TCCTTGGTTTA






TTCTTTGTGTTT






GTTCCTTCAGT






AAACATTTATT






GAGC







 17.

Chromosome 2
TTTTAACGGAG






CCTGAATATAG






CAGGTTTAAAA






TTTAAACAGGT






ATAAAATGAA






AAATAA







 18.
Mm.36571
Chromosome 4
TAGCATGAACC






ACCATGTTTGG






CAATACTGTAT






TTTAGAAAGAA






TTAATGGACTG






GAGAG







 19.
Mm.46424
Chromosome 11
CCTGAGCTCAC






TGTTTCTCATG






CTGTCTTGAGA






CAAAGTATCCA






TATGGAACCTA






GGTTA







 20.
Mm.44508
Chromosome 1
GCTGGTGTTTG






TGTCAAGAAA






ATGGCTGAAGC






TTGTTTCCAGG






CTGTAGGAATG






TTGAAC







 21.
Mm.4909
Chromosome 4
ACTTAAGTTAT






CTGCATAGAGG






CAATCCTCCTG






GGTTTGCTTTA






TGTCTCGAAAA






TCTAA







 22.
Mm.26437
Chromosome 12
GGGCAAAGGT






ACTTTCTGACA






AACTGAGTACC






TGAGATCAACC






CCCAAGAAGG






GAAAAAA







 23.
Mm.295683
Chromosome 5
ACTATGCAATT






GGACAGATGG






ATTACCAAGGA






GACTAAAAAT






ATATTCTTTGA






CTTTGGG







 24.
Mm.195099
Chromosome 5
TCACTGACCTC






AACCCCTCCTG






CAGAGAAGCC






TGAAGACCCCA






AAAGCTGCCA






GTCCAAA







 25.
Mm.196617
Chromosome 10
GATATAATGTG






ATAAAGTTCCA






AAAGGATCTCT






CTGGCTGAAGG






AGATACTGGAT






GGAAC







 26.
Mm.260421
No Chromosome location
CTGAACCCCAA





info available
TTAATAGCAAA






GGATATATCTC






TCTTCAAAAAC






GGATAGATTTC






TGAAG







 27.
Mm.3333
Chromosome 6
TTTTGTTCTCTC






CATCTGTTAGC






CGTTCTGAGGA






CTGAATGCAGA






TTGTCAGCTCA






AAAA







 28.
Mm.37657
Chromosome 16
GCCAATCTCAG






AACCCACATAG






AAGGGTCTGCA






GTATTATTCCT






GTTTCATGTGT






GCACA







 29.
Mm.156914
Chromosome 11
AGTGCAAAATT






TGGTTTGTTGG






TGTGCTTTTCT






GGTTTAGGAGC






CTGAAACAAG






CACACT







 30.
Mm.30074
Chromosome 11
CATGAGTAAGT






TGTGAAGGCTG






GACCCACATCT






TGATACTTGTT






TTCTGCATCTT






GGGCA







 31.
Mm.217705
Chromosome 12
TAGACGTTGTA






AAAAGGAGCC






AAGTTTATCAT






TTTGTTCCTTA






AATCCGTCATA






TGTGGG







 32.
Mm.1650
Chromosome 11
ACTGTGGTGAC






AGCTTCCTAAC






GTGTTTGTGTC






TAAAATAAACT






ATCCTTAGCAT






CCTTC







 33.
Mm.103987
Chromosome 15
TATAAATAGAA






AGTGAACCTGT






AACCTACCACG






GTATCTATCAT






AACACTAGACT






TTCAG







 34.
Mm.22753
Chromosome 14
CATCCTACAAA






GAGGATAAGC






ACTTTGGGTAC






ACTTCCTACAG






CGTGTCTAACA






GTGTGA







 35.
Mm.143774
Chromosome Multiple
CCTGAAAATCT





Mappings
GTCATGTCCAC






CTTGGAGCCTG






AGTAACTTTGA






ACAGCTGGTAA






CTAGT







 36.

Chromosome 17
AGTCAAGGAG






CCTAAAGATTA






TTATGTCAGAG






AGACCAGCTTT






AGATACACCCC






TGAGCA







 37.
Mm.19325
Chromosome 10
TTATGCTGCAG






TTTCACTTGGA






AAAGGGACAA






GGAGCCTTCTA






TTGTCCCCTGT






TTGTAG







 38.
Mm.100525
Chromosome 9
GTAACCAAGA






GCCCTGAATAA






GGAATTCATTG






TAGTAGTGAAA






GGGAAACTAA






TGCTCTT







 39.
Mm.32810
Chromosome 4
TCCCATGCCTT






CCCAGAGGGA






ATTTTAACAAT






GTAACAATAA






ATGCTTGGCCT






TGAAGCT







 40.
Mm.182645
Chromosome 9
AGGACATCTTC






CCAGATCTCAA






AAGAAGAAGA






GAGCCTGTAAC






CACCTCCATGA






CCTAAA







 41.
Mm.262549
Chromosome 6
TCCTGTGGGAG






ATCCCATAAAT






CCTGAACCTCA






CGTAGTGTTAC






TTTTCCAGGTC






ATTCT







 42.
Mm.253853
Chromosome 13
CGACGACGAG






TTCGAAGACGA






CCTGCTCGACC






TGAACCCCAGC






TCAAACTTTGA






GAGCAT







 43.
Mm.171544
Chromosome 12
GAAGAGATGG






AAGATGGTAGT






GCCTTGAACAC






AGCCACCCAA






GCAAAGTTGA






AGAACAGG







 44.
Data not found
Chromosome 9
GCCTGCAGGA






GTTTGTGTTGG






TAGCCTCCAAG






GAGCTGGAAT






GTGCTGAAGAT






CCAGGCT







 45.
Mm.1682
Chromosome 2
CTGTCTTCTAA






TTCCAAAGGGT






TGGTTGGTAAA






GCTCCACCCCC






TTTTCCTTTGC






CTAAA







 46.
Mm.123240
No Chromosome location
TTCACAGGGTT





info available
CCTGGTGTTGC






ATGCAGAGCCT






GAACAAAAGA






CTCAGGTGGAC






CTGGAA







 47.
Mm.41932
Chromosome 17
TCTACAAGGAA






GCATTCAACCA






CCAAGAGGAG






CTTGGACCACG






TTCACTCTGTA






TTCTTT







 48.
Mm.173544
Chromosome 4
GGGCCTGAACT






ATGGCTTAATT






TACATTAATTA






GTTAACATTAA







 49.
Mm.149539
Chromosome 2
TCACACAGTAA






GGAGC






TGTGTTGTGAT






TTCAACTCCCA






AGACGCCCTTT






ATGTCCATTCT






GGAAAAATAC






AATAAA







 50.
Mm.370
Chromosome 4
ACTGATGTTTC






TGCACACTGCC






CAGTGGTTTCT






TTAAGCACTTT






CTGGAATAAAC






GATCC







 51.
Mm.28614
Chromosome 1
TCACAGATGTA






TGTGGAGGGGT






TGTTTTCTGAG






TACTAGACTAC






CCTCTGTGGTT






ATAAA







 52.
Mm.24145
Chromosome 12
TCGGGGATGG






AGCTGAGATGT






TCCACCACAAC






CCAAGATCTAA






GAGTATTGTTT






TGAAGA







 53.
Mm.159218
Chromosome 16
GGAGACTGAA






GCTTTTATTGT






TTAATGTTGAA






GATATTGATCT






ACAAGGTGGG






AATGGTG







 54.
Mm.217664
Chromosome 2
AACTGTGGGTA






TAATTGTAAGA






GCCTGAAACTT






CCAGAACTGG






AGAAACTGTCA






CTGGGA







 55.
Mm.221743
Chromosome 17
GTGTTGTGATT






GTCGTCCCTGC






TTAATGAACCC






ACCTGAGGGA






CAGTTAGTGTC






TTACCC







 56.
Mm.206775
Chromosome 5
CTATATGAACT






GAGAAACAAC






ACGTATGCTGA






ACCCCAATTCT






ACAACAAAGT






CTACGCC







 57.
Mm.32373
Chromosome 3
GGAATATATTA






TGTAGACTATT






CTGGCCTGAAC






CTTGTGGTTGA






CTGATGCTCTG






CCTCC







 58.
Mm.261771
Chromosome 14
TTGGGTGATCC






ATATTTTTCAA






ACCCATACTCC






CAAAAGGAGA






CCTACTTAAAT






TTCTCT







 59.
Mm.252843
Chromosome 10
GTTCCTGAAGC






TCTTGATATTT






TAGGACAAAA






CCCACCACGAC






AAAATGAGAA






GGAATTT







 60.
Mm.27451
Chromosome 7
TGACTTCAAAT






GTCCCATCCCA






CCCAAAGAGC






CTGTGATAACA






GATGTCTCTGG






CTATAT







 61.
Mm.15781
Chromosome 7
TGGGTAGGTTC






CTAGGTCTCCC






TGATATCTAAG






CTACAGTTATA






CTGTAGCTGTG






TGACA







 62.
Mm.170657
Chromosome 19
CCTGTCTCAGA






ACTCAAAGAAT






AAATCCAGTGT






ATCTTCAGAGT






CACTTTGTAAC






CCTAC







 63.
Mm.152120
Chromosome 6
TACTCCCTGGA






GACTAGAACC






GTGGCTATAGC






GGAGCATGCTC






CAGAGCACAG






GACTGAT







 64.
Hs.5831
No Chromosome location
GGGACACCAG





info available
AAGTCAACCA






GACCACCTTAT






ACCAGCGTTAT






GAGATCAAGA






TGACCAAG







 65.
Mm.389
Chromosome 15
GAAAACCAAA






ACTCTTGGTCA






GAGACAATAT






GCAAAACAGA






GATGTCAAGTA






CTATGTCC







 66.
Mm.86910
Chromosome 1
TCAAGGAGACT






GTAGACTTAAA






GGCAGAACCC






CGTAACAAAG






GGCTCACAGGT






CATCCTC







 67.
Mm.9537
Chromosome 2
CACCACGGACT






ACAACCAGTTC






GCCATGGTATT






TTTCCGAAAGA






CTTCTGAAAAC






AAGCA







 68.
Mm.22650
Chromosome 12
GTACCCTCTGA






CTGTATATTTC






AATCGGCCTTT






CCTGATAATGA






TCTTTGACACA






GAAAC







 69.
Mm.221600
Chromosome 5
AAGAACTACTG






ATACAGAACC






ACTTCAGTTGT






TCAGTTAGAAT






CTTTTTAAGAC






TCTCTC







 70.
Mm.173358
Chromosome 2
CTTGACCTTTA






GATGGAAATTG






TACCTAGAGAC






GAGAAGGAGC






CAAACTAAGGT






CTGTCA







 71.
Mm.2534
Chromosome 9
GGAACGGACA






ACGTGGCTTTG






TCCCTGGGTCG






TACTTGGAGAA






GCTCTGAGGAA






AGGCTA







 72.
Mm.28280
Chromosome X
TTCGAATGCAC






ATCATTGACAA






GTTTCTCTTAT






TGCCTTTCCAC






TCTGGATGGGA






CCCTG







 73.
Mm.23172
No Chromosome location
GCCTGGAGACT





info available
GAAGGCAGTTT






TACAAAGGAA






AACTTAGATTT






CTATTCATTTG






CTTTTG







 74.
Mm.10809
Chromosome 1
CTGGATGAAG






AAACAGAGCA






TGATTACCAGA






ACCACATTTAG






TCTCCCTTGGC






ATTGGGA







 75.
Mm.23955
Chromosome 5
TTAATATTGTC






AATGTCAGGG






GGTTCCCTGTC






TCAGAGCATTA






TGTGTACTAAC






TGTAGC







 76.
Mm.268680
No Chromosome location
CCAGAGTTTTT





info available
TCCATCATGTT






TTGCCCCAAAG






ACCTCGGTTTG






TAGAAGCCCA






AGGAAA







 77.
Mm.90241
Chromosome 6
GACAGGGTCA






ATGTTTATTAT






ACATACTGCAC






TGATGAGAAC






AATATCATATG






TGAAGAG







 78.
Mm.230635
Chromosome 8
ACTCTCAGCTT






CCTGTTGGCAA






CAGTGGCAGTG






GGAATTTATGC






CATGTAAATGC






AATAC







 79.
Mm.270136
Chromosome 7
GACAGGGACT






CCATATGGAAG






TAAGGACGTTT






ACCTCATTACT






AAGTCTCGTCA






AAAGAA







 80.
Mm.266485
Chromosome 15
CTCGGATCTTC






ATGTTCTTCAG






TAAGAATCTCT






CTGTGGATTTG






GAACAATCGTA






AATAA







 81.
Mm.32929
Chromosome 7
CTAAGACACCT






GTGATTTGGCA






ACTGGTCAATT






CATGCTTGTTA






CATTCAGAACT






CAGGA







 82.
Mm.145
Chromosome 11
TCCCTCTCTGT






GAATCCAGATT






CAACACTTTCA






ATGTATGAGAG






ATGAATTTTGT






AAAGA







 83.
Mm.288474
Chromosome 5
TTCTCAGTTCA






GTGGATATATG






TATGTAGAGAA






AGAGAGGTAA






TATTTTGGGCT






CTTAGC







 84.
Mm.18626
Chromosome 6
CTGACCAAGGT






GGCTGACTCCA






GCCCTTTTGCC






TCTGAACTGCT






AATTCCAGATG






ACTGC







 85.
Mm.173282
Chromosome 4
GATACCTGGCT






TATCTTTTATC






AACAGCAAATT






ATGCAGTGGTG






GAAATGTCATC






ACAGA







 86.
Mm.76649
Chromosome 3
GTTTGAGAAGA






GACATTATTTA






TAAAACCCAG






ATCCTTAATAC






TGTTTATTACA






GCCCCG







 87.

Chromosome 13
CTCTGATACTG






AATAAACCTGA






TGTGATGTACT






TATAGTCCTTA






AGTCTTGAGAG






TTAGA







 88.
Data not found
Chromosome 3
GGCAACTACG






ACTTTGTAGAG






GCCATGATTGT






GAACAATCAC






ACTTCACTTGA






TGTAGAA







 89.
Mm.1643
Chromosome 19
ACTTCATAGGA






TTCACAATGGA






GAGGGCTAGG






AAGATACTGG






ACAATTTTCAG






CAGTGTG







 90.
Mm.247493
Chromosome 4
CACCTCTTGTC






TCCAGCCATGC






CCAGGATCAAT






TCTAGAATCAG






AGGCTACCCCT






GCCTG







 91.
Mm.182599
Chromosome 16
CGTCAGTGACC






CACTCAATACT






GTGGTGGGAA






GTAAGATGATG






CCAAATCTATA






ACCTGT







 92.
Mm.4159
Chromosome 2
CGAATGAGAA






TGCATCTTCCA






AGACCATGAA






GAGTTCCTTGG






GTTTGCTTTTG






GGAAAGC







 93.
Mm.259061
Chromosome 10
CCGGCGGGCCC






TAGTTTCTATG






TATTTAGAATG






AACTCGTGTAC






ATATGTAAAGA






TCTTT







 94.
Mm.27498
Chromosome 6
CAAGCTGGTTG






GAGCCTCCAGC






CTTCAAAATTC






TGAATCTAATA






AACATTAATGC






ACACT







 95.
Mm.267078
Chromosome 5
CAATCCTAGAA






CAACTACTTGA






GTGTTGTGAGT






GTTCAGATACT






CATTAATATAT






ATGGG







 96.
Mm.24457
Chromosome 4
TCCCACCTCTC






TGATGAGTTAT






AGCCAAGAAG






CCTTAGGAGTC






TCCATAAGGCA






TATTCA







 97.
Mm.116862
Chromosome 3
AAGAAATATTC






CCACTTCAGAG






TGTGTAAGCAA






TATTTAAACCC






AGATAAAGAT






GCATGC







 98.
Mm.196869
Chromosome 5
TTTGGGAGTGG






GCTTCATGAAT






GCGCTCTTACC






AAAGGAGCCA






TGTTTCCATTG






TATCAA







 99.
Mm.21013
No Chromosome location
TTTCATTAAAC





info available
TAATATTTATT






GGGAGACCAC






TAAGTGTCAAC






CACTGTGCTAG






TAGAAG







100.
Mm.153315
Chromosome 1
AAGTGACTCCA






TTTTCATATGT






ACTTAAACACA






GAGTTCCTGTG






GCCTCTGTAAG






CTCAG







101.
Mm.22479
Chromosome 15
CAAGGTGAAG






AGCCTGGAAA






CTGAGAACAG






GAGACTGGAG






AGCAAAATCC






GGGAACATCT







102.
Mm.44876
Chromosome 3
GCATGTGATTG






ATTCATGATTT






CCCCTTAGAGA






GCAAGTGTTAC






CAAAGTTCTGT






TGAGC







103.
Mm.290934
Chromosome 12
TGCTCCAGATG






TGAAACTTATA






GACGTAGACTA






CCCTGAAGTGA






ATTTCTATACA






GGAAG







104.
Mm.221788
Chromosome 2
TGTACAACTGA






ACTCACCTCTT






GTGAAGAATTA






TGATTGTCTTA






CTTGTAAAGAA






AGCAC







105.
Mm.33498
Chromosome 16
TTTTGCAGGGG






TCGAGTGTGAT






GCATTGAAGGT






TAAAACTGAA






ATTTGAAAGAG






TTCCAT







106.
Mm.87180
Chromosome 7
CAAACAGAAA






ACAGGGAGAT






GTAAAACAGTT






TCAACTCCATC






AGTTATGAAAC






CATAGCT







107.
Mm.133615
Chromosome 9
TCAGCAAATTG






GCGATTTCGGA






ATCCTATGACA






CCTACATCAAT






AGGAGTTTCCA






GGTGA







108.
Mm.1956
Chromosome 14
CATGTGCAACC






TCATGGGAAA






AATAGTAACTT






GAATCTTCAGT






GGTTAGAAATT






AAAGAC







109.
Mm.60230
Chromosome 17
GTCTCAAGGAT






CTGGGACCAG






AACTGGGAAA






GAAAAGGAAT






GACCAAGACA






AGATCATAC







110.
Mm.143141
Chromosome Un
TGAATCAGAG






AAAAGAGAGT






TGGTGTTTAAA






GAATATGGGC






AAGAGTATGCT






CAGGTGAC







111.
Mm.40268
Chromosome 3
AAAGGAAATC






ATATCAGGATA






AGATTTGTATC






TGATGAGTATT






TTCCATCTGAA






CCCGGA







112.
18413
Chromosome 11
CAGTCCTCTTG






AAAGGTCTCAG






AAGCTGGTGA






GCAATTACTTG






GAGGGACATG






ACTAATT







113.
Mm.2877
Chromosome 16
AGAGGAGTCTC






CTTATATTAAT






GGCAGGCATTA






TAGTAAAATTA






TCATTTCCCCT






GAGGA







114.
Mm.297591
Chromosome 11
GCATGAGTGTA






TAGGTGAAGGT






TTCACTTTAAG






ATGCTGTCTTC






AGTTCTCTTGC






CTATG







115.
Mm.2284
Chromosome 9
ATCGTCTCTGA






TTATGACAAGG






GCTATGTGGTG






TGGCAGGAGG






TATTTGATAAT






AAAGTG







116.
Mm.15622
Chromosome 4
CTGTTCGTGTT






GGGTTTTGTTC






ATGTCAGATAC






GTGGTTCATTC






TCAGGACCAA






GGGAAA







117.
Mm.196692
Chromosome 10
GTGCAATAGA






AATATATGATT






TCAAACACATT






TCTGAACTGCC






AGGGCAAGAA






AGTATAG







118.

No Chromosome location
CTTGTCGTTTT





info available
TGGGGGTTGTA






ATATCTAAGGG






TGAAAAAATTA






ATTTCCAAAGC






CAAGA







119.
Mm.29268
Chromosome 4
CAACTGTTTAC






CTGGAAATGTA






GTCCAGACCAT






ATTTATATAAG






GTATTTATGGG






CATCT







120.
Mm.220988
Chromosome 8
CCTTCCAGAGC






TTTGCCAAATT






TGGAAAATTTG






GAGATGACCTG






TACTCCGGATG






GTTGG







121.
Mm.173383
Chromosome 2
TAGGTGAGTTA






GGAATCTGCCA






TAAGGTCGTTT






ATAGGATCTGT






TTATATGAAGT






AATGG







122.
Mm.249937
Chromosome 8
ATGACTTTCTC






TGCTTGGTTGG






AGAAGAAGAA






TCTTTACTATT






CAGCTTCTTTT






CTTTTT







123.
Mm.28559
Chromosome X
CCGGGGTGGG






AAGTTGTTTTT






TCCTGGGGGTT






TTTTCCCCTTA






TTTGTTTTGGG






GCCCCT







124.
Mm.38094
Chromosome X
GGAAGATGGG






TAAATAGTAGA






CTGTGGTGTAT






TTGGAACAAG






GTAGCTTTAAA






GACACAA







125.
Mm.41694
Chromosome 5
CCAGGTTCAGA






GCGGACTGCTA






ATAATAATGTG






TGTATTGATCG






AGGAAAAAGT






GCGGAG







126.
Mm.32947
Chromosome 14
TGCATGGGAA






ATTTCTACGTG






GCTCACTTCAC






CAAGGCTTATT






GCACTGGGAA






AAGAAGA







127.
Mm.486
Chromosome X
TTAACCTAAAG






GTGCAACCTTT






TAATGTGACAA






AAGGACAGTA






TTCTACAGCTC






AAGACT







128.
Mm.5624
Chromosome 17
TCCCCACTACT






ATAAGGCCAA






GGAGCTAGAA






GATCCCCATGC






TAAGAAAATG






CCCAAAAA







129.
Mm.6888
Chromosome 19
ATAGGTACTCC






CCGATTCCCAA






GGAGCAGCTA






GTGGAACCCTG






GAGTTTTGGGT






AGTAGA







130.
Mm.2271
No Chromosome location
AGTAGTATTTC





info available
CAGTATTCTTT






ATAAATTCCCC






TTGACATGACC






ATCTTGAGCTA






CAGCC







131.
Mm.1155
Chromosome 1
ACCGCTACTTG






GAGCCTGTTCA






CTGTGTTTATT






GCAAAATCCTT






TCGAAATAAAC






AGTCT







132.
Mm.8856
Chromosome 17
TGAACTCTGAC






CTTTTGCAACT






TCTCATCAACA






GGGAAGTCTCT






TCGTTATGACT






TAACA







133.
Mm.2580
No Chromosome location
GTCTGTTCTTG





info available
GGAATGGTTTA






AGTAATTGGGA






CTCTAGCTCAT






CTTGACCTAGG






GTCAC







134.
Mm.35104
No Chromosome location
CCAGCCTGACC





info available
AGATTTTAGTT






ACCTTTTAAGG






AAGAGAGATTT






ATTCTAATGCC






ATAAA







135.
Mm.22673
Chromosome 1
CACCTCTGTGC






TTTGAAGGTTG






GCTGACCTTAT






TCCCATAATGA






TGCTAGGTAGG






CTTTA







136.
Mm.2720
Chromosome 2
CTGAGCTCAGG






CTGAGCCCACG






CACCTCCAAAG






GACTTTCCAGT






AAGGAAATGG






CAACGT







137.
Mm.4487
Chromosome 7
AGAACAGCAG






TTAGTTCCTGG






TTCTGAGAACC






ACTTGTCCCAG






TATGACACCTC






TTACTA







138.
Mm.4348
Chromosome 12
ATGTGTGTACT






CAGGACAGAA






TCCAGAGATTT






CTTTTTTATAT






AGCTTGATATA






AAACAG







139.
Mm.448
Chromosome 8
ACGTTTCACAC






AGTGGTATTTC






GGCGCCTACTC






TATCGCTGCAG






GTGTGCTCATC






TGTCT







140.
Mm.23942
Chromosome 11
TTTTTTAATTCT






GCAAATTGTCT






CACAGTGGAAT






GAGGAAATGA






GTTAGAGATCA






CAGCC







141.
Mm.1109
Chromosome 6
GTGCTATCTTT






ACTCACTCCCA






AGACATACAC






AGGAGCCTTTA






ATCTCATTAAA






GAGACA







142.
Mm.2692
Chromosome 3
GAGGTCCAAGT






TTAAATGTTAG






TCTCCTAACAA






CTGTCAAATCA






ATTTCTAGCCT






CTAAA







143.
Mm.21630
Chromosome 9
CTTCTAGATCC






TTCTGCAGAAA






TCATCGTCCTA






AAGGAGCCTCC






AACTATTCGAC






CGAAT







144.
Mm.17537
Chromosome 18
ACTTATTCATC






CTTGCCTATAC






CCACCCCCCAA






AAACAGGTTTT






ATTAATAAAAA






ATGTG







145.
Mm.1585
Chromosome 13
TACAGTAACAA






GCAAGCTATCA






TCCATTTTTAC






AATAAAGTTGT






CAGCATTCATG






TCAGC







146.
Mm.103104
Chromosome 15
TTATTTACTTT






ATCTTAGTATG






TAACCTTAGCT






GACCTGAAACC






CACTGGTAGAC






TAGAC







147.
Mm.206536
Chromosome 4
CCTGTCCTGAG






TTCATGGCCAA






AACTTAAATAA






GAGAAGGAGG






AGAGGGTCAG






ATGGATA







148.
Mm.156600
Chromosome 6
AAAGGGGCCT






GAGTATACGCT






GTTGCAAGCTG






TATACTTCATT






TCCTTCGGCTG






GTTTAT







149.
Mm.254385
Chromosome 3
TATCCGGACAG






TCTATGTGAAA






TAGGACCAAG






GTCGAAAGCC






GGAAAGACAT






CAACAGAA







150.
Mm.2570
Chromosome 4
CTGCTTTTCCC






TGACATGGATG






CGTAATCACGG






GGTCAAATTAC






ACCTATCCAAC






ACCAT







151.
Mm.153911
Chromosome 14
AACAAAGAGG






ACAGTATGAAT






TTGAATAGCTC






CCACTAGATAA






GCAATTTCCAC






GAGAAC







152.
Mm.28130
Chromosome 1
CTGACTGTGAA






TGTCGTGACTC






AGAGCAAAGA






CAGAGAATAT






ATTTAATTCAT






GTTGTAC







153.
Mm.3267
Chromosome 14
GCCTGAAGAA






CATGACAGAA






CTCTTCTCAAT






ATTCGTTGGGC






TTTCAGAATCA






TAAACAT







154.
Mm.45436
Chromosome 10
CCTGTGTGAAT






AAAAATACAA






GAACTGCTTAT






AGGAGACCAG






TTGATCTTGGG






AAACAGC







155.
Mm.200362
Chromosome X
GTAAGAAATAT






TAGACTGATTG






GAGTTAAAGTA






GCACTCTACAT






TTACCATGGTG






TTTGG







156.
Mm.143819
Chromosome 1
TGTGAAAGATT






GTGCATCTGCA






TTCAACTACCC






TGAACCCTTAG






GGAAGAAATG






GATTCC







157.
Mm.27923
Chromosome 11
AGCTGCCTACT






AGCAGTTTAAC






AAGGAGCCTTG






CTGTCTCAGAC






AGGTGAAAGA






AAATGT







158.
Mm.40894
Chromosome 3
CCATGTTTGAA






AGTATGTAATG






AAGAGGAGCC






TATTAACCATA






TGAAAGACAG






GAATACT







159.
Mm.160389
Chromosome 7
GTGAATTGGAT






GCATAGCATGT






TTTGTATGTAA






ATGTTCCTTAA






AAGTGTCACCA






TGAAC







160.
Mm.142524
Chromosome 8
ACCCACTGACT






AGGATAACTG






GAAAGGAGTC






TGACCTGAATG






ACGCATTAAAC






TCCTGCA







161.
Mm.2970
Chromosome 14
CCCGCTTCAAT






GAGAACAACA






GGAGAGTCATT






GTGTGTAACAC






GAAGCAGGAC






AATAACT







162.
Mm.24138
Chromosome 2
ATATTAACTCT






ATAAAATAAG






GCTGTCTCTAA






AATGGAACTTC






CTTTCTAAGGG






TCCCAC







163.
Mm.275894
Chromosome 14
TGTGGGTTTTT






TGAAGAATTAA






TGAGCATGTAC






ATAGAAATAGT






GACTGCTTGAA






TCCTG







164.
Mm.566
Chromosome 5
CTACTCTTAAT






GATGTTATCTT






AACACTGAAAT






TGCCTGAAACC






CATTTACTTAG






GACTG







165.
Mm.40268
Chromosome 3
TCGACCATTTC






TAGGCACAGTG






TTCTGGGCTAT






GGCGCTGTATG






GACATATCCTA






TTTAT







166.
Mm.24208
Chromosome X
TCTGAATCTGG






GCACTGAAGG






GATGCATAAA






ATAATGTTAAT






GTTTTCAGTAA






TGTCTTC







167.
Mm.34490
Chromosome 11
GATCCTTAGGT






CTCCATAGGAT






GATTTTTGAGG






TAGTTAATCAG






TGTAAACTCTT






ACACA







168.
Mm.9749
Chromosome 19
CTCAGCAGTAA






CAGAGAAAAG






ATGAATGAAG






CCACTGAGGCT






TCGTGAATGAA






TGAATCT







169.
Mm.154804
Chromosome 8
CTTTGTTCCTA






CCCAGCCACCA






AAGCCACCTAC






ATAACAATCCA






CTCATGTACTA






GCAAA







170.
Mm.90787
Chromosome X
AAATTGTCTAC






GCATCCTTATG






GGGGAGCTGTC






TAACCACCACG






ATCACCATGAT






GAATT







171.
Mm.36217
Chromosome 6
ACATGATGTGA






AAGAATCATTG






AAGATCACAGT






TGTCTACCGAG






TTCAGATTTCC






TTACA







172.
Mm.1834
Chromosome 4
CACCCCCCAGA






AAATGAGACT






ATTGAACATTT






TCCTTTGTGGT






AAGATCACTGG






ACAGGA







173.
Mm.214593
Chromosome 9
AGTGATGGGG






ACCATGACGA






GCTGTAGCCTG






AACCTCAAGGC






CTGCAACCAGT






CTACTGA







174.
Mm.24045
Chromosome 12
AAAGGTCCCA






GGTTTCGATCT






GTTTGGAGTTT






GGAGTCTAATG






GTTGCATAGAT






AAACAG







175.
Mm.18517
Chromosome 8
TCTATGTGCAT






TAGGGGGTGA






CCCAGGGAAA






TCCAAAGGGA






ACAGTATTTGA






TTTCTCAC







176.
Mm.249873
Chromosome 5
CTACACATGTA






CTTTAGGATTC






TAGGTTTCTCC






CTGAGCCCTGC






TTTCGATGTAA






CACTG







177.
Mm.2128
Chromosome 3
AAGTCTAAAG






GGAATGGCTTA






CTCAATGGCCT






TTGTTCTGGGA






AATGATAAGAT






AAATAA







178.
Mm.254240
Chromosome 15
GGAAGAAAAA






GACCTCAGGA






AAAAATTTAAG






TACGACGGTGA






AATTCGAGTTC






TATATTC







179.
Mm.19185
Chromosome 19
GGATGAAGAA






ACTGAGTTTGT






CCCTTCTGAGA






TCTTCATGCAC






CAAGCAATCCA






CACCAT







180.
Mm.10222
Chromosome 15
CTGTTCAGGCT






CAAACAATGG






GTTCCTCCTTG






GGGACATTCTA






CATCATTCCAA






GGAAAA







181.
Mm.141936
Chromosome 1
AGGAGTTCCCA






GTTTTGACACA






TGTATTTATAT






TTGGAAAGAG






ACCAACACTGA






GCTCAG







182.
Mm.18821
Chromosome 17
CTCAATAAAAG






CTCTAAGGAGA






CATCACAACCC






AGTCTTAAGGG






TTCATGAGGTT






TTAAT







183.
Mm.29487
Chromosome 19
ACTTAAAATGT






AGACTGTTCAT






ACAGTGGGTAC






CAGTATGAGTT






GAATGTGTGTA






TTACT







184.
Mm.117294
Chromosome 10
TTTCATAATAG






AACCGTCTACC






AGTGACCTCTT






GATTATGATTT






GATTTGACTGC






AAAAC







185.
Mm.114683
Chromosome 3
ATCCATGTGGC






ATCAATTCAAT






TATGTATAATA






ATGACTTTACA






AGGGCCCCTTA






AAACC







186.
Mm.21596
Chromosome 6
CACAAAAGTC






AAATGTGGATA






TCGTACGCTGC






ATCACGTCATA






GACAAGTCTAA






AGAAGA







187.
Mm.4213
Chromosome 6
CTATCAGGATA






GTGATAAGAA






CGTCATTCTCC






GACATTATGAA






GACATGGTAGT






CGATGA







188.
Mm.212279
Chromosome 6
GGAGATCATCA






CTCTTGTATGA






AATATACTAAC






TCCAAACCTTT






TTAGAGCAGAT






TAGGC







189.
Mm.89568
Chromosome 12
ACTATTAAGCA






CTCAGGAGAAT






GTAGGAAAGA






TTTCCTTTGCT






ACAGTTTTTGT






TCAGTA







190.
Mm.10878
Chromosome 5
AAAGAGAAAA






TATGTCAGATG






GTGATACCAGT






GCAACTGAAA






GTGGTGATGAA






GTTCCTG







191.
Mm.980
Chromosome 4
GAGAGAGGAA






TGGGGCCCAG






AGAAAAGAAA






GGATTTTTACC






AAAGCATCAA






CACAACCAG







192.
Mm.37773
No Chromosome location
GTTGTACTACT





info available
GGAAAGATTTT






GCTGGGACATA






CAATATGTGTG






AGAAAAATAG






AGTTGT







193.
Mm.24498
Chromosome 5
AGACCAAAGA






CACGGACATTG






TGGATGAAGCC






ATCTACTACTT






CAAGGCCAAT






GTCTTCT







194.
Mm.28406
Chromosome 19
CAGAGCAGGG






GGCTTTTATTT






TTATTTTTTAA






TGGAAAATAAT






CAATAAAGACT






TTTGTA







195.
Mm.39856
Chromosome 7
CTTGGCAGCTC






TCCTTACTTCT






GGGACATTTGC






CACTGTGGTAC






TGCCAGGAAG






GAATCT







196.
Mm.275813
Chromosome 12
ACTTATAGAAA






AGGACAGGTT






GAAGCCTAAG






AAGAAAGAGA






AGAAAGATCC






GAGCGCGCT







197.
Mm.684
Chromosome 7
TAGTTCAGTGA






ACAAGTATCTG






TCAATGAGTGA






GCTGTGTCAAA






ATCAAGTTATA






TGTTC







198.
Mm.217227
Chromosome 2
CATGAATGTCA






AAACCTAATTA






CAAAGCATCG






GTCTCTTTGTT






GTGAGGTATCA






GAACCC







199.
Mm.202348
Chromosome 15
CCTGTCTCATG






GGAGATTTGAA






TCATAAGGAG






AATCACTTTTT






GTAACTTTATT






GAGGAA







200.
Mm.6272
Chromosome 9
AAGTAAATATG






CAAAGGAGAG






AAGTTAGAGA






AACTCCTCTCA






TAAGAAAAAT






GTCTTCCC







201.
Mm.254859
Chromosome 17
TCGGAACTGTC






CCTTAAGGAGG






GTGATATCATC






AAGATCCTCAA






TAAGAAGGGA






CAGCAA







202.
Mm.8249
Chromosome 1
TTAGTGGGCTG






AACCTATCGGT






TTTAACTGGTT






GTCTTAATTAA






CCATAAACTTG






GAGAA







203.
Mm.147226
Chromosome 8
TTTTGTACAAC






CCTGACTCGTT






CTCCACAACTT






TTTCTATAAAG






CATGTAACTGA






CAATA







204.
Mm.10727
Chromosome 8
AGACTTGGAA






AAGGCTTGGGT






ACAATTAAGA






AAAACCCTACA






TCCCACCCTCC






TCTTGAC







205.
Mm.221768
Chromosome 6
TAATAAAGAA






ACTGTGGAAAT






ACTTGGATTTC






TACTGAAGACA






AAAGACTTCTA






GGCTGG







206.
Mm.214742
Chromosome 10
AGGTTAAACAT






ATATTCTTGGA






AACATGAAATC






ACAACTCTCAA






AAACCGTGAA






CCACCA







207.
Mm.1359
Chromosome 7
CCTCGTGTTGT






CTTCTTTGGAC






CTCAGTTTTTC






CATGAACCAG






AAGAGAATTG






GAACAAG







208.
Mm.217839
Chromosome 10
AATAGCAATGT






ATCAAACAATG






GATGTGAAAA






AGATGCGCTCT






ATCATCATGAA






AATGCC







209.
Mm.4619
Chromosome 17
TCTCTGGAGAA






ATCAGTAACTG






CAAAAGGAAG






AGAGGGTCTTT






AAAGCACATGT






AGTAAT







210.
Mm.173427
Chromosome 2
TGGAATGTTGA






AGAATGAAAT






CTCGAGGGAAT






TAGAGGTTGAG






GTCATCTGGAT






ATTCAG







211.
Mm.27789
Chromosome 6
ATAGAACCAAT






GTAGGAAAAT






CAGGCAAAAT






AAAATGATGAT






CAGTCCATGTC






ATCATGG







212.

No Chromosome location
AGATGGGAAA





info available
AAGTACTGTAG






GTTCCTGAACT






CTGGATCTCAA






GCAGAAATGT






ACTGTCT







213.
Mm.221816
Chromosome 18: not
AGGAAAACCC





placed
CGGTAGTTAGG






ACATCTGAATT






CTCAATTATTG






GATTGCCAAAA






GTGAAA







214.
Mm.273506
Chromosome 2
GTTTTTGGAAT






TTGGACCTGAA






AATTGTACCTC






ATGGATTAAGT






TTGCAGAATTA






GAGAC







215.
Mm.21104
Chromosome 17
TGGGACCTGTG






AAGCGACTGA






AGAAAATGTTT






GAAACAACAA






GATTGCTTGCA






ACAATTA







216.
Mm.182542
No Chromosome location
TCCATTATTAC





info available
ATACAACAATC






AAGAAAAAGA






CAGAAAACTA






CCCTTAGAGAG






ATCAGGG







217.
Mm.260378
Chromosome 4
ATTCAACAGCA






TTCTAGGAAAA






TGGCAAGAAA






GTAAATTATCA






TCCATTTCAGG






TCTGTG







218.
Mm.260433
Chromosome 18
CCATATGATCA






CAGTCGTGTTA






AACTGCAAAGT






ACTGAAAATG






ATTATATTAAT






GCCAGC







219.
Mm.29216
Chromosome 18
GGGCCATATTT






TAAAGATAAG






GAGAGAGAAA






CTAGCATACAG






AATTTTCCTCA






TATTGAG







220.
Mm.248291
Chromosome 1
GAAAGGCGTTT






ATTCAGAAAAT






GATGGTAAGAT






TCAGACTTTAA






AGCACAGTTAG






ACCCA







221.
Mm.2681
Chromosome 13
TAAGGTGTTTT






CTCCAGTTAAG






TTCAGTTCCTG






AATAGTAGTGA






TTGCCCCAGTT






GCAAC







222.
Mm.119383
Chromosome 2
CCACCATAAAG






GAAAAAGGAC






ATGTGTATGAG






TAGGTGTTCAT






CTATGTGCATA






ATTGGC







223.
Mm.263733
Chromosome 12
GCACAAGATG






GAGTCATTAAA






ATTAAGGCATC






ATCATTTTCAG






CATATAACATA






GCAGAG







224.
Mm.221860
Chromosome 1
GATTAAAAAC






ATTAGGGATGA






GAAATAATAA






GGGCTTGCAAC






TGTGTAGAAGC






TAGAGCC







225.
Mm.46455
Chromosome 4
TGAAGTACACT






CTCTAAATGAA






AATGGGCTATA






AATATGTTTGA






GTAGGATAGG






AGGAAG







226.
Mm.5522
Chromosome 9
GTGTAAGAAA






AGATGGGACT






GACAATAAAA






ATGAAGGTCA






GGTAAGAAGT






ACCAGACTCC







227.
Mm.25836
Chromosome 16
GGGAAATATG






CAGCGTTCTAT






GTTTCCATAAG






TGATTTTAGCA






GAATGAGGTAT






TATGTG







228.
Mm.1401
Chromosome 1
GTAGGACTGTA






GAACTGTAGA






GGAAGAAACT






GAACATTCCAG






AATGTGTGGTA






AATTGAA







229.
Mm.222325
Chromosome 16
TCATAGGTCTC






CATTTAGTTCA






AGTGTTTTATG






GACAATCAGC






AAGTTTAGGCT






CATAGG







230.

No Chromosome location
TTGGAATATAT





info available
GAATGACAAA






GAAATGGGAA






AAACTGCTGAA






CCCGAGTCTCT






GAATGTC







231.
Mm.174026
Chromosome 10
CTATCTTGAAT






TGCTAGATTAA






AGAGAAAGAA






AATGTTAGAGC






AAAATAGGAA






CCTGGCC







232.
Mm.173446
Chromosome 9
AATCCCTAGAG






AAAATGGGAA






TAGAAATAAG






CTGCATACAAA






CTCAAAGACAC






AGATACT







233.
Mm.182873
Chromosome 1
AGACTGAAGA






AAACCTTAAAA






TACCCAAAATT






CAGGGGAGAC






ATAGCAACTGA






GTCTCAT







234.
Mm.1781
Chromosome 11
AGAGGACTTCC






TGTCTGTATCA






GATATTATTGA






CTACTTCAGGA






AAATGACGCTG






TTGCT







235.
Mm.216167
Chromosome 10
ATGGAGATGTG






TAAACAGTAG






GACATTTCGAT






AACTATGTCAG






GTCAGTTCTTA






GTTCAG







236.
Mm.221604
Chromosome 12
GAGGCTATTAT






AAATAACCTGA






AATGCATATGA






GAACTGAACGT






GTAATAATTCA






GCTCC







237.
Mm.222320
Chromosome X
AAGTCGGAAT






ATGTCTTAGTG






TTCTTCTCACT






TAGCTCAGTGT






AAGATGGTAG






CTCAAGT







238.
Mm.1430
Chromosome 4
CACTTTTCTAT






GAAGAAAGCC






GTGTGTAAAGT






TTCCGTGACAG






TAGTAATGGAA






ATATCT







239.
Mm.24905
Chromosome 15
TGTAAGAATAC






AAGGTAAAAC






AAAATAGAGA






AATACAGGCAT






CATATCTGCAA






ATCGCCG







240.
Mm.221718
Chromosome 3
CAGAAACAGT






AGTATGGGGTT






AAATCACAATG






AGGGAAATTAT






AGGGATATGC






AGCCAAG







241.
Mm.217090
Chromosome 9
ACTGAAAGTTG






GGGAGATACA






TGTAATTTAAT






AGGATAGGGT






ACTTAGGTCCA






GACAACC







242.
Mm.102470
Chromosome 15
AAGCTGTTGAA






TATGGACGTAA






CTGTAAATCCC






AGAGTGTTTTA






TTTTGAGATGA






GAGTT







243.
Mm.217865
Chromosome 6
TTTATCAAACA






TGGAAACATCT






AGAGACTATG






GGAGAGAAAA






TGGGTTTTTAG






ATATGGG







244.
Mm.203206
No Chromosome location
GGAAGTTAATA





info available
GAACTGTTCAA






AATGTGAAAGT






GGAAATAGCG






TCAATAAGGA






AAGCCCC







245.
Mm.9714
Chromosome 11
AGTGTAGTTTT






CAGTGGACAG






ATTTGTTAGCA






TAAGTCTCGAG






TAGAATGTAGC






TGTGAA







246.
Mm.249965
No Chromosome location
GAAAGTGGGG





info available
AATGAAAAGT






ATAACAAAGT






AAAAAGAGAA






TTTCTAGGCCC






TTTAGGCCC







247.
Mm.11186
Chromosome 11
GGTTTTCTCTT






GTTTTATCATG






ATTCTTTTTAT






GAAGCAATAA






ATCCATTTCCC






TGTTGG







248.

No Chromosome location
CTTTTTGAGGT





info available
TTATTTTTCCA






CAGTTTTCATT






TGTTCATTAGG






CATTTTCCCTT






TTACT







249.
Mm.250102
Chromosome 9
AGTGTTTTTCT






TTAATTCTTGA






GGTTGTTATTG






TAATATTTACA






TATAGTGCAAG






AATGT







250.
Mm.138048
Chromosome 10
TAAAGTATCCA






CTGAAGTCACT






ATGGAAAACA






GCCTTTTGATT






TATGGACTATT






TAGCTC







251.
Mm.212863
Chromosome 19
GCCTAGTTTTT






TCAGCATCAAT






TTTGGAAAACC






TTAGACCACAG






GCATATTTCGT






CAAGT







252.

No Chromosome location
TCATTTTTCAA





info available
GTCGTCAAGGG






GATGTTTCTCA






TTTTCCGTGAC






GACTTGAAAA






ATGACG







253.
Mm.78729
No Chromosome location
CTGAAAATCAC





info available
GGAAAATGAG






AAATACACACT






TTAGGACGTGA






AATATGTCGAG






GAAAAC







254.
Mm.107869
No Chromosome location
GCGAGAAAAC





info available
TGAAAATCACG






GAAAATGAGA






AATACACACTT






TAGGACGTGA






AATATGGC







255.
Mm.36410
Chromosome 2
AGAAAGCTAT






GGACTGGATA






GGAGGAGAAT






GTAAATATTTC






AGCTCCACATT






ATTTATAG







256.
Mm.68486
Chromosome 12
ACAAAAAGGT






TACCTATGAAG






ACAGTGAAAT






AAGAGAGAAA






TGTTTAGTACC






TCAGGTTG







257.
Mm.249862
Chromosome 7
CTAAGGGAGG






AAATGTTGGTA






TAAAATGTTTA






AAAGAACTTG






GAGGCAAACTT






GGAGTGG







258.
Mm.182670
Chromosome 6
CCACATCATTG






GAAAGAAATA






CACTTATCTTA






ATTGCCATGGA






ATAGGAGCAT






GAAAGTC







259.
Mm.221086
Chromosome 4
ATGAGAAATA






CACACTTTAGG






ACGTGAAATAT






GGCGAGGAAA






ACTGAAAAAG






GTCTATTC







260.
Mm.269426
Chromosome 4
CCTGTGAACTG






AAAATGCAGA






TGATCCACAGG






CTAAATGGGA






AACCTGGAGA






GTAGATGA







261.
Mm.107869
No Chromosome location
GCGAGAAAAC





info available
TGAAAATCACG






GAAAATGAGA






AATACACACTT






TAGGACCAGA






AATATGGC







262.
Mm.25571
Chromosome X
TGGAGGAAATT






GATTGAAAAA






CGATTGGTCAA






ATCGAAAATG






GAGAAAACTC






ATGTTCAC







263.
Mm.103648
Chromosome 1
CTTCATCCTGG






TTTTCACGGCA






ATAATAATGAT






GAAAAGACAA






GGTAAATCAA






ATCACTG







264.
Mm.123225
No Chromosome location
ACTGAAAATCA





info available
TGGAAAATGA






GAAACATCCAC






TTGACGACTTG






AAAAATGACG






AAATCAC







265.
Mm.871
Chromosome 11
CAAGCACTGTG






CTGCAAAATGT






CGGTGGAATAT






GATAAGTTCCT






AGAATCTGGAC






GAAAA







266.
Mm.217877
Chromosome 1
TTTGAGAAGAA






AGGCATACACT






TGAAATAAAG






GCAAAAACATT






ATACTGTCTAC






CGAGAC







267.
Mm.38058
Chromosome 13
GAAGAAAACG






AGGTGAAGAG






CACTTTAGAAC






ACTTGGGGATT






ACAGACGAAC






ATATCCGG







268.
Mm.43952
Chromosome 13
ATCATAAAAAC






TGTGGAAATCC






ATATTGCCCTT






TTAAAAGAAA






ACTATGGGGAT






GGAGAG







269.
Mm.8709
Chromosome 5
AAATGGCAGA






AGAAAGGGTT






AATGGCTGGA






AAAATGGATC






AGTAGTCTTGC






AGAGGAACC







270.

Chromosome 10
ATTTTAGGGGG






CTTTATTGTTA






CTTGACGTGGA






ATTTGAAAACT






AAAAAGATGA






GTCTGG







271.
Mm.276728
Chromosome 11
GTGGAAATCA






GAGATCTAAGT






ACGTTTATGCA






TAGGAGTAGG






AATGAGGGGTT






ATTAAAG







272.
Mm.30052
Chromosome 4
AAACCCCCCAA






GTAGCCCAAA






GGCCCGCTTCC






CACCAAAATGT






TTTTTATGTTTT






AAGGA







273.
Mm.105080
Chromosome 16
ATTATGATGCC






TGTAACACACA






GAAGTATCTGA






CTGTGAACGAA






TCAACCTCATG






GATGA







274.
16169
Chromosome 2
AGAAGAGATA






CTGAGCCAATG






AACCCTTTCGT






ATAGGATTCAT






GACAAAACCA






AACTCAG







275.

No Chromosome location
CTGCCTTCCCA





info available
TAAAAATAAA






AGGCATGCAA






AACCAATTTTT






GGCCAGGCCC






AGTTAAGA







276.
Mm.28088
Chromosome 5
ACAAGCCCTGG






GCCTCTGAGAC






CACCCGACACA






CCATCCTACCA






AGAAGCCTCTA






AGTAT







277.
Mm.29981
Chromosome 13
CAAGTCAGCA






AGAAGCCAAC






CTTGGTGAAAT






AATTCTGGTTG






TTTGAAAGCTA






GGTCTTG







278.
Mm.250067
Chromosome 1
GGTCAAGAGA






GTGCCAACTAG






CTTTGTTTAAA






AAATCCTAGTC






CTGAATCCACA






AGCCTG







279.
Mm.222100
Chromosome 9
AGTGGAAGCCT






TATAAGCATTG






AACCCAGGAT






GAGTCGCTCGT






ATTTCCACCTT






ACTCAT







280.
Mm.259829
Chromosome 15
CTTCCCACAAC






CCCACCGTACC






TTGTCTATGTA






TGCATGTTTTT






GTAAAAAAGA






AAAAAG







281.

No Chromosome location
TGCCTGACTCC





info available
AAGAAAAGAA






GCCAGAACTCG






GAACCATAGTC






ATCTTTAAAGA






TCTTCT







282.
Mm.45194
No Chromosome location
GTTAATATTAT





info available
TAACTGAGCCT






GCCCATACCCC






CCGTGGTCATT






GGTGTTGGGTG






CAGTG







283.
Mm.157778
Chromosome 7
GGAGGACGAC






ATCCTCATGGA






CCTCATCTGAA






CCCAACACCCA






ATAAAGTTCCT






TTTAAC







284.
Mm.295706
Chromosome 15: not
TCTGAACCTCA





placed
ACCCATCACCA






ACCCCGTGTCT






TCAACATTACT






TTCCAAAAAAG






TCTGG







285.
Mm.217064
Chromosome 10
AGGAGCCTGTG






TCCTTATAGAG






TTGGAATTAAC






TTCAGCCCTCT






ATCTCACTTCC






TCTGT







286.
Mm.29587
Chromosome 2
GAAAAAAGAT






GAGATCTCCTC






CATGACAAGA






GCCTGCATACA






ACATTTGAGTA






CCCTTCT







287.
Data not found
No Chromosome location
TTTGATTTTAG





info available
CAGAAACCAC






CACCAAAATTG






TGCCTTAGCTG






TATTTCTGTTT






AGGGGA







288.
Mm.103701
Chromosome 1
AGATACTATGG






TACTGTCATGA






AATGCAGTGG






GACTCTATTCA






AACAACCCTCC






AAAATG







289.
Mm.157781
Chromosome 7
AGAGAACCCA






CACTCCTTTCA






TCAAGACTTGC






AGAGCATCCCA






CAACCAAGAT






GCTATTT







290.
Mm.218764
Chromosome 17
TATGAGCCTGA






CCCACACTCTC






TGTAAGGTGTG






ACTTTATAAAT






AGACTTCTCCG






GGTGT







291.

Chromosome 9
ATACCCCACCA






CAACCTCTCAA






AAGAGGGCTCT






TAACTTGGAAG






GATAAAATAA






ATCAGG







292.
Mm.1994
No Chromosome location
TATCCTCCCAC





info available
AAAGATGAGA






GGAGCCCATCC






AGTGTTACTGT






TAGAAGTCACA






GTGAAA







293.
Mm.221745
Chromosome 3
TATTGTCCAAT






GAAACCCACA






AACTACCCTCT






ATCTGGAGTTG






GAACATTTATC






TGCATT







294.
Mm.250157
Chromosome 9
TAAGGAGACT






GCCCTACAAAA






CTACGATACTA






CTATCACTTTA






AAAATTAGTGT






AAAGGG







295.
Mm.48757
Chromosome 7
TCAAGGCCAA






GTTTCTGCAAG






AAGCAAGGAT






CCTGAAACAGT






ACAACCACCCC






AACATTG







296.
97587
Chromosome X
GATTGCCAGAG






ACTTACACTTA






ATAGAGTCATA






AAGCCCATAG






AGCCTGAGTGA






GAGCCA







297.
Mm.103259
Chromosome X
TTATTCCTGAA






GCCCCCGCTAC






AGATGTTTCCA






CAACCGAAGA






AGCGGTCTCCA






AAGAGC







298.
Mm.9911
Chromosome 7
AGCTCCACATG






AACTCACAGA






AGAACCAGGC






TAAGTACCCAA






GGACCGAGCTC






AAGGACA







299.
Mm.276293
Chromosome 15
ACCATTATTCT






TTTAAAAAACC






CAAAAACCAC






CAGCAAGGGG






GCCTTTGGTTG






GCCTCAA







300.
Mm.218714
No Chromosome location
CTTCATCTTAA





info available
AACTCCAGAAC






AACTCCCTTCC






TAACCTGGAAC






CCAGCAGCTTT






CAGTT







301.
Mm.261348
No Chromosome location
CTGCACGCCCC





info available
AGGAGCCTGG






GTGAAGCATCA






CAGCACTAAGT






CATGTTAAAAG






GAGTCT







302.
Mm.200585
Chromosome 2
CACTGGAGCAC






TGAACATGATG






TACAAGTATCA






CACAGAAAAG






CAGCACTGGAC






TGTACT







303.
Mm.202727
Chromosome 12
ATAAGAACTTA






TAGGAACCCCA






ACTCCCCATGA






AAAATATAAG






ACCTCAAGGCC






TGGGGA







304.
Mm.100527
Chromosome 3
GCCCACCAACT






CTAATTTGTGC






TACTTATATAT






ATTCCTGGGAG






TAGGACTGTCC






TCCTG







305.
Mm.2364
Chromosome 10
CAGTCAGGTCT






TCCAGAACAAT






TACAACCCCGA






GGAGAACCTC






AATGACGTGCT






TCTCCT







306.
Mm.169672
Chromosome 11
CGTAGCTCGCT






GGTAGAAAGC






CTGACCACCAT






GCATACGATCC






TGGGTTTCAAC






AAGGAA







307.
Mm.196532
Chromosome 19
GAGCCTGAGAT






CTACGAGCCCA






ATTTCATCTTC






TTCAAGAGGAT






TTTTGAGGCTT






TCAAG







308.
Mm.41803
Chromosome 11
GAGTCTGTGGG






TATTCGCCTGA






ACAAGCATAA






GCCCAACATCT






ATTTCAAGCCC






AAGAAA







309.
Mm.98232
Chromosome 11
AGCATCAAAC






AAAGCACATA






AACTCGTACAT






AAGCAAGGGA






TGTCCTTATTG






GTCAAACA







310.
Mm.197271
Chromosome 2
GGGAAAAAAT






AGCAAAACCC






CAAACTCCACA






ACCACAAAAA






CCTGTTAATTA






TGGTGGCA







311.
Mm.30058
Chromosome 9
ACACAGAGCC






AGAAAACCCA






GGCCTGAAGA






CATCCCCTAGT






CCTGCTGAGAG






ACCACAGT







312.
Mm.259849
Chromosome 11
CGACCAATCTG






CCTGGGAAAC






AACACCCCACA






GAACGGGGCTT






CAGAAACACG






TGAGTGA







313.
Mm.45054
Chromosome 19
GTTTAGGTGAG






TTTCCATTGTA






TCTTATAACAG






AGAAACCCATT






AGGCAGTAGTT






AGTTC







314.
Mm.268521
Chromosome 10
TCGAAACACCT






ACCAAATACCA






ATAATAAGTCC






AATAACATTAC






AAAGATGGGC






ATTTCC







315.
76964
No Chromosome location
TGCTACCCTCC





info available
AGGACCAACG






ATGGATGCACC






ACGGAGTCCCA






AGAGCTGAAA






AGCAGAA







316.
Mm.23149
Chromosome 14
CGGAGCTCTTC






AGAACCCCAA






CTCTCTCTGGC






TGGCTACCCCC






AGAACTCCTAG






GTTTAT







317.
Mm.362
Chromosome 9
ATAAAGAGAA






TTCCCACCACC






CTGGGCGAAG






GAATTACCAGC






AATAAAACCTA






TTCCTTC







318.
Mm.11484
Chromosome 7: not placed
ACTTTCAAGTC






TGAATCCTATG






AGCCTGAAGTG






AGATCTTATTT






AGAAACAGAA






CCCCAA







319.
Mm.40331
Chromosome 5
GACAAGCCCTT






AGGGAGCCAG






AAAAAGAGCA






GGAAGAAGTT






AAAATGTTTAA






TTTTTTAA







320.
Mm.188475
Chromosome 16
GCCCAAGAGCT






AGAAAACCTA






CTCTATGTGTA






GAGATACTTCC






TATTAAAATAA






TAGTAC







321.
Mm.216782
Chromosome 9
CTCCACTTTTA






AAGTCTGTAGG






AATAGGAGCC






GATTAGACAAC






TCTCGGTCTCA






TGCTCA







322.
Mm.2877
Chromosome 16
TTTCTGGGATC






CCACTGCACCG






CCATTTCTTCC






CAGATTTATGT






GTATAACTTAA






ACTGG







323.
Mm.27723
Chromosome 17
ATACAGTAGAT






GCTGAACACAC






TTGAGTCCATC






ATGAGGGGGT






AATAAGTCTCA






CCAGCA







324.
Mm.39040
Chromosome 2
TCTTATACTTT






CAACAAAGCT






GAACCCTAACA






TTACACTAACC






AGCAGCTCAAC






ACGAGT







325.
Mm.26700
Chromosome 7
CTGAATGTATA






CACACCCACAG






GAGACTGTGGC






TGAGCGTTCAT






CCAAATAAATT






TGAAT







326.
Mm.35046
Chromosome 4
GTTCCTGTTCA






GAGTGCCTGAA






AACCCAAAGT






GTCTGAGAGTC






TGAAGGAATTC






AACTGT







327.
Mm.24781
Chromosome 6
AAACACCCAC






ACTTGAAACTT






CCATGAACCCA






CTCAAATTCAT






TTCTATCCCCC






TTTGGA







328.
Mm.945
Chromosome 7
TCATGGAGATA






TAACTATAGAG






ATAAAGAGCG






ACACCCTGTCT






GAAGCAATCA






GCGTCCG







329.
Mm.31482
Chromosome 4
GGACACTGTGA






ACACTGTGTGG






ACAGAGCCCA






CAACTTCTCCA






TTTGTGTCTGG






CAGCAA







330.
Mm.14302
Chromosome 9
AGGAAAGAAA






GGGGTTAGAAT






CTCTCAGGAGA






TTAAAGTTTCT






GCCTAACAAG






AGGTGTT







331.
Mm.28451
Chromosome 16
CTCAAGACTTT






GCCAACATGTT






CCGTTTCTTAC






ACCCTGAACCC






TGATCGGAACA






TTCAT







332.
Mm.200891
Chromosome 11
TCTGTACATGG






CCGAAAATCA






GAGTCCACCAT






ATTCTTTTGAA






TATCCAGGGTT






CTCTGA







333.
Mm.193835
Chromosome 6
TTCTGGCTCCT






TATTTCAGTTC






TCTTTAAAACC






AGTTCAACACC






AGTGTGTTAAA






AAGAA







334.
Mm.31129
Chromosome 9
GCAGATTTAAC






AACTAGCAACT






CTGTCATCTTT






TTCTAAAAATG






ACCAACTGCTG






ATTAC







335.
Mm.154695
Chromosome 17
CTTAAAAAGG






GAGATACAGTT






TTACTCTGATC






CAGCAAATCTA






GTTAAGACACT






AGAATG







336.
Mm.9043
Chromosome 7
CTTCCTGAACC






ATTACCAGATG






GAAAACCCAA






ATGGCCCGTAC






CCATATACTCT






GAAGTT







337.
Mm.134338
Chromosome 11
GTAACGGAGC






CTGGGGGTTGA






AGGTTATCTTT






ACATATATGTA






CAAACTGTTGT






CAAGAG







338.
Mm.259795
Chromosome 7
TCCCCACCACT






CATGGGGATCT






TCAAGAAGCAT






CACCATTCACT






GAAAGGTCCTA






AAAAA







339.
Mm.24449
Chromosome 10
GCGCAGAGGC






AAACCAACGT






GGAGCCAGAC






ATTGGTGAACC






CAACCTATCCA






CACCTTCA







340.
Mm.259702
Chromosome 6
CTTATTTTAGA






CAGATCCAAA






GTTCTCACAAG






CCCCCTTTCTT






TGCTCTGCCTA






TCATCG







341.
Mm.11161
Chromosome 8
AACCTCTGAAC






CTAATCACTGT






GGATTCCCACC






AACACCATATA






TGAAAATGCA






GGCCGA







342.
Mm.1428
Chromosome 14
TGCGGAAGGA






GGGGATTCAA






ACCAGAAAAC






GGAAGCCCAA






GAACCTGAATA






AATCTAAGA







343.
Mm.275718
Chromosome 2
CCCTAGTCCGT






TTTCTGATCAG






TCAGAACCCAC






AATAACTACTA






GTAGTCCTGTG






GCTTT







344.
Mm.218038
Chromosome 9
GTAGCCACCAA






GCCACAAGTA






ACAAATGATCT






CTGTGAATGCC






ATATGGAAACT






TTTATT







345.
Mm.216977
Chromosome 9
GGCTCCATTTC






TGAACTCTGTG






TTAAGCTAATA






AGATTTTAAAT






AAACGCTGATG






AAAGC







346.
Hs.158323
No Chromosome location
TGCTGGGGGCC





info available
TAGAACCCTGA






GACATAGACC






ATGGATAAATG






GCAACCGGGG






TGGCAAA







347.
Mm.217130
Chromosome 18
AACGCAAAGA






GCAAGAACCA






AACAAAGACA






GGAACAACTC






GCAGAAGAAA






TCCCGCCTGG







348.
Mm.57171
Chromosome 6
TGTTTTCTGAT






GACCAAAGCA






ATGACAAGGA






GCAGAAAGAA






GAACTGAACG






AATTGATCA







349.
Mm.174523
Chromosome 10
CCCACCACTGA






ATATAGACCAT






ACTGTGAGAG






GACCATAATTA






GGTCCTGAATT






TTTAAT







350.
Mm.103389
Chromosome 3
GTATGACTTCC






AACCAGAAAA






AGGCTCTAAAA






GCTGAACACAC






TAACCGGCTGA






AAAACG







351.
Mm.80565
Chromosome 3
CTTCTGGCTCC






CTTACATGAAG






GACTGATTTAA






GAAACCAGAC






CATTCCTTTAC






TTTGAA







352.
Mm.215689
Chromosome X
GCAGGGTGCTT






ACTTTCTCAGA






GCCTGAAGTTA






CTTCCATTGTT






TTGGCACTGAA






TAACA







353.
Mm.200518
Chromosome 6
TTAGCACAAGA






GAAAAGCTGA






GAACGTGGGTT






TTGCCTCCTTC






AGAAATATGTC






TGGCTC







354.
Mm.152289
Chromosome 17
ACACAGCACCC






ACAACTAATCT






TGGGACACCCC






TATCTGGTTGG






AAGAGAGTAA






ACTAAT







355.
Mm.24767
Chromosome 19
CAATGGCCTAT






TCTGTCAGATG






GGTGTCCTTTC






AAGGGTGACA






ACTACAGAAC






ACAAGTA







356.
Mm.260244
Chromosome 12
AAAGTAGGTTC






ACACAGTAAA






GGGATAATACC






ATCTGGAACAA






TGATCAGTGTA






GAGTTA







357.
Mm.249888
Chromosome 4
CACCTGGGTCT






ACAGCTACTCT






GATTCTACAAA






GACAGGGTCA






AGCATCTCTAA






CAAAGT







358.
15182
Chromosome 10
TATTAAACCCA






GGAGATACAA






GGAGTCTGCCA






TTAACCTCTCT






GTAACTCAAGA






GTAGTT







359.

No Chromosome location
TTCCTCCCAAA





info available
ATGGAGTTTCC






TCTTCAAACCA






CAGCTCCCCCA






AGATCTATCCT






GATAT







360.
Mm.21836
Chromosome 5
TATGTCTTGAT






ACTGGACCCAC






ACTACTGGGGC






ACTCCAAAAA






ACCGTTGTGAA






CTACAA







361.
Mm.208743
Chromosome 11
AGTAAAGGGC






ACCGGAAATGT






TAAATCCTTGT






TTAGGATATGA






AAGGAATTAG






GGGATGG







362.
Mm.217288
Chromosome 11
GAATGTCTGAT






ACATGACCCAT






CAGTTAGGAAC






CACTGAACTAG






AGGAGTAGCT






AAACTC







363.
Mm.45371
Chromosome 13
GCTTCTACTGG






CTCTTGTATGC






ATATGTGCACT






TATCCAGACTG






AGGATTTTACA






AAGCA







364.
Mm.113272
Chromosome X
CTGTCTAAGCG






CTGAACCACTT






AGCAGAAATG






ACACCCATATG






AGAGCTTGTGC






CAAATA







365.
Mm.173357
Chromosome 14
AAAGGAGACT






GCATCAGGTAT






TCTGATAGAGA






GCTGAGGAAG






AGATTGAGGTA






TGGGATT







366.
Mm.234023
No Chromosome location
TGACTGGAATC





info available
ACCACCCTTGC






CTGAGTTTGCG






ATCTCACAGTT






GGAACTGAGA






GTTTCC







367.
Mm.5675
Chromosome 12
GGATCAGATG






ATGCACCATTG






CTTTCCATTGC






TACATTTAAAA






TCTTTTACTAG






TCAACC







368.
Mm.11978
Chromosome 1
TTGAGACCTTA






AAGAAATAAC






AAACTCAAGG






AAGATTAGGGT






CCAGTGTTTAA






GTCATGG







369.
Mm.228379
Chromosome 12
GTCTCCTTTGT






GTTATTGCCTT






CCCAACACTTC






TAAGTCCCAGC






TCAACAGCTAC






TTCTA







370.
Mm.17675
Chromosome 1
CACAGCTGCTT






GTAGTCATCAT






TCCAGTGAGGA






GTAAGAAGAA






TTTTATGTGTG






TCTCTA







371.
Mm.20355
Chromosome 4
AACTTAAACAG






TCTCCCACCAC






CTACCCCAAAA






GATACTGGTTG






TATTTTTTGTTT






TGGT







372.
Mm.28721
Chromosome 2
CAGCAGAAA






GGCTCCCACCA






AGAAGGCCAA






CAGCACAACC






ACAGCCAGCA






GGATGTGTT







373.
Mm.25072
Chromosome 17
GGCTTCACATC






TAAGTGGGGA






CTATTTTAACT






TATTTACAGGT






ATATGGTGTGG






AAATAA







374.
Mm.233802
Chromosome 7
CGCTCAGTTGT






AGAAAGCAAC






AAGGACACAA






ACTTGATTGCC






CAAAGTCACTG






CCAGTTA







375.
Mm.1389
Chromosome 7
GTCTGAACACA






CTATTATGTAT






CCATCCAATCT






CAACTGAATAA






AGGGAGATGC






CTTTTG







376.
Mm.221547
Chromosome 14
AAAGAATTTCA






AGAACGAAGC






ATAGGTGGTTA






TGTAGTTTGAT






TACAGAAAAG






AGATGCC







377.
Mm.4697
Chromosome 11
AAACCACCTTC






AGTGTGAGGA






GCCCACGTCAG






TTGTAGTATCT






CTGTTCATACC






AACAAT







378.
Mm.100116
Chromosome 6
GCACTCCAGCC






TGATTCTTTGA






GACTTTGGGGT






ACACATATTGA






AAGTACTTTGA






ATTTG







379.
Mm.231395
Chromosome 7
ACTGTATCGGT






TCCATGTAAGT






CTGACCAGTCA






AAGGCAAGAG






GTATCAAGGTG






GAGAAA







380.

Chromosome 18
GTGTTTGAATT






AAAACCCCCAC






CCTCGGAGGCC






TTTAAAGAAAT






GGTTTTTGTCC






GTTGT







381.
Mm.149642
Chromosome 6
CTCTCGACAAA






ATATAAATGGA






CAGTACCAAAC






TAAGAGGGAT






ATAAGTGGGA






GCAAAGG







382.
Mm.202311
Chromosome 11
TATGGTACGAG






TTTAGGGCTTA






GTCAGTTTACA






ATGGGGATTGA






ATTTTGTGTCA






AAACC







383.
Mm.235234
No Chromosome location
CTGGCTCCTAC





info available
TGGCAACAGG






CATACTTGTGG






TTTAATACAGA






GAAACAAAAC






ATTCATA







384.
Mm.27968
Chromosome 1
TTTGACCTAAT






GAAATACCCAT






TTCATCTGTGA






CAACACATAGC






CCAGTAAACAT






CACTG







385.
Mm.37806
Chromosome 14
CCTGTTCCTAG






TATCCTGGCGT






CCACATATACC






CAAAGTTAGGC






ATACTAACCAA






GAGAT







386.
Mm.2901
Chromosome 2
CTGGAACTCAG






CACTGCCCACC






ACACTTGGTCC






GAAATGCCAG






GTTTGCCCCTC






TTAAGT







387.

Chromosome 12
CCTGGAGGTCT






CCACCTGAAGT






TCCCTGATGCA






GGGTCAGTCCA






GCCTTGGTAAG






GGCCA







388.
Mm.182611
Chromosome 12
AAATGAGAAC






CAGATTACCAA






AATTACCACTA






CCACCAAAATA






ACCCCTCTGAT






TCCTTG







389.
Mm.225096
Chromosome 2
CAGATAGATG






ACAGCAGGAA






ATTTTCTTTATT






TCCTGAAAGAA






AATACCAGACT






CTCAAC







390.
Mm.174047
No Chromosome location
GGTGCCAAATG





info available
CGGCCATGGTG






CTGAACAATTT






ATCGTCAGAGG






GGAAGAACAG






TTGACC







391.
Data not found
No Chromosome location
CCAAAACAGA





info available
GCCAACACCAC






CGACAACAAC






CCCACAGCAA






ACCCGGAGAG






AAACCCAAA







392.
Mm.12900
Chromosome 1
TTTCAACCCGC






CCATTATTTCC






AGATTTATCCG






CATCATTCCTA






AAACATGGAA






CCAGAG







393.
Mm.143742
Chromosome 5
TGGAGACTGA






GTTCGACAATC






CCATCTACGAG






ACTGGCGAAA






CAAGAGAGTA






TGAAGTTT







394.
Mm.250079
Chromosome 11
GATACAACAG






CATCTGTTTTC






CAAGGAGAAA






TCATTTGAGGA






ACAAAACCTAT






CAAGAGA







395.
Mm.45048
Chromosome 2
AACTAGAAAA






CATAGATGCAC






AGGACTCGGAT






CCATGATATTT






ACACTGGGAA






ATGTTCT







396.
Mm.34384
Chromosome 6
ATCTCAAGATT






TCTATCCAAGT






GGAAACAAAC






TGAATCATGCA






CACGACTTATC






TGTGTG







397.
Mm.19839
Chromosome 13
AGAGGAGCCA






CACTTGATGTG






AATTAAACTCA






TAAACATTATG






CCACTAACAGC






TTTTAT







398.
Mm.4312
Chromosome 4
CTGCCGCCTGT






ACAAAGGAAA






CTGAACCTTTT






TCATATTCTAA






TAAATCAATGT






GAGTTT







399.
Mm.206841
Chromosome 3
AAGCTGAGATT






AAACGGCTAC






ACAATACCATC






ATAGATATCAA






CAACCGAAAA






CTCAAGG







400.
Mm.28865
Chromosome 4
GACTTGGGAA






AACAATGCAA






CTCCCATAAAC






CAAAACTCCAA






TTCCATGCCTA






ACTTGCT







401.
Mm.2666
Chromosome 4
AGCAGGGAAC






AATTTGAGTGC






TGACCTATAAC






ACATTCCTAAA






GGATGGGCAG






TCCAGAA







402.
Mm.6251
Chromosome 1
AGCTCCAACTC






AACAGATGGCT






ACACAGGCAG






TGGGAACACTC






CTGGGGAGGA






CCATGAA







403.
Mm.103450
Chromosome 10
ACTAGCTGCAT






TGTAAAGAAA






CAAATCGAAA






CTGAGTCTTTT






CACATATTGTG






ACGGACA







404.
Mm.24276
Chromosome 6
GTAGGGTCATC






ATACACCCAGA






CTACCGCCAAG






ATGAACCTAAC






AATTTTGAAGG






AGACA







405.
Mm.11092
Chromosome 11
TCCCCACCACG






AATTATCGTGG






CTAGTGGATGA






AGGCCACTAAT






ACAGGTTCAAA






TTGTT







406.
Mm.23837
Chromosome 5
TATGTGCATAG






GCTGGAGTTTT






GGTTATACATG






GTACACTTTTG






GGCCAATATAA






TAGGA







407.
Mm.9706
Chromosome 12
CCACACTCCCT






GGAGACAATG






TCTGCCATTTT






TGCATCACTTG






TCAAACCACTA






ACTTCT







408.
Mm.173615
Chromosome 6
TCGGTTGACCT






GATTCCACCAA






GGAGAAGGAG






ATCAAGGAAG






AGTAAACTGTA






AGAGCAT







409.
Mm.133615
Chromosome 9
GAGTGCTTTGA






TGGTTGTTAGG






GACCGTAAGA






ATAGTCCTGTG






TCAGACAGCA






GATTCTA







410.
Mm.255931
Chromosome 19
AACTGTCATAA






AATCCAACGTG






CCTTCATGATC






AAAGTTCGATA






GTCAGTAGTAC






TAGAA







411.
Mm.289605
Chromosome 5
ACTCTCATCTG






TAAAGCCTTCC






CATCTCATTAT






TCCTTGCACTA






ACCACAGCCAC






TAGGA







412.
Mm.197224
Chromosome 11
CAGACTGAAA






GGAAATTCCAA






AGAAAACAAA






AACCTTTCAAT






CTATGAACTCA






ATGGCTG







413.
Mm.219663
Chromosome 15
CTGAGAATAAC






CTACTACCACC






TCTCTTTTCCC






ACCAACATCCA






AGTGCCAGCG






GTGGTT







414.
Mm.134516
Chromosome 14
AGCGACATGC






AACCAAATACC






ACTCAAAACA






AAAATCCAGC






AAAACTGAGTT






GTGAGGGA







415.
Mm.35474
Chromosome 14
GTTTGTACATG






TAAAAGATTGA






CCAGTGAAGCC






ATCCTATTTGT






TTCTGGGGAAC






AATGA







416.
Mm.173781
Chromosome 3
ACTTAGACCAC






AACAGCATCTA






AGCATCATTAC






CTTAAGTACTA






AAGCAAAAAT






CTAGTC







417.
Mm.60590
Chromosome 15
TAAACCACTCT






TAAACTGCTGG






CTCCAGTGTTT






TTAGAATGATA






TGAAGTCATTT






TGGAG







418.
Mm.2408
Chromosome 6
AGTAAGTGCCA






TTATCCACCCA






ACTACCAACCA






ATGCCTAAGCA






GATTCTATATC






TTAGC







419.
Mm.31672
Chromosome 5
GCTTCTGGCAG






AGATCTGTTTA






GCATAGTGTGG






TATTAATTATA






GCAAATGTTAA






GGTAG







420.
Mm.250054
Chromosome 15
GTTGTCTGAAT






AATAGCACCCA






AGAAAAAGTG






TGGAGATCAGT






AGGTATTCATT






AAGCAT







421.
Mm.5202
Chromosome 10
TAAAGGAGCTT






TCCACATGAAC






TCACAATTTTC






TTGAAATAAAC






TTCTTAACCAA






CTGCC







422.
Mm.987
Chromosome 1
GTCACTTGGAT






GGTGTATTTAT






GCACAAAAGG






GCTCAGAGACT






AAAGTTCCTGT






GTGAAC







423.
Mm.159956
Chromosome 7
GTCATGAACCC






AATACACTGTG






GAAATGTGTGA






TTCTTTATATT






AAACGTCTGCT






GTTCA







424.
Mm.31672
Chromosome 5
TGTCGATACCA






TCTAAAGACCA






CAACTTCTAGC






CATAGGGTATT






TCATATATGTC






CATTT







425.
Mm.221754
Chromosome 7
ATGCAAACCTA






AAAAGCACCC






AAAAAATTCAC






ATTGGACTGAA






GAAGAGTGAT






CCAAGCA







426.
Mm.1114
Chromosome X
TTTGAGACCCT






TTCATAAGCCC






AATTATACAGA






TATCCAATATT






ACTGCAATCAT






TGGAG







427.

Chromosome 13
ACCTAAATTTC






CACAGGCAACT






TACTTTGTTAT






TAAATTTGGGG






ATCATATCCTG






TGCCC







428.
Mm.260376
Chromosome 9
TTTTTTCAGAC






TTAAGAACAGC






TAAACAAAAC






CTTCCTCTAGC






TTTTTCATCAC






ATCCAG







429.
Mm.249886
Chromosome 17
ATAATGATGAT






GATAACAACA






AGAAAACAGA






CTCGAACCTAA






AGACGCTGGTC






TCAGATA







430.
Mm.4987
Chromosome 5
CGCAAACATAC






CCTGTATAAGA






AGGCTCCTAAC






GAGAGATTTAT






TAACAACACTA






TATAT







431.
Mm.233117
Chromosome 19
TTTGACTGGGA






CCAGCCCAGCC






ATTCTCAGCCT






CTCGACATGTA






ATTTCATTTCT






TTTAC







432.
Mm.24576
Chromosome 3
AGGACTCATAG






ACTTACAGAAT






GATGCCGAATG






GAATGTTTTGT






GCATGACCTTT






TAACC







433.
Mm.143813
No Chromosome location
CCACCTCGCCC





info available
AAGTCTCCTTT






TACTGAAATAA






AATTTGAGGGG






AAGAGAAAAA






ATTTAC







434.
Mm.15383
Chromosome 15: not
GATGTTCTTCT





placed
GTAAAAGTTAC






TAATATATCTG






TAAGACTATTA






CAGTATTGCTA






TTTAT







435.
Mm.23572
Chromosome 2
CTTAAGATTCA






GGAAAATGGTT






CTTTCTGCCCT






TCCTAGCGTTT






ACAGAACAGA






CTCCGA







436.
Mm.24138
Chromosome 2
TATATTGACAT






CCATAACACCA






AAAACTGTCTT






TTTAGCTAAAA






TCGACCCAAGA






CTGTC







437.
Mm.3645
Chromosome 9
TCTTTAGTGCT






GCATTTAAGTG






GCATACAAAAT






ACAATCCCATA






TGTATGAACTG






TTGTG







438.
Mm.86813
Chromosome 16
AATCTATGCCA






GATACTGTATA






TTCTACCATGG






TGCTAATATCA






GAGCTAAATG






ATACTC







439.
Mm.136022
Chromosome 2
AATTTACACAT






GTGGTAGTAGT






AGGTCCAGATT






CCTAAGTTACA






GTGTGCTGAAA






AATAA







440.
Mm.11819
Chromosome 1
ATGAGGCTAA






ATTTGAAGATG






ATGTCAACTAT






TGGCTAAACAG






AAATCGAAAC






GGCCATG







441.
Mm.86813
Chromosome 16
TCTACTACTTT






GCTTATCATGT






TCACTGCAAGG






GAGGCAACGT






ATGGGTTGCTC






TCTTCA







442.
Mm.196253
Chromosome 16
GTACTGAACTC






ACAAGCGTATC






TCCTATTTTAT






GAGAGAATAC






TGTGATAACAA






AAAGTG







443.
Mm.6483
Chromosome 7
TTGGCCCACCC






CCAAAGGGCC






AAGATTATAAG






TAAATAATTGT






CTGTATAGCCT






GTGCTT







444.
Mm.3453
Chromosome 4
CTGGGAACCAC






CTAATGGTATT






ATTCCTGTGGC






CATTTATCAAT






ACCTTATGAGA






CTATT







445.
Mm.22929
Chromosome 4
TCCTCTGGGGT






AAATGAGCTTG






ACCTTGTGCAA






ATGGAGAGAC






CAAAAGCCTCT






GATTTT







446.
Mm.233891
Chromosome 2
GCCGCAACGC






AACAGAAATT






GTTTTTAATTT






CATGTAAAATA






AGGGATCAATT






TCAACCC







447.
Mm.25941
No Chromosome location
ACTTTTGGGTC





info available
TTTAGAACTGA






GCCCACCTACT






GAGTCTCAGTT






TCTGTTGGTGT






GACCT







448.
Mm.17185
Chromosome 12
TGCTTACTAAG






AAGCCAGTTTG






GGTGGGTAAA






GCTCTCTGGAA






GAAGGAACTTT






GCTTCT







449.
Mm.3266
Chromosome 11
TCCCAATGTGT






AGAATTCAACT






ATGTAACGCAA






TGGTACATTCT






CACTGGATGAG






ATAGA







450.
Mm.930
Chromosome 13
CTTATGGACAC






TATGTCCAAAG






GAATTCAGCTT






AAAACTGACC






AAACCCTTATT






GAGTCA







451.
Mm.29454
Chromosome 12
GCCATATGATG






AACAGAATTTC






AAGAATGCTGT






TTTATGCCTTT






TAACCTCCAAA






GCAGT







452.
Mm.288252
Chromosome 15
TCATTTTCCTG






TCTAGGCTAAA






GCTAAACTTAA






ACTATGGCTTT






ACGTAAATTAA






GCTCC







453.
Mm.24223
Chromosome 16
CAACATCTAAC






GCTTTACATAA






ATGCCCTTTTA






GCTTCTCTATT






TCGACACAACT






GTGAT







454.
Mm.9277
Chromosome 17
TTACCCAAATA






AGCATTTTTTA






AATATACCCTG






TACTGTAGGAT






AGTGATGAAC






GCCTAG







455.
Mm.459
Chromosome 1
ATAAGCCGTAT






CTGGGTCTTGG






ACTACTTTGGT






GGACCTAAAGT






AGTGACACCTG






AAGAA







456.
Mm.4190
Chromosome 8
AAGTGGAATG






GAGCCGGCCA






AGCTGAGCCTG






ACTTTTTTCAA






TAAAACATTGT






GTACTTC







457.
Mm.4159
Chromosome 2
CTTAAAACTAC






TGTTGTGTCTA






AAAAGTCGGT






GTTGTACATAG






CATAAAAATCC






TTTGCC







458.
Mm.19352
Chromosome 14
CAGCTGCCTAA






CCCGCAACATT






TGCATTATGTT






CAGACTGTAAC






CTGCTTACTGA






TGGTA







459.
Mm.233010
Chromosome 10
CTGTGGTACCA






AGGAGTTATTT






TGGATGATTAG






AAGCACAGAA






TGATCAGGCCT






TTAGAG







460.
Mm.2580
Chromosome Multiple
TTGTTTTTGTTT





Mappings
TTAACCTAGAA






GAACCAAATCT






GGACGCCAAA






ACGTAGGCTTA






GTTTG







461.
Mm.42193
Chromosome 13
TGCCTGAAAAC






ACTTAACACTG






ATTGTCTAAGA






GATGAAAGTCC






TCCAAAGATGA






CACAG







462.
Mm.134712
Chromosome 10
ACTTCAGTTAA






TGGGTTTATAA






AGTCAAGCACT






GGCATTGGTCA






GTTTTGTATGA






TAGGA







463.
Mm.34883
Chromosome 9
TCCCCTATGCG






GTACGACCTTT






ACTGTCAGAAA






TATATTTAAGA






AAATGTTCTAA






ACGGT







464.
Mm.9953
Chromosome 9
GATCCAGCCTT






CTATGAAGAAT






GCAAACTGGA






GTATCTCAAGG






AAAGGGAAGA






ATTCAGA







465.
Mm.741
Chromosome Multiple
CATGACTGTTG





Mappings
AGTTCTCTTTA






TCACAAACACT






TTACATGGACC






TTCATGTCAAA






CTTGG







466.
Mm.19844
Chromosome 5
CTTGTAATCAG






ACACGTGTTTT






CCTAAAATAAA






GGGTATAGAC






AAAATTTAAGC






CCATGG







467.
Mm.3468
Chromosome 11
TGTCTGAAGAT






GCTTGAAAAAC






TCAACCAAATC






CCAGTTCAACT






CAGACTTTGCA






CATAT







468.
Mm.369
Chromosome 4
TACTCCCATTA






CTATTTGCTGG






TAATAGTGTAA






CGCCACAGTAA






TACTGTTCTGA






TTCAA







469.
Mm.22753
Chromosome 14
CAGCCGATGCT






TTTTCAATAGG






ATTTTTATGCT






TTGTGTACCTC






AACCAAGTATG






AAGAG







470.
Mm.2012
Chromosome 6
GGGACACTTAA






TTTACATGTAC






TTTAACCCCAT






GAAAGAGTCT






AGATAGAGAG






AAGACAC







471.
Mm.22547
Chromosome 8
GCCTGCCAGTA






ACCCCAGGAA






GAGTCTAGCTT






CAAAAACCCA






CAAACTCATTA






TTTTTAA







472.
Mm.39298
Chromosome 3
AATCTAGATGT






TAGAAATCAAT






GTGTATGATGT






ATTGTATTTAG






ACCATACCCGT






GACCG







473.
Mm.57177
Chromosome 2
ACGATGAGCA






GTGTTTGAAAG






CTTTCCAGTGA






GAACTATAATC






CGGAAAAATG






AATGTTT







474.
Mm.41333
Chromosome 10
GATGCGTGAA






ATGTTCCTCCA






GGAAAAGCCA






TTCAAGCCTGA






TTATTTTTCTA






AGTAACT







475.
Mm.100666
Chromosome 1
CATCTTAGATC






TCAGAGACTTG






AACCTTGAAGC






TGTTCCTAGTA






CCCAGATGTGG






ATGGA







476.
Mm.2423
Chromosome 15
CGTGTCCTACA






CAATGGTGCTA






TTCTGTGTCAA






ACACCTCTGTA






TTTTTTAAAAC






ATCAA







477.
Mm.15185
Chromosome 8
AAGGAGCCAC






GATAATACTTG






ACCTCTGTGAC






CAACTATTGGA






TTGAGAAACTG






ACAAGC







478.
Mm.20458
No Chromosome location
GTTTATAGGTA





info available
GACCTAAGAG






ATAAAACTGCA






GGGTATCACAT






TAACGTTGGTT






AAAAGA







479.
Mm.26786
Chromosome 15
AAACTTGAGAC






ATTTTGTAGGA






CGCCTGACAAA






GCGTAGCCTTT






TTCTTGTGTCA






GGATG







480.
Mm.565
Chromosome 12
CTCATACCAAA






GAAATACTTGA






CACTGCTTTGA






AGGAGATAGA






TGAAGTTGGGG






ATCTGC







481.
Mm.290404
Chromosome 12
AAATCCAGCCT






TTAAAAGCTCA






GTTTCTTCCTC






TAAGTGAATGT






CATTACTCTGG






TATAC







482.
Mm.5378
Chromosome 6
ACCAGGAACTC






TGGTAACATTT






GAGGGCATGC






AGATAAAATA






ATAAAGAATG






AGAACATT







483.
Mm.29564
Chromosome 8
TCAACATCTAT






GACCTTTTTAT






GGTTTCAGCAC






TCTCAGAGTTA






ATAGAGACTG






GCTTAG







484.
Mm.261624
Chromosome 13
GACCGAGAGC






CACCACAAGG






CCAAGGGAAA






ATAAGACCAG






CCGTTCACTCA






CCCGAAAAG







485.
Mm.3152
Chromosome 11
TTCTACCTCAC






TAACTCCACTG






ACATGGTGTAA






ATGGTACATCT






CAGTGGTGGTG






ATGCA







486.
Mm.18742
Chromosome 7
TTGGAGAAATT






AGGAGTTGTAA






GCAGGACCTA






GGCCTGCTTGA






TTCTTTCCCAC






CTAAGT







487.
Mm.3137
Chromosome 1
TTATTGAAAAG






TTTGAAGTTAG






AACTTAGGCTG






TTGGAATTTAC






GCATAAAGCA






GACTGC







488.
Mm.227260
Chromosome 2
CACCATTTCCA






ACTTGCTGTCT






CACTAATGGGT






CTGCATTAGTT






GCAACAATAA






ATGTTT







489.
Mm.3295
Chromosome 1
AACAAGAGAT






CCTGTGGATGA






GGGGGTCTGTA






TAAGTTATACT






CCAATAAAGCT






TTACCT







490.
Mm.38783
No Chromosome location
TTTTGACCAGT





info available
TGAACCCATTT






TGTTTTCCTAG






CGAACACTAGC






ATAATATTGGA






AAAGC







491.
Mm.257899
Chromosome 1
GTGAGGATTGG






AATTAGAACAT






TCATAAGAAA






ATATGACCCAA






CATTTCTTAGC






ATGACC







492.
Mm.7500
Chromosome 7
CGCCCTGGAGC






CTCTGTCAAGT






CTTGGACCAAG






TAAAAATAAA






GCTTTTTGAGA






CAGCAA







493.
Mm.142455
Chromosome 14
AAGATGGAGA






GTTGTCCAAAC






AAGATCCCAA






GTCTAAATAGA






GCAAGGGATTC






TGAGGTG







494.
Mm.200886
Chromosome 2
GTTTTAAAAGG






TGCCAGGGGTA






CATTTTTGCAC






TGAAACCTAAA






GATGTTTTAAA






AACAC







495.
Mm.25311
Chromosome 15
TCTGAGGTATT






AAAATATCTAG






ACTGAATTTTG






CCAAATGTAAG






AGGGAGAAAG






TTCCTG







496.
Mm.282049
No Chromosome location
AAGTATTGCTA





info available
GACTGAAACC






ACTTGAACTTC






TCAGAGAGGTT






AGACTGACAG






AAGGTGT







497.
Mm.41264
Chromosome X
ACATTTTTGTC






ATCATCATGTA






AATCCCACGAT






TTCAAACTGTA






AACATCTGTTC






AGTGG







498.
Mm.24584
Chromosome 11
CTGGGGAAATT






GATCTTTAAAT






TTTGAAACAGT






ATAAGGAAAA






TCTGGTTGGTG






TCTCAC







499.
Mm.45173
Chromosome 3
AGGACTCAAA






ACTATATTAAT






CTGCTCTGAGA






TAATGTTCCAA






AAGCTCCAAA






GAAAGCC







500.
Mm.151315
Chromosome 1
GCTCCAACATG






CCATGTATTGT






ATAGACTTTTA






CTACAATTCAA






ATAACGTGTAC






AGCTT







501.
Mm.25492
Chromosome 8
CAGCTGAATGG






GTTTTGGTTTG






CAGGAAAACA






GTCCAGAGCTT






TGAAAAGGCTC






CTAAGA







502.
Mm.227732
Chromosome 3
TGTTTTTATTG






TGTTTGGTGGA






GAAGAATAAT






ACACTTCTTGC






CTAAATCCAGA






AGCCCC







503.
Mm.27061
Chromosome 6
TCCAGTTCCCG






AAGAAGCTGA






TAGGAATTGCC






CTTGTGCATAT






ACTACACAAGC






ATGCTA







504.
Mm.4165
Chromosome 19
CATAAAGACAT






AGTGGAGGTTC






TGTTTACTCAG






CCGAATGTGGA






GCTGAACCAGC






AGAAT







505.
Mm.27098
Chromosome 5
GGATTCGGCTC






GATGAATGAA






GCACTTTATGG






ACTGCGGGGAT






CAGTTACTGCC






ACACCC







506.
Mm.7046
Chromosome 2
TGCTTTTACCA






TGTTCTCGAGG






TTCCTGAACAA






AGAGCCTTACT






GATAGTTCCGC






TGCAA







507.
Mm.29329
Chromosome 4
TGAAGCAAAA






AACATAAAAC






CTCACCACTGC






CTGCTGAACCT






AGAACCTTTTG






TTGGGGC







508.
Mm.381
Chromosome 4
GAATCCTTAGA






TGAAGTTATGG






ATTACTTTGTT






AACAACACGC






CTCTCAACTGG






CTGGTA







509.
Mm.38399
Chromosome 1
GATATTAGTAG






TATATCATAAA






ACTTGAGAAAT






AAAGATGCGCT






CACCCCCTATC






TGTTG







510.
Mm.39298
Chromosome 3
TGTGATAAAGT






TGTGACATACG






TATTAGTTGGC






ACATATTTAAG






CTCCAAATCAG






TTTGC







511.
Mm.260244
Chromosome 12
TAAAAGTTAAA






GTAAGCGAAG






AAAGGAAGCT






GTATCTACACT






GCTTTCCAGTT






TAATCAG







512.
Mm.193099
Chromosome 1
GGAGATTTTTC






TCTTCAGGGTG






TCTACATACCT






TACACACACTT






GTGTCTTAATA






AGCAA







513.
Mm.156919
Chromosome 2
AATCCATGGGA






GGGGGGAACA






AGTCCAGACTG






CTTAAGAAATG






AGTAAAATATC






TGGCTT







514.
Mm.1568
Chromosome 11
AATGTGGAGTG






TGGAGAAGGG






CATTTCTGCCA






TGATAACCAGA






CCTGTTGTAAA






GACAGT







515.
Mm.14860
Chromosome 19
TGACATGAATG






AAATCAAAGT






ATTTTACCAGA






AGAAGTATGG






AATCTCTCTTT






GCCAAGC







516.
Mm.27816
Chromosome 13
ACTGGATACTG






TAACTATGAGA






ATAAAATATAG






AAGTGACAGA






CGTCTACAGCA






TTCCAG







517.
Mm.275696
Chromosome 10
ATACAAGCAA






GCTGTTAAAGA






TCTTGGATCCC






ATTCTATAGTG






TGTATACCTAA






ATCAAC







518.
Mm.245007
Chromosome 1: not placed
AGCATCAACTG






TCCTGTCAAGC






ACAAAAAATG






AAGAAGAAAA






TAATTACCCAA






AAGATGG







519.
Mm.27112
Chromosome 12
CCTCTGTTCTG






AGGAACATTCT






AGCATAGAAA






ATGGAATATGC






TGCAAACATTT






CTAGAT







520.
Mm.212870
Chromosome 1
GTGTAGAAGCC






TATTGAAATAT






CAGTCCTATAA






AGACCATCTCT






TAATTCTAGGA






AATGG







521.
Mm.19182
Chromosome 7
CTGATCCCGCC






TCATCTCGCTG






CTCCGTGCTGC






CCTAGCATCCA






AAGTCAAAGTT






GGTTT







522.
Mm.10727
Chromosome 8
TGTAGAAAATG






TGGCCTCTCGT






TATAAATGAAA






ATAAATGTTTA






ATTTAATGGGA






GTTTC







523.
Mm.6105
Chromosome 2
GGTGCCACAG






AGAAGAGCCC






AGTTGGAAGCT






ATACCCGATTT






AATTCCAGAAT






TAGTCAA







524.
Mm.193099
Chromosome 1
CAGTGTTGTTT






AAGAGAATCA






AAAGTTCTTAT






GGTTTGGTCTG






GGATCAATAG






GGAAACA







525.
Mm.200518
Chromosome 6
ATAACTATATA






TACTTAGAGTC






TGTCATACACT






TTGCCACTTGA






ATTGGTCTTGC






CAGCA







526.
Mm.6419
Chromosome 5
CCTTGGGACAT






TTTTGTGGAGT






AGTTTGCAGTG






AGATAACAGT






GCAATAAAGA






TACAGCA







527.
Mm.46067
Chromosome 14
TCTATACCTGG






ATAAAAAGAA






ACCTACACTTC






ACTGTAAAACT






TCATGTTTCAA






GGCAAG







528.
Mm.192991
Chromosome 8
CCTGTTTACTA






AACCCCCGTTT






TCTACCGAGTA






CGTGAATAATA






AAAGCCTGTTT






GAGTC







529.
Mm.33903
Chromosome 13
ACCGTGTAGAC






ACTCATATTTT






GCATGACATGA






TCTACCATTCG






GTGTAAACATT






TGTGT







530.
Mm.2436
Chromosome 6
GCCAAAGGAA






AATGTTTCAGA






TGTCTATTTGT






ATAATTACTTG






ATCTACCCAGT






GAGGAA







531.
Mm.28392
Chromosome 7
TCCAGAAGCTG






CATTGCCAACA






TCACACCCCAA






AATTGTCCTGA






CATCGCTGCCC






GCATT







532.
Mm.2814
Chromosome X
AAGGACTCTGA






GGCCATCCGTA






GTCAGTATGCT






CATTACTTTGA






CCTCTCTTTGG






TGAAT







533.
Mm.296913
Chromosome 13
ATCTCCCAAGG






CAAAGAACTG






AAACTCAGAG






CTGTCTGGATT






GAAGAAATGT






GTGTTGTT







534.
Mm.1186
Chromosome 3
ATGAAGGTAG






GATAATTAATT






ACAAGTCCACA






TCATGAGACAA






ACTGAAGTAAC






TTAGGC







535.
Mm.296074
Chromosome 1
GGTGTAGCCAT






ACAATACACA






AATACAATAG






ATATTCTCTCT






ACAATCTTTAT






GGTGTGG







536.
Mm.29045
Chromosome 6
GGAGAAGCAG






ATTATCTGTGT






GGCTTCCTCTT






TCTGTTCTAAT






ACTGGTAATCA






GTGGAC







537.
Mm.1314
Chromosome 6
GTGAACACCA






GAATTTAATTT






CCATACTTGTA






CAGGTAGGACT






ATTCTTCAGCT






CTCTAC







538.
Mm.46354
Chromosome 9
GGCTTCACACA






TGTGGAGATAA






GCCCCAAAGA






AATGACCATCA






TATATGTGGAA






GCCTCT







539.
Mm.144089
Chromosome 15
GTTTGTAAAGT






TGGTGATTATA






TTTTTTGGGGG






CTTTCTTTTTAT






TTTTTAAATGT






AAAG







540.
Mm.77396
Chromosome 17
ATGGAATTCTG






TTAGAGTAAAA






AAGAGAAAAG






CAGATACTATT






GGCTGGCCTTG






GAGGTC







541.
Mm.87452
Chromosome 1
AATAGTGCTGA






ATTTGTCTAAA






CAGAATTGAG






AGGTCATAGA






AATCCTTAACA






GGGTAAC







542.
Mm.297991
Chromosome 13
TATGAAGATTT






GGGAAAGAAC






AGCTATCTGAC






ACCTGGAAGG






CTCAGCCAGAG






TAACAGT







543.
Mm.22240
Chromosome 4
GAGGCAACATT






CCTTATTCACC






AACTAGTCTCA






AAAGATTGTCT






TAAGCCCTGAC






GATGG







544.
Mm.248549
Chromosome 9
TAATGAAGGAT






GTATAATTGAT






GCCAAATAAG






CTTGTTCTTTA






GTCACGATGAC






GTCTTG







545.
Mm.46067
Chromosome 14
CAGTTTGCGAA






GTAGAATTTTG






TTTCTAAAAGT






AAAAGCTAAG






TTGAAGTCCTC






ACAGAG







546.
Mm.259614
Chromosome 11
TAGAAAAGAT






CACCAACAGCC






GGCCTCCCTGT






GTCATCCTGTG






ACTAAGAAAT






GATTCTT







547.
Mm.4182
Chromosome 7
TATCTAAGAGC






CAAGTCTATGG






CATTAGCTGTG






AGAAGTAGTTA






CCACTGTAATT






CACCT







548.
Mm.36389
Chromosome 12
AAATTATCACT






TGGATACGGA






GGAACATGACT






AGGCACATTTT






ATGAATACTCC






AAATCC







549.
Mm.11982
Chromosome 10
AACTATTGGTG






GTATATTTTTG






AACACAGGTTA






ACTGTGGAGGT






TATCTGCTAAT






AGCAA







550.
Mm.29058
Chromosome 18
ACCTCTGGAAC






AGGCATTGGA






GGACTGCCATG






GTCACACAAA






GAAACAGAAC






TTTTACAT







551.
Mm.132946
Chromosome 1
CCAGTATACCT






ACAAAATGAC






CCACAAGTAAC






CCGCATGAGTC






CAAGTTGTCAG






CCATAT







552.
Mm.30024
Chromosome 6
GTAAAGGGAC






CATTACTAAGT






GTATTTCTCTA






GCATATTATGT






TTAAGGGACTG






TTCAAG







553.
Mm.24887
Chromosome 1
CTCTAAGTCAT






TCATTTTGTAA






AATTATTATAG






AGAAATCTCTA






CTTATACAGAT






GCAAT







554.
Mm.2668
Chromosome 2
TCTAATCTCAG






GGCCTTAACCT






GTTCAGGAGA






AGTAGAGGAA






ATGCCAAATAC






TCTTCTT







555.
Mm.29181
Chromosome 6
ATTCAGATCAG






GAAAGGTTGA






AATGGTCTTCG






TTACCAGGAGG






TCTACATTTAT






TAATTT







556.
Mm.28908
Chromosome 8
CAGTTATGGGC






TTCCATTTTCA






AATATCTTTTC






AACTGTAATGA






CTATGACAGGA






ACTGA







557.
Mm.4509
Chromosome 17
GCTTTCTATGC






ACGTATTGTAC






AAATTGTGCTT






TGTGCCACAGG






TCATGATCGTG






GATGA







558.
Mm.2777
Chromosome Multiple
TGGCTAGATTT





Mappings
AATTGAGGATA






AGGTTTCTGCA






AACCAGAATTG






AAAAGCCACA






GTGTCG







559.
Mm.29656
Chromosome 5
AGAGGACCATT






ATGAAGAAGC






TGTTCTCTTTC






CGGTCAGGGA






AGCATACCTAG






ACTGAAA







560.
Mm.12616
Chromosome 14
AGAAAAGAAA






AAAGCAGAGA






AAAAGTTCATT






GACATAGCAG






CTGCTAAAGAA






GTCCTCTC







561.
Mm.2144
Chromosome 12
ATATTTGCTTA






TTTAAGCGTAC






GTTCCTTTGGT






TTATAGAGAAC






ACCCCCAAATC






ACCTG







562.
Mm.222258
Chromosome 9
GACTCTCCAAC






TTACAGACTTT






TATCAGATATG






GAGAAGATAA






TGTTAAGAGAC






TTCACA







563.
Mm.143846
Chromosome 1
TAAAATCCCAT






TGAAAGTGGA






CTCAGTTGTAA






GAATAACAAT






GTGTACCATTC






TGGAATG







564.
Mm.4182
Chromosome 7
CCAATGAACCG






ACAGTGTCAAA






ACTTAACTGTG






TCCAATACCAA






AATGCTTCAGT






ATTTG







565.
Mm.182649
Chromosome 11
TCAAATCAGTT






TCAACTTTCAT






AAAATGGATTC






TTTAATGGATG






GAGACTTACTC






GTCGG







566.
Mm.160141
Chromosome 2
CTATACACAAG






ATATGCTAGGA






GATGTGAAAG






ATAATGGAGA






CTTTCCAGTAA






GCACTTT







567.
Mm.34609
Chromosome 5
CTGAGATTTTT






CAAATCTTTGG






CAACTGAGATG






GGATGGATCCA






TTTAATTAGAG






AACGG







568.
Mm.2632
Chromosome 3
AAATGTCTTTC






CAACAGTAATG






GTACTATGTCT






ATCCCCTAATA






AAACTTCACTT






CAGCC







569.
Mm.9652
Chromosome X
TGAACATTCAC






AGGATTTCTAA






CTATACTGATA






TAAACCCAGTG






TTTTCTGGACT






CAGGG







570.
Mm.117294
Chromosome 10
CAACAAAGTTG






ATTTACATGTA






TAATCCACACC






CTTAAAGATGA






ACAGTTAGAGT






AGCAC







571.
Mm.142714
Chromosome 15
TGGACACAGTT






CACTAAATTCC






TGATTTAGTCA






AAGTAACTAG






ACTGAAAGAA






CCTAAAC







572.
Mm.17484
Chromosome 6
TTGTTGTGGCT






TCACACTTAAA






TTGTTAGAAGA






AACTTAAAACA






CCTAAGTGACT






ACCAC







573.
Mm.28252
Chromosome 18
TGAACACATCA






AGTATTCTGGA






GCTTCAGCGGC






AGTTAAATGCC






AGTGACGAAC






ATGGAA







574.
Mm.88747
Chromosome 5
AAGGTCCAAA






ATACAGACATT






TTTGCTAGGGC






CTAGAAATCGA






CCATAAAACAC






ACTGCA







575.

No Chromosome location
GACTGAAATG





info available
AAAGTTCCACT






AACGGTATTTG






CTCTAGTGATA






TGTGGACATTG






TGATAT







576.
Mm.106185
Chromosome X
TCAAATAAAA






AACCCTTAATC






AGGCTGTAAAT






CAAATGACACT






ATGCGATGTCA






CTACAG







577.
Mm.221935
Chromosome 1
GCACTATAAAT






TTCATCTTTTG






AAGGTTGTTGA






CTACAAGGGTA






CAAAAATGAT






ACAGGC







578.
Mm.4973
Chromosome 11
CTTGCATGAGT






GCGTGTTTAAG






TTCTCGGAATT






TCCTGAGAGGA






TGGAGTGCCAT






TGTTA







579.
Mm.265620
Chromosome 2
AGTGTTAGCTG






CAAAGCTACA






AAGCTCTGGAA






TGGTTACATTA






TGATTCTGGAA






CGTTCG







580.
Mm.30241
Chromosome 8
TCCAGACTTCT






CAGAGACAAG






GATCTTGCCTT






ATTTTCAAATG






GTGCTAAATTT






AAATTC







581.
Mm.9772
Chromosome 14
AGTGACTTCCA






CCTTTTAATGT






CATTAAAAGCA






GGAGCTTAAAC






TAAAAGCAGC






ATTCCA







582.
Mm.2241
Chromosome 6
ACATACATTTC






ATCACCAATAT






GTTTTATCTTA






CCCCATCTCTC






AGAGTGTTCCC






TGCAA







583.
Mm.21657
Chromosome 4
TTTTTTGTATT






ATTGTGTTTTG






TGCTACTGTAG






TTTTGGTGTGG






CACTATTATAA






TTAAA







584.
Mm.40298
Chromosome 15
CTTAGGGAGAC






TACTAACATGG






AGAGAATGCC






GTGTATACCTC






ACGTACTGTGT






GCTTTA







585.
Mm.3845
Chromosome 18
CATACATAGAA






GCAAAATACTT






TAACTGCTGTA






AACCTTCAAAA






GTTAGTAGACG






TGAGG







586.
Mm.45759
Chromosome 10
ACTTCCTGCAA






TACATCCCAGT






AGGTACACCTA






GTTTACAATTT






AAACTAGTTTG






TGAAA







587.
Mm.34557
Chromosome 10
GGAGGCACAT






AATTCCAAGCA






ATACAGGCTGT






TAAAATATAAA






TAATGGGAACT






GTGATT







588.
Mm.221706
Chromosome 1
AAGCGTTAGG






AAGGAAATTTC






CTGGAAGGAT






AGGTTGTCTTC






CTAGCAGCCTC






GTCAATA







589.
Mm.24807
Chromosome 3
TTTTTTAACTT






CACTCATGACA






ACAGAGGAAG






AAAGGAATTG






AGGTTTAGGTA






AGTTCTC







590.
Mm.24045
Chromosome 12
AGGCATATCTC






ATAGAGCCTTA






AGTTAGAATCT






TACTCTTATGG






AAGGAGTTATT






TCCTA







591.
Mm.34027
Chromosome 1
GATCACCTCAT






TCCTCGACTGT






GAGATGAGTTT






ATGAAAAGAA






TTAAAAGTGAG






CACTTG







592.
Mm.6404
Chromosome 5
TAAAGGTAACT






CCATCAAGATG






AGAAGCCTTCC






GAGACTTTGTA






ATTAAATGAAC






CAAAA







593.
Mm.8155
Chromosome 17
GGCCAGGTATA






TGTGTACCAGT






GCTCTTCAAAG






GGAGAACCATT






AAAACCAACA






TGGAAT







594.
Mm.2793
Chromosome 9
CCAAGAGATTA






TTTAACATTTT






ATTTAATTAAG






GGGTAGGAAA






ATGAATGGGCT






GGTCCC







595.
Mm.118
Chromosome 8
AGTGAACGAA






AAAGACACCTT






AACATGTTTCA






TCTACTCAGTG






AGGAACGACA






AGAACAA







596.
Mm.8040
Chromosome 12
GATATTTATTG






AGTGTCAAATA






AAAAGGTGCC






ATAATCTTCAG






TAGCGTACACA






GTAGAG







597.
Mm.200608
Chromosome 14
GTGTTACCAGA






AGAAGTCTCTA






AGGATAACCCT






AAGTTTATGGA






CACAGTGGCG






GAGAAG







598.
Mm.29101
Chromosome 6
TGTCTTTATTTT






AATGCCAAAA






GGAAGTGATTA






TGCAGCTGTGT






GTAGAGTTTCA






GAGCA







599.
Mm.201248
Chromosome 1
AGAACAAACT






GGAATTTTATT






CTGAAGCTTGC






TTTAAAGACAC






TGATGTGCCTA






AACGCT







600.
Mm.20156
Chromosome 2
TATGGTCTTTC






CAAGGAAACT






AGTCACAGTGT






CATCTTAATCT






TACTGATCCAA






TAAAAC







601.
Mm.248334
Chromosome 15
ATCCTCCTGAT






TGGTCTGAATG






CATTTCCAATG






ATGTCAGGGA






GTCTGCCTTCC






TCAGCC







602.
Mm.259704
Chromosome 13
TAAGCCCTGTC






TTCTGGGAAAT






ATCAGTTTTAA






AGAGAACTTTT






GTGCAATTCCA






AATGA







603.
Mm.29771
No Chromosome location
GGAAGATTAAT





info available
TTTCCAGGGAT






TGTATCAATCA






GGACCATTTTT






GTGGGGCACTT






GGGAC







604.
Mm.21440
Chromosome 2
ATGTGATCTAC






AGTGGTGTGAC






AACTTGCCTTG






TATCTGATGGA






CTGTCCAGATT






TATGG







605.
Mm.29975
Chromosome 2
AAACGAAGTG






ACTTTCCATGA






ATGCCTTTAAC






ATTCTTGTGTC






AACATTTGGTA






CTAAAC







606.
Mm.17580
Chromosome 13
AATACTCATTA






TGCTGTGTGGG






AATTTCCTGAT






TACTAGAAGCT






GACCTCTGCTA






TCCTG







607.
Mm.2018
Chromosome 8
GAATTATTATA






AACAATAATGT






GTTACAGAAGC






TGATGCTGACC






TTGTGTTACTG






AGCAC







608.
Mm.14627
Chromosome 9
TTCTTGAGGTT






TAAGGACGAC






AACTTTATGGA






CCCTGAATGGA






AACTGAGGAA






TCACAAG







609.
Mm.86611
Chromosome 5
GTCACATGCCA






ATAAAAACAG






GAAACTCTGAA






AATAATATGAA






TGTACAGTATC






AGACCG







610.
Mm.252080
No Chromosome location
CCCTATTGCAA





info available
ATTGATTTGTT






TTCCCTTAACC






CTGTTCCCTTT






TAACCCCGGCT






TTTTT







611.
Mm.25264
Chromosome 10
CATTGCATCGT






TTTCCAACATA






CTTTTAGATTT






ACAAAGTAAA






ACCAACCATGG






ATCTGC







612.
Mm.173217
Chromosome 17
TTGAGAAATTA






AAAACAAATA






TCCAAAATCGA






CTTTTCCTCAA






GGCTATGTGCT






TCGTCC







613.
Mm.105182
Chromosome 5
ACGACTCTTGT






TAATGTGCGTT






TCTCATGGAGT






AATTTTCAGAG






CCTGAACTTGT






AGCAC







614.
Mm.3596
Chromosome 2
GTTGGTGTGTC






CTGAAAGGGA






TGGAGTTATGG






CAGAAGTGCTT






TTGTGATCAAC






TGGTTT







615.
Mm.143818
Chromosome 2
CAGAAAACTC






AAGTCATGGAC






TATGCGAGTCA






AGAATTAAAAT






ACAACTGTATT






ATGTGC







616.
Mm.4587
Chromosome Multiple
AAATTTCTCAT





Mappings
TTAATTTTCCA






GTCTCGATTGC






AGTAACAAAG






TCAACCACACA






GTCAGA







617.
Mm.5236
Chromosome 2
GGAGGAAGAC






AACTGAACATT






TGTATAAAACG






TAAAAAGTTTA






CTGATTGGGGT






GGGACA







618.
Mm.31402
Chromosome 16
CAGCAGCTTAC






AAACACTGAA






GTTAGGCGACT






AGAGAAAAAC






GTTAAAGAGGT






ATTAGAA







619.
Mm.68134
Chromosome 14
GAGAAATGTTA






GTAAAATGGTA






AAAGGGAATC






ACGTGACATTC






AGGGTAGGAA






GAGCTTG







620.
Mm.180776
Chromosome 3
TCAGGAAAAA






TGTCATAAGCC






ATCTGGTAAGT






TTTCTTAAAGG






ATGTTGTTAAG






AAGTCC







621.
Data not found
No Chromosome location
CAAAACAAAT





info available
ACATATTATAA






AATAAAAGAA






AAGGCGTGAT






AAATGGATGTG






ACAAAATT







622.
Mm.265969
Chromosome 15
GTAGGGAAAA






TATGTCCATAG






GTTTTAGGAAA






CACTTAGCCTT






TAATATACTGG






TTGTAG







623.
Mm.26430
Chromosome 4
GTATACAGATG






GTAGTTAGAAA






TACTGGATGAA






CTGATCAGTTA






TTGTGTGTAGA






AAGTG







624.
Mm.171547
Chromosome 4
TTGTATCCCAA






AGGGAAACGG






GAATCAAGAT






ACGGACCTATG






CTTTTCATATG






AAACCGT







625.
Mm.4639
Chromosome 16
TGCAGCTAAGG






TACATTTGTAG






AAAAGACATTT






CCGACAGACTT






TTGTAGATAAG






AGGAA







626.
Mm.31530
Chromosome 6
GGCAATGGAA






AATGTTGAAAT






CCATTCAGTTT






CCATGTTAGCT






AAATTACTGTA






AGATCC







627.
Mm.28867
Chromosome 1
CCCCAAAGAA






AACTGGAAAA






ATTGTTTTCCA






CTCCTGAAATT






TCTTGGATGGG






CCCCCTG







628.
Mm.181004
Chromosome 5
CCAGACAGTGT






ATTCTTCGGAC






AAATGGTGTGA






AAGTGAAATA






AGAATTCATAA






TGTAAC







629.
Mm.179011
Chromosome 2
AGCAAAAGTA






TGTATATTTTA






GCTTGTCATGA






AATGTCAACGA






AGGACACTGA






GAAAGAG







630.
Mm.212874
Chromosome 6
TAGAATGGGA






ATTTTCTGTCT






CATAGTGACAT






ATTGCTATGTT






TAACAGTGAAC






ACTCAC







631.
Mm.254904
Chromosome X
TGACGGTATAT






TTGCAAAAAG






AGAAAGAAAA






ATCTGGTATTT






GCAATGATCTG






TGCCTTC







632.
Mm.86150
Chromosome 16
GAAATATCATT






TGTAGCTTTAA






GGCTAGAAAA






TGAAAAAGAA






TCCAAGCCAGT






AGAAGGC







633.
Mm.275985
Chromosome 8
ATACCAGGAA






AATAAAAGTA






CCAGTAAGGA






AGCATCAAATC






AAGATGTCATA






GTCAGTGG







634.
Mm.17827
Chromosome 3
CAGTGTAAATA






TAGCATATGGT






TAGGTGGTGAG






AAAATGATCTT






GAGACTGATA






AGAATC







635.
Mm.86385
Chromosome 3
ATCCTTTAGAT






GTTAGTACAGT






GTTTATGAGAA






AACTGTTACTA






GAAGCTGAAG






AACAGC







636.
Mm.2655
Chromosome 17
AGTGTTCTATA






TGTGTAAATTA






GTATTTTCAAC






TGGAAAATGTT






GGCTGGTGCAA






AAGGC







637.
Mm.188851
Chromosome Multiple
GTCTGGGCTAG





Mappings
TGCCCGTTTTT






AACCCTACCCA






TTGATCATTTC






AAGAAACCTCT






GGTTA







638.
Mm.228682
Chromosome 16
TGTAAGACCAT






TTCTAAATTGC






TGGTAATAGAA






ACTCATGGCAG






TAAAAATGTAA






CCTCG







639.
Mm.2992
Chromosome 18
ACTGGAATAG






GAATGTGATGG






GCGTCGCACCC






TCTGTAAATGT






GGGAATGTTTG






TAACTT







640.
Mm.28580
Chromosome X
TCTACTAGAAG






GGTTAAAAGCC






ATATGAATGCA






AGAAATCATTT






GAGGCTTAAA






ATGCTG







641.
Mm.62886
Chromosome 8
GGACACCATTT






TTCATGTTAAA






TAGATTTTAAC






CTCGTATCTAT






GCATAGGCTAA






GGTGG







642.
Mm.65363
Chromosome 2
TAGATAAAGCC






CGTATGAGAA






GAGAAAACCA






AATTAATCCAC






TTCAGCAAAAA






GAAAGCC







643.
Mm.22288
Chromosome 7
CAATGTCAGAC






TGCCATGTTCA






AGTTTTAATTT






CCTCATAGAGT






GTATTTACAGA






TGCCC







644.

No Chromosome location
CTTTGGGGGGG





info available
GTTTTGGAAAA






CCGGTTTTTTC






GGGGGGGTTTC






CTTTTGGGGGG






TTTTT







645.
Mm.283461
No Chromosome location
GCCATACAGCT





info available
TATATTTGTAC






TGGTATGTCCA






GAAATCATGG






AGGAAAGAAA






AGTAAAA







646.
Mm.143724
Chromosome X
TGGTGTTTTGA






TTACAGTGAGA






CATCACAGGTT






ATCTAAAAGCC






CTTCGTTATAA






CCAGC







647.
Mm.217092
Chromosome 3: not placed
TATTTGGTGGT






AAAGAATATG






GTTGAAAATTG






TCATCCACATG






CATGCATCAAG






TAACAC







648.
Mm.87062
Chromosome 6
CGAGGAGTTAT






TAGGGAGAAT






CATGGAGCCAC






ATAAGAAAAT






CTTGGGCAAGA






AAAGAGG







649.
Mm.21126
Chromosome 1
TGGTGACAGG






ATTACGTGAAA






ATCTCTGACAT






TGTGATAAACT






CGATAAAGGCT






TAAGAG







650.
Mm.11778
Chromosome 1
ACCCTTTGCTT






AAATAGTGGG






AAAACGTGAA






TGTTTAGCATA






ATATAAAAAC






ATGCAGGC







651.
Mm.261448
Chromosome 14
GTTGGACTCTA






ATACAACTGAC






CATTGAAAAAT






GAACAACGGC






TTATTGTTTTG






TAACAG







652.
Mm.250414
Chromosome 7
TTCCTACAAAG






TGTGTTTCTAT






AGGATTACTAG






AGTAGCGGTTT






TGTACTGTGAG






GAAAC







653.
Mm.33764
No Chromosome location
TAGATAACAGT





info available
GACTATTGACG






ATTTTAGTAAA






AGAAAGTTGA






CATGCGTACCG






CTACCT







654.
Mm.27579
Chromosome X
GGGGGGACAG






TTAATATCGTT






TGTTAGATACC






ATAAGTGGTGG






AAATAAAGTG






ACTAAAG







655.
Mm.71633
Chromosome 13
AAAGAGGAAA






CTGTCCTATTT






CTCAACTGATA






AGTACTCCTGG






TAAGATGTAAT






ATTTGC







656.
Mm.173983
Chromosome Un: not
CAAATGTACTG





placed
AGAAACAAAA






TCATGAACGAC






CTTGAAATCAC






CTTCTTATTTC






AGCTCC







657.
Mm.117055
Chromosome 5
AACATAAATCA






AAATATACTTA






GGAATATTTAC






AATTAAACATG






ATGTTTTAAAC






TTAGT







658.
Mm.141083
Chromosome 2
GACTATTTATT






AGATTAGAAA






GTCATGTTTCA






CTCGTCAACTG






AGCCAAATGTC






TCTGTG







659.
Mm.198119
Chromosome 1
ACAAACACAT






GAAAAAATCA






AGTAGGAACT






GGAGAAACGT






CTCACAGTTAA






GAATGTTTG







660.
Mm.6156
Chromosome 5
AATTCACAGAT






GGCTTACATTT






ATGTAAAGAAT






TCCTGTAAGGC






ACTCATGTTTG






ACATC







661.
Mm.6456
Chromosome 5
TATACCAAACT






GAAAACGTTTA






AATCTCAAATG






AAGTAAGCAA






GGTTTTGTTCT






CCCTGC







662.
Mm.30015
Chromosome 4
TAGCCATTTAG






GAGATGTCCCT






TCAAAGTGACG






TGATGATGGAC






TTGCACTTGGG






AATCA







663.
Mm.31079
No Chromosome location
GCTCAGCTTAG





info available
GCTAGACTTTG






ACCAGGTAAG






CAGAAGAAAT






GAGAAACAAA






ACTCAGCA







664.
Mm.295618
Chromosome X
TATCACTGGAA






TATTGAAAGGT






TGTATGTAGTA






TGGGAGATCA






ACTTTCTTCCC






TAAGGT







665.
Mm.171323
Chromosome 4
ACTGCTGAGAA






AAACAAAATTC






ACTACATACCT






CAATAGTTATT






TACCATGAGAT






TGGCG







666.
Mm.173106
Chromosome 1
GAAGGAAATG






CAAACACCTTT






GAACTTCAATT






CTTTCAGTAGG






AAAACAAGAA






TTGTCCC







667.
Mm.206737
Chromosome 1
AGAAAAACAC






TAAACTCCAAA






TTAGTATAATA






ACGAGCACTAC






AGTGGTGAAA






AAGCTCC







668.
Mm.19945
Chromosome 14
AAAGGAATCTT






AAGAGTGTAC






ATTTGGAGGTG






GAAAGATTGTT






CAGTTTACCCT






AAAGAC







669.
Mm.182061
Chromosome 11
GAAATGGATTT






TGAGGCTTTGA






AAATGAAAAT






GGCTAGTATCT






CAAAGATGTCA






GTATCC







670.
Mm.265618
Chromosome 14
ACTATTTCTTG






TCAATAGTTTG






GCAAAAGACG






ACTAATTGCAC






TGTATATTGCC






AGTGTA







671.
Mm.22687
Chromosome 7
TCCTCTAAAGA






TGTGTCTTATA






TACATGATTGT






CATTGGTGGGC






TCAAACAATAA






GGGTG







672.
Mm.56769
Chromosome 10
TTGGAAACTAC






AAGTAACCCTC






AGACGGCCTA






ATTCTTATAAT






CCGGAAAAAC






ACCCCAA







673.
Mm.34356
Chromosome 8
GTGTGATAATC






TTTTCATGTTTT






CTAGAGCAAA






GACAAAGCAG






TTACTCTTCTA






TCGCAA







674.
Mm.34510
Chromosome 3
GGCTTTAGAGA






AAACTTCGGTC






TTCAAAGAACT






CTTCTAATTAG






TTCCTTCTTGG






AAAAA







675.
Mm.4664
Chromosome 4
AAAGTAGGAG






ATGAGATTTAC






ATTTCCCCAAT






ATTTTCTTCAA






CTCAGAAGAC






GAGACTG







676.
Mm.24730
Chromosome 2
AGTCCTCTGCA






TGTTTCCAAAA






TTTCCTTTACA






TGAAGGCTATA






TTGGATCAGAG






CTTAC







677.
Mm.159840
Chromosome 5
AAGAATAAAT






CACTTGAAATC






ATACTGTTTTT






GGAAATCCAA






ACTGTTTAAAG






AAAACTT







678.
Mm.291487
Chromosome 6
GTTAGATGCCA






TTGAAGGGGA






AATAACTTTGG






CTAATAGCTTG






GAAAACTCAGT






ACTAAG







679.
Mm.73777
Chromosome 18
AGCAGATATGT






GACTTCTCATA






TACACAGTTAC






GCTAACTCAGG






TGTATGATGAA






TACAG







680.
Mm.221709
Chromosome 19
TGTCTATGGGA






GAAGTAATAG






CCTGAAATAAG






ATAAGGCTCAA






ACAAACACTAC






TTACTT







681.
Mm.259122
Chromosome 5
GGGAAGAAAA






AGAATTGGTCG






GAAGATGTTCA






GGTTTTTCGAG






TTTTTTCTAGA






TTTACA







682.
Mm.218530
Chromosome 11
CTTGAAGAAA






AGTATATCACG






TAGGCATAGAT






GAGAAAGCCG






TTTGATCAAGT






CTGGTTA







683.
Mm.108076
Chromosome 13
TCCTTCAGTCA






GATATCTGTCC






CAGAGAAAGG






AAAATAAGGA






GCATGGTAAG






AAATGAGT







684.
Mm.204920
Chromosome 13
TATGGAATGGA






GAAATAAATA






CATCTGTGTTG






AAGAACCTTTT






GATGGAACTA






ATACCGC







685.
Mm.162073
Chromosome 6
AGGTCAATGTT






AAGTTTTCTGA






GTTTAATATAT






AGTTAGGGTGA






AAGACTTAGCA






CACGG







686.
Data not found
No Chromosome location
AATGCTTAACT





info available
TTGAGTCACAC






TGTTTACCCTT






CCTATGAGGTT






GCATTTTGACA






ACAAC







687.
Mm.248267
Chromosome 18
TAAAGGGAAC






CCCCATTTCTG






ACCCATTAGTA






GTCTTGAATGT






GGGGCTCTGAG






ATAAAG







688.
Mm.20847
No Chromosome location
CCCCTTTTTGT





info available
AACTGGGATAT






AAATCCTTGAA






AGAAAGGAGA






ATTTAGAGTTT






TGCCCC







689.
Mm.200366
Chromosome 5
GTCAGTGAGTT






GGTTTCCTTTC






CATCAGGAAA






AATGGATTCTG






TAAAGAGTCA






GGGCGTT







690.
Mm.28835
Chromosome 8
GAAAGCCGTC






AGCGAAAGTTT






TCTCGTGACCC






GTTGAATCTGA






TCCAAACCAGG






AAATAT







691.
Mm.25148
Chromosome X
GAAATATGTTA






ACTAAGAGCA






GCCCAAAAAT






ACTGGATATGC






TTATCCAATCG






CTTAGTT







692.
Mm.10760
Chromosome 15
GTATACAATGC






TATTTTTAGGT






TAAGGCCTAAA






CTTCTGAAGAT






CTTGGTAACAG






CAGAG







693.
Mm.89961
Chromosome 13
GGATGAAGTG






GAAGATTACTG






GCAGGTCCAA






AAACCTGATTT






TCTAGTACATT






TCACTCT







694.
Mm.8655
Chromosome 1
TTCAATCAAGA






AAGTAGATGTA






AGTTCTTCAAC






ATCTGTTTCTA






TTCAGAACTTT






CTCAG







695.
Mm.28890
No Chromosome location
AAATTTTCTTA





info available
AAGCTATGAAC






TCTGACTTTTG






ATTTTGTGTTT






CCATTTAGTAG






AAACT







696.
Mm.3368
Chromosome 13
AGAATCTCACT






ACTAAAGTCAA






GTATAGAAATA






ACTGTTCTTAT






GTTTTCCTCCA






AGGCC







697.
Mm.143689
Chromosome X
ATCTTTGGCTA






TATTTTCCTGG






TAGCATATGAC






AAATGTTTCTA






CAGTGAGAAG






CTGAGA







698.
Mm.27385
Chromosome 15
GGGTTATAATG






CACTGAGATCC






AGAAGTTGGG






AAAACTCAATA






AATGTACAAA






GGAAAGC







699.
Mm.171399
Chromosome 1
TACTTGTGTGA






CAAGCTAGAG






AAGTTACAGA






AGAGAAATGA






CGAACTAGAA






GAGCAATGC







700.
Mm.4554
Chromosome 4
TAAATAATCCC






TTCCCATGAGC






CCACTGCTCTG






AATGGACAAG






CTGTCCTTATC






TTCAAT







701.
Mm.27800
Chromosome 18
AAATAGTTGTT






TTTAAGGTTGA






AGGAAGAGAC






ATTCCGATAGT






TCACAGAGTAA






TCAAGG







702.
Mm.268027
Chromosome 2
TGAATCTACAG






GCAACTCTTCA






TCTCTGTAATG






CTACCTGACTT






CTCTTGTGAGG






AGCTG







703.
Mm.103300
Chromosome 14
TGGCAAAGAG






TAGATGAGAA






AATGTTGGATT






TAAATCAGCAG






ACTCATTTCAT






ACTTTGC







704.
Mm.22194
Chromosome 10
ACCACGTTTAA






ATGACCAGTCT






CAGGATAAAG






AGTTTTACAGA






AAATTTAAAAT






GCCTGG







705.
Mm.29820
Chromosome 14
GACATCGTTTT






CTCTCTAAATT






CAGTAGCAGTT






TCATCGACAGT






GCCATTGAACT






ATGGG







706.
Mm.4859
Chromosome 4
TCTGTGGGGTT






CTCATGCCAGT






GTCTGAAATCT






CACCTCACTAG






AGATGTTTCTC






GAATT







707.
Mm.30111
Chromosome 11
TTCCAGTTCTC






ATGTCTTGAGA






TTTCAAGTAAA






GATGTGTTAGT






GTAAGCTCAGA






TCCGA







708.
Mm.37770
Chromosome 14
AACCATTGGGA






AAATGCAATAC






AGATAAACTA






GAGATTCGTAT






AATGCCACGTG






TTAGCT







709.
Mm.447
Chromosome 1
GTGAATGGAGT






GTTTACTGTAT






GTAAGAAAGA






AGAAAAGTGG






AACTACATTTG






CTATGAG







710.
Mm.182857
Chromosome 5
TTCACAATTTA






GACACAAGATT






TGGAAGATTGA






AACTGACATGA






AAGTCTTCTTC






CTGAG







711.
Mm.2277
Chromosome Multiple
GAAGATTTTTT





Mappings
GATGTATAAAA






GTGGCGTCTAC






TCCAGTAAATC






CTGTCATAAAA






CTCCA







712.
Mm.27436
Chromosome 15
AGAATGAACC






AGAATGGAGA






AAACGTAAAA






TTTGAAGAATC






TCGTTGAAGAG






CTATTTGC







713.
Mm.133824
Chromosome 10
TCGACAAGAG






GTAATCCGAGA






AATGGAGCAG






AAAACCTCCTT






GCACTTCAGTG






ATATACA







714.
Mm.27829
Chromosome 4
TATATGCAACT






TCATAGATCCT






CTGCAATATGT






ACTTAGCTACC






TAAGCATGAA






ATAGAC







715.
Mm.34674
Chromosome 19
CGTCATATATC






CTATTTGTAAT






CAAGAGGAAA






GACTACATTAA






GAAGATAGGG






TGCATAG







716.
Mm.4481
Chromosome 6
CTCAGATCAGT






TCTTTAGAAAG






AGCTGGTATAG






AAATGGGTGAT






GTAAAACTTGA






GAAGC







717.
Mm.203928
Chromosome 19
AATGAAAATCT






GCGTCTAACTT






TTGAAAGTAAG






TGTTAACTTAC






TTGAATGCTGG






TTCCC







718.
Mm.214553
Chromosome 15
AATCTTCGACC






AGACATTGGAT






ATTTGAACTAT






CCTGAAACATT






TTAGAAATATC






CAGGC







719.
Mm.22370
Chromosome 8
TACCCCATTAA






AGGCATCAAAT






CCGGGTTTAGA






TCAGTCCCTCT






GAAGAATGGG






TACAGT







720.
Mm.4462
Chromosome 15
TTTTTTCTCTTG






CCAATGTATTT






TTGTAAGGCTC






GTAAATAAATT






ATTTTGAACAA






AACA







721.
Mm.18635
Chromosome 7
CACACCCTCTG






ATGTTCCAAAA






GCTCCAGGACC






AGATCTTCAAT






CTCATGAAGTA






TGACA







722.
Mm.162929
Chromosome 19
CCCAGGTATTT






CTAAGCATGCT






AGGTTTGAGGT






CATTTACCATG






TTCAAATAAAA






GACGG







723.
Mm.255070
Chromosome 9
GGAGCAAAAC






TTGAATAATGT






CCTTTATCCTG






ATTTGAAATAA






TCACGTCATCT






TTCTGC







724.
Mm.173654
Chromosome X
TGGAATAAGA






AAGAATCTGTG






GTAGAAATAAT






AGACTTGCTAC






ATAGGGTTAGC






TAAGGC







725.
Mm.30664
Chromosome 11
ACCACAGTTTA






TCAGCATTTGA






AGATTTCCTTG






ATGATCCATAC






TTGTCTTGGGA






TAGGG







726.
Mm.788
Chromosome 15
AGGGTCAGCG






CCGAATCTTGT






GGACACACTG






ACAAGGATGTC






TAATCCAAATA






GATGTAT







727.
Mm.248907
Chromosome 9
AGTGGAGTATT






CAGTCTGGAGT






TTCAGGATTTT






GTGAATAAATG






CTTAATAAAGA






ACCCT







728.
Mm.34399
Chromosome 4
TTTGGGCCCTT






AAAAACATATT






TCAGTTTTGCC






CAAGTGAGGC






CTTAAAAATTG






CCCATG







729.
Mm.424
Chromosome Multiple
AAAGGAAAAT





Mappings
AAAGTGGATCT






GAAAGTAGAC






TCTGCTTCTGC






GCATGTGTGAG






TGGTGCC







730.
Mm.259278
Chromosome Multiple
TTCACTCCTGG





Mappings
ACTGTGATTTT






CAGTGGGAGA






TGGAAATTTTT






CAGAGAACTG






AACTGTG







731.
Mm.219676
Chromosome 2
CACCATCCTTC






CAGAATATGGT






ATGAAAAATCT






ATGCAAACTGT






GTAAGCTTTTG






CTCAT







732.
Mm.145306
Chromosome 12
TTGTGGAGTGT






GAAATAAAGG






ATAATTGCCTA






CCTCTAGCAAG






TGGATCTTATT






ATGTTG







733.
Mm.22564
Chromosome 17
ACCAGAAAGG






ACAGTCTGGAC






TTCAGCCAACA






GGACTCCTGAG






CTGAGATGAA






GTAACAA







734.
Mm.274876
Chromosome 7
GATACTGCCGG






CTTTGAAAATG






AAGAACAGAA






GCTAAAATTCC






TGAAGCTTATG






GGTGGC







735.
Mm.205421
Chromosome 14
CCATTTGAGCC






TCACTGCAATG






TTAGTGCAGAG






GAGAAAACAA






TTTTTAATGTA






ATCTTG







736.
Mm.268911
Chromosome 1
GGCAACTTGTA






AAGTGTGTTCA






TTCTAACTGTT






AAACTGAGAA






AACTTGAGAAC






ATACTG







737.
Mm.269064
Chromosome 14
CAGAAGAGAT






TCTGAAAATGT






TAGTTGTGGTG






ACTCTAATGTA






GATCCATAACT






GAAAAG







738.
Mm.218665
Chromosome 15
TATCGTAAGTT






GCACCTATTGT






TAAGTGGAAA






ATGCTCTGATT






ACACTCAGGA






AGCTGGG







739.
Mm.21450
Chromosome 5
TGTTTTGTCCC






TAAATCACCAC






CACTCACTATT






TCTCCCAGGGT






CTGATAATGCC






TTTAC







740.
Mm.28908
Chromosome 8
AGCCACTTTAA






CTCTAAACTCG






AATTTCAAAGC






CTTGAGTGAAG






TCCTCTAGAAT






GTTTA







741.
Mm.259295
Chromosome 17
GCTTTGTTTAA






ATGGTCAGACT






CCCAAACATTG






GAGCCTTTTGA






ATGTGTTCTGA






GACCT







742.
Mm.154623
Chromosome 18
CCTTAGAAAGA






TGGTAATTCAC






TTTAGGTAAAA






GTACTATTTCA






CGCCATTATGA






AACCC







743.
Mm.29628
Chromosome 19
TAAAATGAGG






CTTTTGGAAAG






AAAGATGAAA






ACGTAGAATGT






AGTGCTAAGA






ACGTTTCC







744.
Mm.163
Chromosome 2
GCAGTTACTCA






TCTTTGGTCTA






TCACAACATAA






GTGACATACTT






TCCTTTTGGTA






AAGCA







745.
Mm.6272
Chromosome 9
TGCTTAGAACT






ACATAGAATCA






GAAGCAAAAT






GGATGCCTTAG






CACTGAGGAA






AGGTTTC







746.
Mm.70065
Chromosome 10
GGTTTTCGAAC






CACGTACCTTT






ATGCCTCGTGA






TTGTGAAACAT






TGACTTTTGTA






AACCC







747.
Mm.21579
Chromosome 13
GTTCACTGTAG






AAATTTGTGAT






AAGAAAGACA






CACAGACGTA






GAAAATGAGA






ATACTTGC







748.
Mm.21138
Chromosome 14
AAAGACTTTTT






TGGACTTAATA






CTGATTCTGTG






AAAACTGAAG






AAGTGTAGATG






TCTCCC







749.
Mm.486
Chromosome X
CTGGTGTGGGA






TATTTTCCACA






CTTTAGAATTT






GTATAAGAAA






CTGGTCCATGT






AAGTAC







750.
Mm.247440
Chromosome X
TAAAGGTTTTA






GTGTCCTAACT






CCCCAGGATCA






GGAGATTATCC






CAACTATTTCT






GGGGT







751.
Mm.34462
Chromosome 5
CTGAATTTTGA






TCACTTGTGGT






TTCTCATGGTG






ACCTCCATTTG






CAACAAAAAG






ATGTCT







752.
Mm.154684
Chromosome 2
TGTGCTTTACC






AAAATGGGAA






ATAATTCTGCT






TTAGAGGATAC






TATCAAGACAA






CCTTAC







753.
Mm.30251
Chromosome 17
TCTGTGAGATG






TTGTAGACATT






CCGTAAGAGA






ATCCAGAATGA






TAGCAGGATCA






GGAAAG







754.
Mm.243085
Chromosome 6
CTTACATGATC






TCCTAAAAGGA






TGGGCCCCTCC






TTCCTTTTGCG






GGTTGAAAGTA






ATGAA







755.
Mm.41525
Chromosome 10
CTGTTTAAAAA






ATGAAATCAG






GAAGCTTGAA






GAAGACGATC






AGACGAAAGA






CATTTGAGC







756.
Mm.45563
Chromosome 1
TGAATATAGTA






GGGCCATGAGT






ATATAAAATCT






ATCCAGTCAAA






ATGGCTAGAAT






TGTGC







757.
Mm.221705
Chromosome 19
GGGGGAAATT






CTATATGAGCT






TCGTTTTCTAA






TGACTTACATG






GATAGTATGGA






AACTTC







758.
Mm.19142
Chromosome Multiple
AAACTTGAAA





Mappings
ACACAGACATT






GAAGGAATCA






TAGGTATTTTT






GCTTTATGCTC






TCTGGCA







759.
Mm.139860
Chromosome 16
AATAAGCAGG






AAGAATTTGAC






TTGGAAAACTA






ATACACGCATG






TTAGGCATTCT






CAAGGC







760.
Mm.200936
Chromosome 5
TCCCACTGTTT






ACAGATGTAGT






TCTTGTGCACA






GGTGCCACTAG






CTGGTACCCTA






GGCCT







761.
Mm.37562
Chromosome 7
TATTTTTGTCA






TTGCCTCTAGT






GATTTTTGTAA






ATGGGAATGG






AAAAGTACAA






GGCAACC







762.
Mm.35600
Chromosome 9
TTAACTGGCCT






GTCAAACTGGT






CTTGAAGCGTC






TCTAAGTGAAG






AGCCAGAAGA






AACCCT







763.
Mm.213128
Chromosome 9
CAATGTGATTT






TTCAATGGTAT






TAGTTCAAATT






GACGTGGATTC






ATGCCACATGG






AAATC







764.
Mm.46501
Chromosome 12
AACTGAATAA






AGTTGACCAGA






AAGTGAAAGT






CTTTAACATGG






ATGGAAAAGA






CTTCATCC







765.
Mm.227202
Chromosome 3
GGATATAAAGT






GTATTTCTTTC






AGTGATTTCTC






AGTGCATAAG






AAGTGCATAA






GTCTCAG







766.
Mm.43444
Chromosome 6
TAGCTTTTTAA






AAGAAGTTTTT






CTACCTACAGT






GACCATTGTTA






AAGGAATCCAT






CCCAC







767.
Mm.248456
Chromosome 13
ATTTGCAAGGT






CAGAAACTAG






CCAAGGTCCTT






CTCAGGCATCT






ATCCTTAACTT






GGTCTC







768.
Mm.30103
Chromosome 11
TTGGAATTTGA






GGAGGAGAAA






TGAAAAAACA






GTGTGTCCCTG






GTGTCACCCTG






GCATCAT







769.
Data not found
Chromosome 10
TCTTATGATTT






AAGTGATTGGT






GGATAAATGTA






TAGGAATTTTA






CACTCCAGCAG






CATGG







770.
Mm.26658
Chromosome 9
GCCTCAAATGG






AACCACAAGT






GGTGTGTGTTT






TCATCCTAATA






AAAAGTCAGG






TGTTTTG







771.
Mm.34702
Chromosome 8
CCGTACACAAA






AGTGAAGATTT






CAGCGAAATG






CCAAGGAAGT






GCCATCTATCT






GGCTTCT







772.
Mm.2433
Chromosome 17
AAGAAAATGC






TGTATGATGTT






AGAAGACATT






GTAATTATCAT






CCCGTGTCTTT






GCTGTAC







773.
Mm.270044
Chromosome 8
GGCATTTCAGT






TTATCTTGGGT






TTGTAATTAGT






TAAAACAAAA






ACCAACCTAGG






TCTGTG







774.
Mm.268014
Chromosome 10
ATTAGCCAAGG






AGTCCGGACAT






AATATTTATCC






AGATCTCTAAG






CAGTTAGCTTT






AAATT







775.
Mm.549
Chromosome 10
TACATTAGCTA






ATACTAACCAC






ATAGAATATCA






GACTTAGATAC






GTGAATAGGG






ATCCTG







776.
Mm.276062
Chromosome 4
AAGATTTTCTA






GTCACTGCATA






AAGGAAACGC






CTAAGAGTTGC






CGTATTGCTTT






CTGAGA







777.
Mm.26939
Chromosome 3
ACAAGAATTCA






TTCTTAACATT






TGAACGAGTGT






ATTTGCTTAGG






TCGATGAAAGT






GTTGC







778.
Mm.247480
Chromosome X
AGGATTTTCTC






ATGAAGAACC






AGATGACATGT






GGTAATAACAT






TAGCTGTCTAG






TTTCTC







779.
Mm.182877
Chromosome 1
TAGAGTCATGA






AGAACAGAAA






TTCAAGGTCAT






TTTCAATTACA






GAGTGAGGTTA






GAGCCA







780.
Mm.121973
Chromosome 15
TCTAAAACATG






CCAAATGACTT






ATGTCACAAAG






AATAGGTCCTA






ATATACTGTAT






ACCCC







781.
Mm.139738
Chromosome 13
GTGTTTCTTCC






CATTTGTAAAT






GTCCTGAACCA






TAAATTACTAT






CAGGATTAACT






GACAG







782.
Mm.27969
Chromosome 1
GAAGCTGGAA






GCATTTGTTTT






TGAAGTTGTAC






ATATTGATAAG






TCAGCGTATGT






GTCAGA







783.
Mm.222307
Chromosome 2
TTACATGGCAA






ATCTGAAAGG






AAGACTTAAGC






AGGGTAAAGTT






AATTGAAAGG






AGGAGCT







784.
Mm.1258
Chromosome 6
AGCAATCTTTG






TATCAATTATA






TCACACTAATG






GATGAACTGTG






TAAGGTAAGG






ACAAGC







785.
Mm.24933
Chromosome 1
GGTGTATGGAA






ATAAAGTTTAG






TCAATGTTGAA






AATCTCTCCTG






GTTGAATGACT






TGCTC







786.
Mm.1940
Chromosome 15
CTTTCAGTCTC






CTTCTGTGTCT






CGAACCTTGAA






CAGGATGTGAT






AACTTTTCTAG






ACCAC







787.
Mm.215584
Chromosome 2
GACTGTTTCTG






GGAAAATAAG






TATGTGAAGTG






ATGCAGAAAA






TCCATCTAGAC






AGTTGAG







788.
Mm.216113
No Chromosome location
TGGTGGCTTGA





info available
TTGATTTGATC






TGAGAGCAGTT






TATAACATAAT






GGAGAACTGTT






TGCAG







789.
Mm.2623
Chromosome 13
AGAAGTCTACC






TTTAAGATGAC






CTATATTGGAG






AGATATTCACT






AAGATTCTGTT






GCTTC







790.
Mm.264709
No Chromosome location
ACTCTCTGGTC





info available
ATGATGGTTTT






CCGAAATCAG






GTTCCTGACCT






GAAAATTTGGG






TTAATC







791.
Mm.20323
Chromosome 5
GTTTTCATGCT






TTGGAAGTCTT






TTCTTTGAAAA






GGCAAACTGCT






GTATGAGGAG






AAAATA







792.
Mm.222093
Chromosome 1
GTGTGTAGGAA






AATGTAATTAA






GTACAAGGCTT






GTTTATGGGTG






GCTATGGAATG






CAGTC







793.
Mm.25035
Chromosome 6
GTTTCCTCATC






AGGTGTAATGG






CGTGTCCTAAT






GAAGCTATTTC






TTATGTATAAC






AGAGA







794.
Mm.103545
Chromosome 11
TGAAAAAATG






AAAAGAATCA






GAGATGAAAT






AGGAGCGCTC






AGAAGTTTTTA






TGTTCTCCC







795.
Mm.26229
Chromosome 11
AAAGAAATGA






AAACCGTCATT






TGCGATTTTCA






GGGTACGTTTC






TAATGTATCCA






GAAGTC







796.
Mm.22699
Chromosome 15
TTTCCAGTGTT






CTAGTTACATT






AATGAGAACA






GAAACATAAA






CTATGACCTAG






GGGTTTC







797.
Mm.275510
Chromosome 17
TTTTGACTCAG






TTGACTGTCTC






AGACTGTAAG






ACCTGAATGTC






TCTGCTCCGAA






TTCCTG







798.
Mm.275745
Chromosome 3
CCCGAGTTACT






AACAACATTCT






TTTGCTATATG






TAGATCAAGAT






TAACAGTTCCT






CATTC







799.
Mm.80676
No Chromosome location
GTTTTGGTGCA





info available
AAAGTCGTCCT






GTGTCTCTTGT






TCCCTTCATTA






GAAAACATGCT






AGAGG







800.
Mm.197381
Chromosome 12
AGGAAGGAAA






ATAGGCTTTGT






TGTATGTACAT






AAGTGGAATTA






ACAAGAGTCTT






TAGTCC







801.
Mm.276618
Chromosome 15
TACAGGGAAT






GGTCTAAGCAT






ACCATTTCATT






CACTGTATTAG






TAGACATAACT






GTTGAG







802.
Mm.46636
Chromosome 10
GAAACGGGCTT






TGTTGTAAAGG






TAATGAATAGG






AAACTCCTCAG






ATTCAATGGTT






AAGAA







803.
Mm.132926
Chromosome 13
AAGTTAAGGA






AATACTGAGA






ATCGGTCAGTT






AACACTCTGAA






AAGCTATTCAA






AGCATAG







804.
Mm.62
Chromosome 15
AAATACATGCA






TTTGTACAGTG






GGCCCTGTTCT






TGTGAAGTCCA






TCTCCATGGTC






ATTAG







805.
Mm.117473
Chromosome 9
CCGTTTTATTG






ATTGGAAATGT






AAGACTCAAA






GAACTCAGGTT






TACTGGCCAAG






ATGGCA







806.
Mm.2692
Chromosome 3
GGAAAGAGAG






ATCAAACTAGG






AACCTACAAG






ATAGTTCACTA






GCCTAAGATCT






TTACTTG







807.
Mm.196533
Chromosome 9
TTGATTGGTGT






TTCTGAGCATT






CAGACTCCGCA






CCCTCATTTCT






AATAAATGCA






ACATTG







808.
Mm.216997
Chromosome 10
CTAGTGAAATT






TATGTCAGAAT






GACATATCTGA






ACTCTGAATTC






ATCTCTAGTTT






CCACG







809.
Mm.29476
Chromosome 11
TAGTTAATACT






TCTCTGAAATA






CATGGTAACAA






CTAGTAAGCAA






GAGATACCGC






AGATTG







810.

No Chromosome location
TGGATTATTCC





info available
CGCCAAAGCA






CCCAAGTCGGC






CTGTTTAATTG






GAGAAAGATG






GAATTAA







811.
Mm.41781
Chromosome 19
GATCCAGGCA






ACCTCTGTTTA






CCCTGGGGCCT






ACAATGCCTTT






CAGATCCGTTC






TGGAAA







812.
Mm.142105
Chromosome 3
GTTCCATCTGA






CTTAAACAAAA






ACCGTAGTTTC






CAGCTCAGAAT






CATCCTAACAT






AGAAA







813.
Mm.203928
Chromosome 19
GTAGGGGAAT






AACTAACCAA






AGTAGAGGGA






ATTCTAAGTTT






AGTAGTAAATG






TGGCTTGG







814.
Mm.24128
Chromosome 6
GGTGTGGGACT






TATGGGGTCTA






CACAAAGGTA






AAGAATTACGT






GGACTGGATCC






TGAAAA







815.
Mm.221784
Chromosome 1
AGGTATGACAT






TTTACATCCTT






GAATCTTACTT






ACTATGTGCTA






AACAATTGGCA






GAAGG







816.
Mm.86699
Chromosome 16
TGCTTGTGTGA






ACTACCTCAGG






ATGAAGGGTA






ATGTTTAACAT






TCCATACATGC






CTACTG







817.
Mm.3317
Chromosome 5
CGATGGACCCA






AGATACCGAC






ATGAGAGTAGT






GTTGAGGATCA






ACAGTGCCCAT






TATTAT







818.
Mm.22383
Chromosome 15
GCAGCCAAAA






TGGAAATGTTT






AAATTAACTGT






GTTGTACAAAT






GTACCCAACAC






AAAACC







819.
Data not found
Chromosome 13
TTGACATGATA






CATTACGCCTT






TGCAGTGAGCT






AATAAGCTAAC






ATTTGTGCACA






GATAA







820.
Mm.257567
Chromosome 15
TCTCAACTCAT






CTCAGATTAGG






AAGTATTTGGC






AGTATTAGCCA






TCATGTGTCCC






TGTGA







821.
Mm.12912
Chromosome 7
ATTTTCATGCC






GAATATTCCAG






CAGCTATTATA






AAATGCTAAAT






TCACTCATCCT






GTACG







822.
Mm.465
Chromosome 6
GAGAATTAATC






ATAAACGGAA






GTTTAAATGAG






GATTTGGACTT






TGGTAATTGTC






CCTGAG







823.
Mm.21764
Chromosome 18
CATGAGCAAA






GCCCACCCTCC






CGAGCTGAAG






AAGTTTATGGA






CAAGAAGTTAT






CATTGAA







824.
Mm.222266
Chromosome 19
CTCTGTAAAGT






CAAGTTGCATT






GCATTTACAGT






TAATTATGGAA






AAGTCCTAAAT






CTGGC







825.
Mm.40285
Chromosome 12
TTTTCAGGGCT






ATAAAAGTATT






ATGTGGAAATG






AGGCATCAGA






CCACCGGACGT






TACCAC







826.
Mm.41940
Chromosome Multiple
AAGAAGCTGA





Mappings
GGAAAAACAG






GAGAGTGAGA






AACCGCTTTTG






GAACTATGAGT






TCTGCTCT







827.
Mm.263124
Chromosome 15
CCTGATGGAGT






CTGTGTTACTC






AGGAGGCAGC






AGTTATTGTGG






ATTCTCAAACA






AGGAAA







828.
Mm.21974
Chromosome 9
AGCAAATGGG






CATTTTACAAG






AAGTACGAATC






TTATTTTTCCT






GTCCTGCCCCT






GGGGGT







829.
Mm.25843
Chromosome 6
CTGCACTTGAA






TGGACTGAAA






ACTTGCTGGAT






TATCTAGAACA






ACAAGATGAC






ATGCTAC







830.
Mm.896
Chromosome 1
AGATTTCACCG






TACTTTCTGAT






GGTGTTTTTAA






AAGGCCAAGT






GTTGCAAAAGT






TTGCAC







831.
Mm.30466
Chromosome 15
ATAAAACCAC






AAACTAGTATC






ATGCTTATAAG






TGCACAGTAGA






AGTATAGAACT






GATGGG







832.
Mm.260433
Chromosome 18
ACCTAAATGTT






CATGACTTGAG






ACTATTCTGCA






GCTATAAAATT






TGAACCTTTGA






TGTGC







833.
Mm.145384
Chromosome 4
TTTATAGTTCT






AGGTTTACACC






AGAGAGGAGT






TAATTTATCAA






CAGCCTAAAAC






TGTTGC







834.
Mm.28385
Chromosome Multiple
TTCTTCCACGA





Mappings
ACAGATATTAT






GTCATTTTATC






CAATGCCGATA






AAGGAGAAAC






AACTTG







835.
Mm.27254
Chromosome 10
TACGTGGTCTG






GGGACCTGATG






TTGGAATCCTA






TTGTTGTTAAT






AAAACTGAGT






AAAGGA







836.
Mm.233891
Chromosome 10
ACCAACTTCTG






TCAAAGAACA






GTAAAGAACTT






GAGATACATCC






ATCTTTGTCAA






ATAGTC







837.
Mm.24021
Chromosome 7
TGACACAAATA






GAGGGGTCAA






TAAATTTTTAG






CCAAAAGCTTC






AAATTCTTTCA






GGAAGC







838.
Mm.141021
Chromosome 7
ATCACCATTGT






TAGTGTCATCA






TCATTGTTCTT






AACGCTCAAA






ACCTTCACACT






TAATAG







839.
Mm.292081
Chromosome 8
GCCGCTTTTTT






GTAACCTAAAA






GGCCCCATGAA






TAAGGGCCCAT






GTTTTGGGCAT






TTGTA







840.
Mm.275315
Chromosome 12
CCAAGAACAA






GTATAAACTTA






AGCTCTGTAGA






ACTGAAATTCT






TTCAAGTCCTT






TCGATC







841.
Mm.221696
Chromosome 6
AGGACATCTTG






CAACTTCTATG






CAATAATAAG






GATTTCCATCT






GACAAATAAG






ACAAGTG







842.
Mm.33922
Chromosome 5
GGGGAGTTCTA






ATAATAGTACC






ATTCATATCAG






CAAGAACCTA






AAAATGGTTCT






GACTTT







843.
Mm.27182
Chromosome 11
TGCCACTAGTT






CTGACTTGGGG






AATATGGTCCC






TTAAACATGCC






AAAGTGAGCTT






TTTAA







844.
Mm.2923
Chromosome X
CATCAATCCTT






TGATGGAACCT






CAAAGTCCTAT






AGTCCTAAGTG






ACGCTAACCTC






CCCTA







845.
Mm.8766
Chromosome 14
CAGTTGGAAA






AATGGATGAA






GCTCAATGTAG






AAGAGGGATT






ATACAGCAGA






ACTCTGGCA







846.
Mm.249306
Chromosome Un: not
TCAGTCAAATG





placed
TGCATAACTGT






AAATCAACACT






AAGAGCTCTGG






AAGGTTAAAA






AGGTCA







847.
Mm.87337
Chromosome 7
AGCAGGTGTTT






CGGACTTGCAA






TGAGCAATGCA






ATTTTTTCTAA






ATATGAGGATA






TTTAC







848.
Mm.258225
Chromosome 5
CTTGCTTCTTT






AGCAAAATATT






CTGGTTTCTAG






AAGAGGAAGT






CTGTCCAACAA






GGCCCC







849.
Mm.24159
Chromosome 11
TCTCAATTTTC






AAGGTGTATTT






CCTATCAGGAA






ACTTGAAGATA






ATATGGTCTGA






ACCCA







850.
Mm.14301
Chromosome 5
ACTGGACAAA






GTATTATGACT






TTCAACACCAG






GAGGTCTCCAA






ATACCTGCACA






GACAGC







851.
Mm.233547
Chromosome 4
GGCTGTTGAGT






GTAAAATGTGC






TTTGTGTTTGC






TTACAACATCA






GCTTTTAGACA






CACAG







852.
Mm.72173
Chromosome 14
TGAGTGCAATG






TGTCAGATTTC






ACCAAGAGAT






CTCCAAGGTTT






GTAGGTAATTT






GTGGTT







853.
Mm.101992
Chromosome Multiple
GTCATTGTCCA





Mappings
AGGTGACAGG






AGGAACTCAGT






CGTTAAAATGA






CGAGCCTTATT






TCATGA







854.
Mm.9336
Chromosome 3
TCTTAGAATCT






GGAATTGAGTG






CCATATTTTCT






GTTCTCCAATG






ATACCTGGAGA






AATCC







855.
Mm.15801
Chromosome 4
TGCTTTCTTAT






TCTTTAAAGAT






ATTTATTTTTCT






TCTCATTAAAA






TAAAACCAAA






GTATT







856.
Mm.159173
Chromosome X
CTGCATGTTAT






AACTTTATATG






ATGGTGTAGTG






CATATAAGCTA






TGAGAATCAGT






TATAC







857.
Mm.235074
Chromosome 8
CGTGCTGGAGG






ACGAGAGATTC






CAGAAGCTTCT






GAAGCAAGCA






GAGAAGCAGG






CTGAACA







858.
Mm.141157
Chromosome 3
TGGAGGCTTTG






TACCCAAAACT






TTTCAAGCCTG






AAGGAAAAGC






AGAACTGCGG






GATTACA







859.
Mm.39046
Chromosome 6
TGGAGGATCTG






TGTGAAAAAG






AAGTCACCCTC






ACAAACCGCC






GTGCCTAAGGA






CTCTGTC







860.
Mm.12090
Chromosome 1
CTATTTTGTGT






AGACATCGTCT






TGCCTGAATAG






ACTGTGGGTGA






ATCCAAATTTG






GTCCA







861.
Mm.221891
Chromosome 5: not placed
TAATTATCTAC






ATTGGGGTAAT






TGAAGTAGAA






AGATCCATCTT






AACTACGGTAA






TCTCCG







862.
Mm.235020
Chromosome 5
TTGGGTATCGT






TTATGTTTCCA






TCATAACACAT






GCAATAACATC






TAGGAAATCTT






TACCG







863.
Mm.269006
Chromosome 4
TCTGATGTGGA






AGTGCGGTCAT






TCCTGGTTTAA






CTCACAGCAAC






TTTTAATTGGT






CTAAG







864.
Mm.12829
Chromosome 1
ATCTCCTGTTA






ATGTATTTGGG






TCAAATGCAAG






GCCTTAATAAA






GAAATCTGGG






GCAGAA







865.
Mm.222131
No Chromosome location
GCAGCAAGAG





info available
AAAAGAGCAA






GAGAGCCAAA






GGCAAGAAAT






CTCTCTGTCAC






TCCCTTTTA







866.
Mm.288200
Chromosome 16
TGAGGAAAAG






CCCCATGTGAA






ACCTTATTTCT






CTAAGACCATC






CGTGATCTGGA






AGTCGT







867.
Mm.213420
Chromosome 11
ACCGGCTGTAC






CCAAATAGAA






CGTCATTTTGA






TATGAAGGATT






TCAGCCCCTGA






AGATTT







868.
Mm.131026
Chromosome 2
ATGGTTTCTTC






CAGCAATTTAG






CATTGCCTGAG






GGGTCTAAAA






GAATAAGTTGG






TTCTTG







869.
Mm.3401
Chromosome 19
ACAATCTCTGT






CAGCGAAAAG






TTCTACAACAG






CTGTGCTGCAA






AACATGTACAT






TCCAAG







870.
Mm.18830
No Chromosome location
AACTGTTACTG





info available
GATTGAAATTC






CCATCCCCTTT






CCCTAAAAATT






GTGCCTTAGAA






AACCC







871.
Mm.46184
Chromosome 5
CGACTGAGGTT






ATGACATCCTT






AGACTTTGTTG






TATGCTGCTTC






GAATGAACCA






GAGATA







872.
Mm.10117
Chromosome 9
TGCCTCTTCAT






CGCCAGTGGTC






CAAAGGGCGC






AGAGAGCGCA






CTAGCAGTCAA






TAGTGTT







873.
Mm.30219
Chromosome 8
CCACTAATATT






TAGCCAGCCTT






CATGTAGAAG






ACACATGGAA






ACACAGAAGT






AAACTTTT







874.
Mm.276229
Chromosome 10
AGAAATGAAC






ATACATTGTCA






GCATTTAGAAG






TAAGTTGTGAA






GACAGGGACA






TTAAGTG







875.
Mm.260594
Chromosome 5
CAAACGGGAT






CCTGTCTTCTT






CTTTTCTAATA






GAATTTTGTAA






AGGAAATGAA






TGTAGCC







876.
Mm.29467
Chromosome 8
ACCGTTCTATC






ACTGTGGATGG






AGAAGAAGCG






TCACTATTGGT






CTATGACATTT






GGGAAG







877.
Mm.154121
Chromosome 7
CTATTTTTGGG






AGATGTCTATT






GCGGAGTACA






GTAATATATAC






CCAGAGTATGT






CTATAG







878.
Mm.260515
Chromosome X
ACCCAACTCCA






GTGCTCTCTGT






CTTTTAGTACA






GGATTTTCACC






CATGTGCATGA






AAAAT







879.
Mm.21686
Chromosome 13
TTACCATTTTT






GGTTAAATGGC






CAAATTCAGAA






AATAACTCCAT






TTGAATCTCCA






GCAGG







880.
Mm.222196
Chromosome 6
TCACCATACTT






TGAAAGTGTAA






ACTACCACATA






TTAACATGTGT






GATTTAAGACC






CTCAG







881.
Mm.275648
Chromosome 13
TGTTGCCCTCA






GATATGTCAGA






TCAACTTGGAA






GGAAAGACCTT






CTACTCCAAGA






AGGAC







882.
Mm.254493
Chromosome 8
TCTAACAAGTG






TATTTGTGTTA






TCTTTAAAATA






GAACAATTGTA






TCTTGAAATGG






TAAAT







883.
Mm.27571
Chromosome 7
CGACACTGGGT






GGCCCTGCGAC






AGGTAGATGG






CATCTACTATA






ATCTGGACTCA






AAGCTC







884.
Mm.41033
Chromosome 2
TCTCAGAGGTG






TTGAAGATTTA






TCATCTTGAAT






CCTCCACAAAT






ACAGATACAGT






CCCAA







885.
Mm.3992
Chromosome 12
TCTTTTCACCT






CGATCAGCATC






ATGAGTCATCA






CAGATCATGTA






ATTAGTTTCTG






GGCCA







886.
Mm.221415
Chromosome 6
TGGGAATTGCA






TTTAGGATAGA






ATTGTATCTGA






TTTGCAAAATC






CATAAGCTCTC






ATGCC







887.
Mm.20437
Chromosome 4
TACTCCCACAG






TTGTATAGAAG






TCGAATAGTGA






AGGAGCTGGG






AGAAAACTGCT






TCAGCT







888.
Mm.27881
Chromosome 3
CCGCACTTAGC






CTAGCACCTTT






CTTACATGATC






TCAAGTTGAAC






CGACTTCCTTA






ACTCT







889.
Mm.29027
Chromosome 5
GCTTTGGAATT






AAAGAGGAGG






ATATAGATGAA






AACCCCCTCTT






TTGAATTAAGA






TTTGAG







890.
Mm.68617
Chromosome 1
AAATCAGATAT






GCAGGTCATCT






GATAAATGAGT






TAATGTTTGAT






ATTCGGGGTAT






CTCAC







891.
Mm.260361
No Chromosome location
GAACCATATGC





info available
TGGAATGAAA






CATAAGAGTTT






TCAACAGTTAT






CCTCTCACCTC






TGTATG







892.
Mm.7995
Chromosome X
GTATCGTCAAT






CCCAGTCAGTA






AGATAAGTTGA






AACAAGATTAT






CCTCAAGTGTA






GATTT







893.
Mm.130433
Chromosome 6
GTCAAAAACG






CCTTCAGGAAG






CCTTAGAGCGT






CAGAAGGAGT






TTGATCCGACC






ATAACAG







894.
Mm.196484
Chromosome 1
AATAGAATCTT






TTCACTTAGGA






ATGGAGAACA






AGCCAGTTCAG






AGGACCCCAA






AGTCTAG







895.
Mm.103615
Chromosome 4
CGTGGAGGAT






GGGCTAGCCTG






AGCTCTGGGAC






TAATCTTTATT






ACATACTTGTT






AATGAG







896.
Mm.24430
Chromosome 3
CTTATAGGGAG






AATGTTCTATT






CCTCAATCCAT






ACTCATTCCTA






CAGTATGCGCT






CTGGA







897.
Mm.33788
Chromosome 6
AGCAGGGGGA






TTATGTTAAGT






CAAATGCGTGT






GTCTCAAAAGT






GACATGTTTAA






CTGCTC







898.
Mm.4079
Chromosome 5
ACTCTGTACCC






TACTGGAACCA






CTCTGTAAAGA






GACAAAGCTGT






ATGTGCCACTT






CAGTA







899.
Mm.258618
Chromosome X
TTACAGGTCAC






TGTTTGTCACT






TTTGTGTACCA






GCTTCCCCATT






AGAATTCAACC






GATAC







900.
Mm.195900
Chromosome 13
ATGGAAGCGA






GGTCATTCTGC






GAACATTGGA






GATCTTTTATT






ACAAGTCTGCT






TGTTAAT







901.
Mm.271829
Chromosome 6
TAAAATTAGTG






TCCTGGGAGAG






ATGACCATTTT






AACTTCTATGC






TTATTTCACAT






GGGAA







902.
Mm.213265
Chromosome 14
TCGACGTCAAT






CTTACCTCTCT






AGGCAACATGT






TATCCCCGGAT






GATCAGAAATT






CCCAA







903.
Mm.17631
Chromosome 8
ACCTGTGTTTT






GTTTTTGTTTT






AAGAAACCAA






AGTGCACCAA






GATAGCATGCT






CTTGAGA







904.
Mm.20852
Chromosome 2
CTGCAGGTAAC






TCTCATTGGAA






GAAAAAGAAA






CTACAAGAGC






AAACAGAAGC






CATGGGAA







905.
Mm.87759
Chromosome Multiple
AAAGATTTCAT





Mappings
CCACGTCTGGC






GTAGTGGAAA






ACCCGAAGGG






AATATGTAATG






ATCTTTC







906.
Mm.242207
No Chromosome location
GTGTTGTACCC





info available
TAATTTGAATT






TAAAGTAGGC






AGTAGGTAGG






GTTAATTGGTA






GACTATC







907.
Mm.32556
Chromosome 17
CTTGGGTTTGA






GCACTCAGAAC






ACATGGCTGCA






ATCATCAAGAC






AGTTCACAGTT






AGCTT







908.
Mm.2074
Chromosome 2
CCCTAAGACAA






TGAAACTCAGA






ACTCTGTGATT






CCTGTGGAAAT






ATTTAAAACTG






AAATG







909.
Mm.268854
Chromosome 3
ATTTATAGAGG






TATCCTTAACA






TGCTGACTTCA






GTAACTGCCCT






TGTTTCTAAGG






AAGTC







910.
Mm.1483
Chromosome 16
ACCTGTAGCTT






CACTGTGAACT






TGTGGGCTTGG






CTGGTCTTAGG






AACTTGTACCT






ATAAA







911.
Mm.103615
Chromosome 4
TAATCCCTGGC






AAAGTCAAGA






CTGTGGGAAAC






TAGAACTGGTT






ACTCACTACTG






CTGGTA







912.
Mm.19738
Chromosome X
TTAGCTTCATG






ACCCCAAGGTT






AAGGTTCTGCC






AACAAGCATTC






TGCCTGACATC






TACTT







913.
Mm.276229
Chromosome 10
AATAAAGGCC






CCTTAGAAGCT






ACTGTAAAGCT






CTTCAAAGTTT






TCATGTAATCA






TAGGCA







914.
Mm.149029
Chromosome 16
AGAGATGGAG






ACTACACTGGG






TAGATTCTAGT






TTTTAGTTCTT






ATTAATGTGGG






GGAGTA







915.
Mm.268534
Chromosome 12
TATGGCCATTT






GGTTTCAGCAT






GTCAGGAGATT






TCTAATGATTT






GTGGCAATATC






AGCAA







916.
Mm.221782
Chromosome 19
TGTGTCAAGAT






AATCCTGAGTC






AACCTGGACAC






TTAATCCCTTT






GGACCTCTATC






TGGAG







917.
Mm.34527
Chromosome X
CCACCCATTAA






AATGACAGTAC






AAGTAGACCA






CAGTTTAAAGT






AGTTAGTCTAA






TTCTAC







918.
Mm.13445
Chromosome 15
CATAGTGGAA






ATATGCTCATC






TTTTATGCTAT






ATGTATTAAAC






CTCGACTTAGC






CCTGAA







919.
Mm.29236
Chromosome 7
GTTGAGGCTGA






CGACCTCCCAG






AGGCAATCTCT






GGATCTGGAAC






TTTGGGCATCA






TCGGA







920.
Mm.12454
Chromosome 1
ACCAACCAGG






GACTAGTTTGA






TGCTATCTTTG






CCTGTCTCTTG






GCTCTTAACAA






TGCCTA







921.
Mm.125975
Chromosome 7
CCAGGGAAGG






AACGATCCATT






CAGTGGTTTTA






AAATATCTCTT






CCTCAACAGAA






AAAGAT







922.
Mm.138073
Chromosome 2
GGTGCAAGCTA






GTACTCACACT






GTCACACCTTT






ACGCATGCGA






AAGGTAATGTG






CTAAAT







923.
Mm.140672
Chromosome 5
AGATCAGTGCT






CTGGACAGTAA






GATCCATGAGA






CGATTGAGTCC






ATAAACCAGCT






CAAGA







924.
Mm.218312
Chromosome 1
ATATCCCTGCT






AACTTAACAGC






AGTTAGTTTCC






TTGTTATGAAT






AAAAATGACA






GTCTGG







925.
Mm.260102
Chromosome 5
AAAGCAAATG






TTAGTAAAAAG






CTGGTGTGCAT






AGTCTTGTTAC






ATTGATGCAGT






TTTTCC







926.
Mm.3096
Chromosome 11
CAACTTGCTGA






ATAATGACTTC






CATTGAGTAAA






CATTTGGCTCT






GGTTATCTTCA






GGGAT







927.
Data not found
No Chromosome location
AGGAATTAGTA





info available
ACGTTTCATCC






AAGTAACCTTG






TTACAGTGAAC






AAGTGTCAAGT






GCTCA










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-Apoetm1Unc) 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-Apoetm1Unc), C57B1/6J, 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 2/3 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 (1-2 mm) 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 1, 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 22 k 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-Apoetm1Unc) (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, Fcerlg, 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 116, 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 Abca1 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 Res 91:9-16). 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, Il1Ora, 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, Col8a1, 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. Wisp1 is a secreted wnt-stimulated cysteine-rich protein that is a member of a family of factors with oncogenic and angiogenic activity. Rgs1 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 Hdac7 and 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) J Immunol 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 down-regulated 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 J Med (“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 2







Biological themes in atherosclerosis. Enrichment analysis of atherosclerosis-related genes


annotated with Gene Ontology and Biocarta terms demonstrates involvement of multiple


molecular pathways and biological processes. Probabilities (p-values) were derived


using Fisher exact test. 8478 of the entire microarray and 513 of genes in our set


(including additional 183 genes which demonstrated Pearson correlation >0.8


with the upregulated pattern) were annotated with GO, Biocarta, or other terms.











List gene #
Total gene #
p-value














Biological Process (GO annotation)





immune response
19
78
<0.0001


chemotaxis
10
23
<0.0001


cell surface receptor linked signal transduction
12
36
<0.0001


defense response
15
60
<0.0001


carbohydrate metabolism
14
67
<0.0001


antigen processing
5
9
<0.0001


locomotory behavior
4
6
<0.0001


inflammatory response
8
30
<0.0001


complement activation
5
12
<0.0001


proteolysis and peptidolysis
25
204
0.001


antigen presentation
4
10
0.002


intracellular signaling cascade
28
269
0.003


zinc ion homeostasis
2
2
0.004


transmembrane receptor protein tyrosine kinase activatic
2
2
0.004


hormone metabolism
2
2
0.004


hair cell differentiation
2
2
0.004


cell death
2
2
0.004


exogenous antigen via MHC class II
3
7
0.006


ossification
4
14
0.008


collagen catabolism
3
8
0.010


classical pathway
3
8
0.010


vesicle transport along actin filament
2
3
0.011


taurine transport
2
3
0.011


nitric oxide mediated signal transduction
2
3
0.011


negative regulation of angiogenesis
2
3
0.011


endogenous antigen via MHC class I
2
3
0.011


endogenous antigen
2
3
0.011


cellular defense response (sensu Vertebrata)
2
3
0.011


beta-alanine transport
2
3
0.011


lymph gland development
4
17
0.017


perception of pain
2
4
0.020


myeloid blood cell differentiation
2
4
0.020


female gamete generation
2
4
0.020


cytolysis
2
4
0.020


ATP biosynthesis
4
19
0.025


regulation of peptidyl-tyrosine phosphorylation
3
11
0.025


neurotransmitter transport
3
12
0.032


sex differentiation
2
5
0.032


exogenous antigen
2
5
0.032


cell adhesion
20
217
0.039


regulation of cell migration
3
13
0.040


wound healing
2
6
0.047


ureteric bud branching
2
6
0.047


cellular defense response
2
6
0.047


acute-phase response
2
6
0.047


regulation of transcription from Pol II promoter
6
44
0.048


hydrogen transport
3
14
0.049


calcium ion homeostasis
3
14
0.049


Molecular Functions (GO annotation)


acting on glycosyl bonds
12
31
<0.0001


interleukin receptor activity
8
13
<0.0001


hydrolase activity
67
641
<0.0001


cytokine activity
13
57
<0.0001


hematopoietin
9
32
<0.0001


complement activity
5
9
<0.0001


cytokine binding
3
3
<0.0001


C-C chemokine receptor activity
3
3
<0.0001


chemokine activity
4
7
<0.0001


cysteine-type endopeptidase activity
11
63
0.001


tumor necrosis factor receptor activity
3
5
0.002


platelet-derived growth factor receptor binding
2
2
0.004


cathepsin D activity
2
2
0.004


beta-N-acetylhexosaminidase activity
2
2
0.004


antimicrobial peptide activity
2
2
0.004


scavenger receptor activity
3
6
0.004


cysteine-type peptidase activity
9
56
0.006


mannosyl-oligosaccharide 1,2-alpha-mannosidase activi
3
7
0.006


receptor activity
42
479
0.009


taurine:sodium symporter activity
2
3
0.011


taurine transporter activity
2
3
0.011


myosin ATPase activity
2
3
0.011


MHC class I receptor activity
2
3
0.011


cathepsin B activity
2
3
0.011


calcium channel regulator activity
2
3
0.011


beta-alanine transporter activity
2
3
0.011


catalytic activity
23
230
0.012


solute:hydrogen antiporter activity
2
4
0.020


protein kinase C activity
2
4
0.020


tumor necrosis factor receptor binding
3
11
0.025


hydrogen-exporting ATPase activity
5
29
0.028


neurotransmitter:sodium symporter activity
2
5
0.032


MHC class II receptor activity
2
5
0.032


heparin binding
5
31
0.037


endopeptidase inhibitor activity
4
22
0.041


protein-tyrosine-phosphatase activity
7
54
0.043


hydrogen ion transporter activity
5
33
0.046


sulfuric ester hydrolase activity
2
6
0.047


Cellular Component (GO annotation)


extracellular space
139
1148
<0.0001


lysosome
26
66
<0.0001


extracellular
23
117
<0.0001


integral to membrane
138
1637
<0.0001


membrane
77
862
<0.0001


integral to plasma membrane
22
205
0.006


extracellular matrix
14
114
0.009


external side of plasma membrane
3
9
0.014


Biocarta Pathways


classicPathway
3
3
<0.0001


il22bppathway
4
7
<0.0001


nktPathway
5
12
<0.0001


Ccr5Pathway
5
13
0.001


reckPathway
4
8
0.001


compPathway
3
4
0.001


il7Pathway
4
10
0.002


TPOPathway
5
17
0.003


cxcr4Pathway
5
17
0.003


blymphocytePathway
2
2
0.004


il10Pathway
3
7
0.006


pdgfPathway
5
22
0.009


ionPathway
2
3
0.011


egfPathway
5
23
0.011


biopeptidesPathway
5
23
0.011


bcrPathway
5
25
0.015


ghPathway
4
17
0.017


fcer1Pathway
5
26
0.018


spryPathway
3
10
0.019


neutrophilPathway
2
4
0.020


mrpPathway
2
4
0.020


trkaPathway
3
11
0.025


pmlPathway
3
11
0.025


srcRPTPPathway
3
12
0.032


plcdPathway
2
5
0.032


ifngPathway
2
5
0.032


il2Pathway
3
13
0.040


RacCycDPathway
4
22
0.041


lymphocytePathway
2
6
0.047


nuclearRsPathway
3
14
0.049


cdMacPathway
3
14
0.049


CCR3Pathway
3
14
0.049


Summary annotation for inflammatory genes


defense
15
54
<0.0001


chemokine
9
22
<0.0001


Interleukin
9
38
<0.0001


cytokine
18
144
0.003


TNF
4
13
0.006


TH2
4
15
0.011


TH1
4
16
0.013


macrophage
3
13
0.040
















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-Apoetm1Unc) 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 2/3 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 Cdkn1a, 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 functions 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 Aoc1 (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 Tgfb4, 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), PPARα, 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 according to claim 1, wherein the isolated polynucleotide molecules are immobilized on an array.
  • 3. A system according to claim 2, 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.
  • 4. 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.
  • 5. 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.
  • 6. The method of claim 5, 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.
  • 7. The method of claim 5, comprising detecting the expression level of at least two of said genes.
  • 8. The method of claim 7, 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.
  • 9. The method of claim 5, comprising detecting the expression level of at least ten of said genes.
  • 10. The method of claim 9, 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.
  • 11. The method of claim 5, comprising detecting the expression level of at least one hundred of said genes.
  • 12. The method of claim 11, 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.
  • 13. The method of claim 5, wherein said atherosclerotic disease comprises coronary artery disease.
  • 14. The method of claim 5, wherein said atherosclerotic disease comprises carotid atherosclerosis.
  • 15. The method of claim 5, wherein said atherosclerotic disease comprises peripheral vascular disease.
  • 16. The method of claim 5, wherein said expression level is detected by measuring the RNA level expressed by said one or more genes.
  • 17. The method of claim 16, comprising isolating RNA from said individual prior to detecting the RNA expression level.
  • 18. The method of claim 16, 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.
  • 19. 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.
  • 20. The method of claim 19, 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.
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
Continuations (1)
Number Date Country
Parent 11387484 Mar 2006 US
Child 12205618 US