39 results on '"Luc J. Smink"'
Search Results
2. T1DBase, a community web-based resource for type 1 diabetes research.
- Author
-
Luc J. Smink, Erin M. Helton, Barry C. Healy, Christopher C. Cavnor, Alex C. Lam, Daisy Flamez, Oliver S. Burren, Yang Wang, Geoffrey E. Dolman, David B. Burdick, Vincent H. Everett, Gustavo Glusman, Davide Laneri, Lee Rowen, Helen Schuilenburg, Neil M. Walker, Josyf Mychaleckyj, Linda S. Wicker, Decio L. Eizirik, John A. Todd, and Nathan Goodman
- Published
- 2005
- Full Text
- View/download PDF
3. Genome bioinformatic analysis of nonsynonymous SNPs.
- Author
-
David F. Burke, Catherine L. Worth, Eva-Maria Priego, Tammy M. K. Cheng, Luc J. Smink, John A. Todd, and Tom L. Blundell
- Published
- 2007
- Full Text
- View/download PDF
4. Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity
- Author
-
John A. Todd, Barry C. Healy, Pau Serra, Dan Rainbow, Laurence B. Peterson, Pere Santamaria, Riitta Veijola, Jane Rogers, Linda S. Wicker, Rose M. Cubbon, Jan Clark, Andrea Gonzalez-Munoz, Show-Ling Chen, Jun Yamanouchi, Sarah Howlett, David V. Serreze, Valerie E S Garner, Paul A. Lyons, Ray Rosa, Anne Marie Cumiskey, Simon G. Gregory, Kara Hunter, and Luc J. Smink
- Subjects
CD4-Positive T-Lymphocytes ,Interleukin 2 ,Transcription, Genetic ,Regulatory T cell ,medicine.medical_treatment ,T cell ,Autoimmunity ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Article ,Mice ,Mice, Congenic ,Immune system ,Mice, Inbred NOD ,Genetics ,medicine ,Animals ,Homeostasis ,IL-2 receptor ,Alleles ,Autoimmune disease ,medicine.disease ,Diabetes Mellitus, Type 1 ,Cytokine ,medicine.anatomical_structure ,Immunology ,Interleukin-2 ,medicine.drug - Abstract
Autoimmune diseases are thought to result from imbalances in normal immune physiology and regulation. Here, we show that autoimmune disease susceptibility and resistance alleles on mouse chromosome 3 (Idd3) correlate with differential expression of the key immunoregulatory cytokine interleukin-2 (IL-2). In order to test directly that an approximately two-fold reduction in IL-2 underpins the Idd3-linked destabilization of immune homeostasis, we demonstrate that engineered haplodeficiency of IL-2 gene expression not only reduces T cell IL-2 production by two-fold but also mimics the autoimmune dysregulatory effects of the naturally-occurring susceptibility alleles of IL-2. Reduced IL-2 production achieved by both genetic mechanisms correlates with fewer and less functional CD4+CD25+ regulatory T cells, which are critical for maintaining immune homeostasis.
- Published
- 2007
- Full Text
- View/download PDF
5. Genetic and functional association of the immune signaling molecule 4-1BB (CD137/TNFRSF9) with type 1 diabetes
- Author
-
John A. Todd, Linda S. Wicker, Laurence B. Peterson, Tania H. Watts, Joanna M. M. Howson, Jennifer L. Cannons, Luc J. Smink, and Giselle Chamberlain
- Subjects
T cell ,Immunology ,Congenic ,Receptors, Nerve Growth Factor ,Nod ,Biology ,medicine.disease_cause ,Receptors, Tumor Necrosis Factor ,Cell Line ,Autoimmunity ,Mice ,Mice, Congenic ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,Immune system ,Antigens, CD ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Immunology and Allergy ,NOD mice ,Autoimmune disease ,CD137 ,Physical Chromosome Mapping ,medicine.disease ,Molecular biology ,Mice, Inbred C57BL ,Diabetes Mellitus, Type 1 ,medicine.anatomical_structure ,Female ,Signal Transduction - Abstract
Idd9.3, a locus that determines susceptibility to the autoimmune disease type 1 diabetes (T1D) in the nonobese diabetic (NOD) mouse, has been mapped to the distal region of chromosome 4. In the current report we reduce the size of the Idd9.3 interval to 1.2 Mb containing 15 genes, including one encoding the immune signaling molecule, 4-1BB, which shows amino acid variation between diabetes sensitive and resistant strains. 4-1BB, a member of the TNF receptor superfamily expressed by a variety of immune cells, mediates growth and survival signals for T cells. Functional analyses demonstrate that purified T cells from NOD congenic mice with the C57BL/10 (B10) allele at Idd9.3 produce more IL-2 and proliferate more vigorously in response to anti-CD3 plus immobilized 4-1BB ligand than T cells from NOD mice with the NOD allele at Idd9.3. In contrast, the response to anti-CD3 plus anti-CD28 costimulation was indistinguishable between the congenic strains, pinpointing the differences in NOD versus NOD.B10 Idd9.3 T cell responses to the 4-1BB costimulatory pathway. These data provide evidence in support of Idd9.3 as the locus encoding 4-1BB and suggest that the 4-1BB signaling pathway could have a primary function in the etiology of autoimmune disease.
- Published
- 2005
- Full Text
- View/download PDF
6. No evidence for association of the TATA-box binding protein glutamine repeat sequence or the flanking chromosome 6q27 region with type 1 diabetes
- Author
-
Georgina Bowden, Deborah J. Smyth, Jennifer Masters, Kjersti S. Rønningen, Jason D. Cooper, Felicity Payne, Neil Walker, Dag E. Undlien, John A. Todd, Alex C. Lam, Constantin Ionescu-Tirgoviste, Lisa Godfrey, Luc J. Smink, Rebecca Pask, Rebecca C.J. Twells, Oliver S. Burren, Cristian Guja, Sarah Nutland, Jeffrey S. Szeszko, Helen E. Rance, and William Y.S. Wang
- Subjects
Repetitive Sequences, Amino Acid ,Genetics ,Polymorphism, Genetic ,Glutamine ,TATA-Box Binding Protein ,Biophysics ,Chromosome ,Locus (genetics) ,Cell Biology ,Biology ,Biochemistry ,Genome ,Molecular biology ,Single Nucleotide Polymorphism Map ,Exon ,Diabetes Mellitus, Type 1 ,Microsatellite Repeat ,Humans ,Chromosomes, Human, Pair 6 ,DNA, Intergenic ,Genetic Predisposition to Disease ,Molecular Biology ,Gene ,Microsatellite Repeats - Abstract
Susceptibility to the autoimmune disease type 1 diabetes has been linked to human chromosome 6q27 and, moreover, recently associated with one of the genes in the region, TATA box-binding protein (TBP). Using a much larger sample of T1D families than those studied by others, and by extensive re-sequencing of nine other genes in the proximity, in which we identified 279 polymorphisms, 83 of which were genotyped in up to 725 T1D multiplex and simplex families, we obtained no evidence for association of the TBP CAG/CAA (glutamine) microsatellite repeat sequence with disease, or for nine other genes, PDCD2, PSMB1, KIAA1838, DLL1, dJ894D12.4, FLJ25454, FLJ13162, FLJ11152, PHF10 and CCR6. This study also provides an exon-based tag single nucleotide polymorphism map for these 10 genes that can be used for analysis of other diseases.
- Published
- 2005
- Full Text
- View/download PDF
7. T1DBase, a community web-based resource for type 1 diabetes research
- Author
-
Gustavo Glusman, Vincent H. Everett, Alex C. Lam, Erin Helton, Josyf C. Mychaleckyj, Geoffrey E. Dolman, Nathan Goodman, Davide Laneri, Linda S. Wicker, Christopher C. Cavnor, John A. Todd, Barry C. Healy, Daisy Flamez, Oliver S. Burren, Lee Rowen, Neil Walker, David B. Burdick, Yang Wang, Helen Schuilenburg, Decio L. Eizirik, and Luc J. Smink
- Subjects
medicine.medical_specialty ,INDUCED GENES ,Biomedical Research ,DATABASE ,Gene Expression ,Genomics ,Context (language use) ,Genome browser ,Biology ,MOUSE ,Genome ,03 medical and health sciences ,Islets of Langerhans ,Mice ,User-Computer Interface ,0302 clinical medicine ,Molecular genetics ,Databases, Genetic ,Genetics ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,KEGG ,AUTOIMMUNE-DISEASE ,Gene ,030304 developmental biology ,0303 health sciences ,Internet ,IDENTIFICATION ,Biology and Life Sciences ,ASSOCIATION ,Genome project ,Articles ,NETWORKS ,3. Good health ,Rats ,GENOME ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,030220 oncology & carcinogenesis ,RAT ,PANCREATIC BETA-CELLS ,Database Management Systems - Abstract
T1DBase (http://T1DBase.org) is a public website and database that supports the type 1 diabetes (T1D) research community. The site is currently focused on the molecular genetics and biology of T1D susceptibility and pathogenesis. It includes the following datasets: annotated genome sequence for human, rat and mouse; information on genetically identified T1D susceptibility regions in human, rat and mouse, and genetic linkage and association studies pertaining to T1D; descriptions of NOD mouse congenic strains; the Beta Cell Gene Expression Bank, which reports expression levels of genes in beta cells under various conditions, and annotations of gene function in beta cells; data on gene expression in a variety of tissues and organs; and biological pathways from KEGG and BioCarta. Tools on the site include the GBrowse genome browser, site-wide context dependent search, Connect-the-Dots for connecting gene and other identifiers from multiple data sources, Cytoscape for visualizing and analyzing biological networks, and the GESTALT workbench for genome annotation. All data are open access and all software is open source.
- Published
- 2004
8. Association Analysis of the Lymphocyte-Specific Protein Tyrosine Kinase (LCK) Gene in Type 1 Diabetes
- Author
-
Dag E. Undlien, Constantin Ionescu-Tı̂irgovişte, David A. Savage, Cristian Guja, Bryan J. Barratt, Eva Tuomilehto-Wolf, Jason D. Cooper, Alex C. Lam, Christopher E. Lowe, John S. Hulme, Joanna M. M. Howson, John A. Todd, Rebecca C.J. Twells, Jaakko Tuomilehto, and Luc J. Smink
- Subjects
Adult ,Genetics ,Untranslated region ,dbSNP ,Genotype ,Endocrinology, Diabetes and Metabolism ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Diabetes Mellitus, Type 1 ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,Polymorphism (computer science) ,Internal Medicine ,Humans ,Genetic Predisposition to Disease ,Child ,Gene ,Allele frequency ,Tyrosine kinase ,Genetic association - Abstract
Prior data associating the expression of lymphocyte-specific protein tyrosine kinase (LCK) with type 1 diabetes, its critical function in lymphocytes, and the linkage of the region to diabetes in the nonobese diabetic (NOD) mouse model make LCK a premier candidate for a susceptibility gene. Resequencing of LCK in 32 individuals detected seven single nucleotide polymorphisms (SNPs) with allele frequencies >3%, including four common SNPs previously reported. These and six other SNPs from dbSNP were genotyped in a two-stage strategy using 2,430 families and were all shown not to be significantly associated with type 1 diabetes. We conclude that a major role for the common LCK polymorphisms in type 1 diabetes is unlikely. However, we cannot rule out the possibility of there being a causal variant outside the exonic, intronic, and untranslated regions studied.
- Published
- 2004
- Full Text
- View/download PDF
9. Fine Mapping, Gene Content, Comparative Sequencing, and Expression Analyses Support Ctla4 and Nramp1 as Candidates for Idd5.1 and Idd5.2 in the Nonobese Diabetic Mouse
- Author
-
John A. Todd, Kara Hunter, Anne Marie Cumiskey, Giselle Chamberlain, Jan Clark, Laurence B. Peterson, Amanda Kingsnorth, Jane Rogers, Luc J. Smink, Joanna M. M. Howson, Linda S. Wicker, Simon G. Gregory, Ray Rosa, Andrea Gonzalez-Munoz, Dan Rainbow, Paul A. Lyons, Paul G. Tiffen, and Sarah Howlett
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Candidate gene ,Molecular Sequence Data ,Immunology ,Congenic ,Single-nucleotide polymorphism ,Biology ,Inducible T-Cell Co-Stimulator Protein ,Mice ,Exon ,Antigens, CD ,Mice, Inbred NOD ,Genetic variation ,Animals ,Humans ,Immunology and Allergy ,SNP ,Coding region ,CTLA-4 Antigen ,Amino Acid Sequence ,RNA, Messenger ,Cation Transport Proteins ,Gene ,Genetics ,Chromosome Mapping ,Antigens, Differentiation ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,Chromosomes, Human, Pair 2 - Abstract
At least two loci that determine susceptibility to type 1 diabetes in the NOD mouse have been mapped to chromosome 1, Idd5.1 (insulin-dependent diabetes 5.1) and Idd5.2. In this study, using a series of novel NOD.B10 congenic strains, Idd5.1 has been defined to a 2.1-Mb region containing only four genes, Ctla4, Icos, Als2cr19, and Nrp2 (neuropilin-2), thereby excluding a major candidate gene, Cd28. Genomic sequence comparison of the two functional candidate genes, Ctla4 and Icos, from the B6 (resistant at Idd5.1) and the NOD (susceptible at Idd5.1) strains revealed 62 single nucleotide polymorphisms (SNPs), only two of which were in coding regions. One of these coding SNPs, base 77 of Ctla4 exon 2, is a synonymous SNP and has been correlated previously with type 1 diabetes susceptibility and differential expression of a CTLA-4 isoform. Additional expression studies in this work support the hypothesis that this SNP in exon 2 is the genetic variation causing the biological effects of Idd5.1. Analysis of additional congenic strains has also localized Idd5.2 to a small region (1.52 Mb) of chromosome 1, but in contrast to the Idd5.1 interval, Idd5.2 contains at least 45 genes. Notably, the Idd5.2 region still includes the functionally polymorphic Nramp1 gene. Future experiments to test the identity of Idd5.1 and Idd5.2 as Ctla4 and Nramp1, respectively, can now be justified using approaches to specifically alter or mimic the candidate causative SNPs.
- Published
- 2004
- Full Text
- View/download PDF
10. Haplotype Tag Single Nucleotide Polymorphism Analysis of the Human Orthologues of the Rat Type 1 Diabetes GenesIan4(Lyp/Iddm1) andCblb
- Author
-
Bryan J. Barratt, Jason D. Cooper, Luc J. Smink, Alex C. Lam, John A. Todd, Helen E. Rance, Sarah Nutland, Neil Walker, Rebecca C.J. Twells, Rebecca Pask, Felicity Payne, and Deborah J. Smyth
- Subjects
Genotype ,Ubiquitin-Protein Ligases ,Endocrinology, Diabetes and Metabolism ,Nonsense mutation ,Single-nucleotide polymorphism ,Biology ,Polymerase Chain Reaction ,Polymorphism, Single Nucleotide ,Linkage Disequilibrium ,Frameshift mutation ,Species Specificity ,Internal Medicine ,Animals ,Humans ,Gene family ,Family ,Genetic Predisposition to Disease ,Rats, Inbred BB ,Proto-Oncogene Proteins c-cbl ,Frameshift Mutation ,Gene ,Adaptor Proteins, Signal Transducing ,Genetics ,Siblings ,Haplotype ,Rats ,Diabetes Mellitus, Type 1 ,Codon, Nonsense ,CBLB ,TCF7L2 - Abstract
The diabetes-prone BioBreeding (BB) and Komeda diabetes-prone (KDP) rats are both spontaneous animal models of human autoimmune, T-cell-associated type 1 diabetes. Both resemble the human disease, and consequently, susceptibility genes for diabetes found in these two strains can be considered as potential candidate genes in humans. Recently, a frameshift deletion in Ian4, a member of the immune-associated nucleotide (Ian)-related gene family, has been shown to map to BB rat Iddm1. In the KDP rat, a nonsense mutation in the T-cell regulatory gene, Cblb, has been described as a major susceptibility locus. Following a strategy of examining the human orthologues of susceptibility genes identified in animal models for association with type 1 diabetes, we identified single nucleotide polymorphisms (SNPs) from each gene by resequencing PCR product from at least 32 type 1 diabetic patients. Haplotype tag SNPs (htSNPs) were selected and genotyped in 754 affected sib-pair families from the U.K. and U.S. Evaluation of disease association by a multilocus transmission/disequilibrium test (TDT) gave a P value of 0.484 for IAN4L1 and 0.692 for CBLB, suggesting that neither gene influences susceptibility to common alleles of human type 1 diabetes in these populations.
- Published
- 2004
- Full Text
- View/download PDF
11. Testing the possible negative association of type 1 diabetes and atopic disease by analysis of the interleukin 4 receptor gene
- Author
-
Lisa M. Maier, E Tuomilehto-Wolf, Luc J. Smink, Joanna M. M. Howson, John A. Todd, Sarah Nutland, Jaakko Tuomilehto, Dag E. Undlien, Constantin Ionescu-Tirgoviste, David Clayton, Alex C. Lam, Cristian Guja, David A. Savage, Francesco Cucca, Helen E. Rance, Oliver S. Burren, Kjersti S. Rønningen, Deborah J. Smyth, Christopher Patterson, Neil Walker, Dennis Carson, and Rebecca C.J. Twells
- Subjects
endocrine system ,Genotype ,endocrine system diseases ,Genetic Linkage ,Immunology ,Locus (genetics) ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,White People ,Atopy ,Gene Frequency ,HLA Antigens ,immune system diseases ,Interleukin-4 receptor ,Genetics ,medicine ,Humans ,SNP ,Genetic Predisposition to Disease ,Allele ,Promoter Regions, Genetic ,Allele frequency ,Alleles ,Genetics (clinical) ,Haplotype ,Genetic Variation ,nutritional and metabolic diseases ,Exons ,medicine.disease ,Asthma ,Receptors, Interleukin-4 ,Diabetes Mellitus, Type 1 ,Logistic Models ,Haplotypes ,Chromosomes, Human, Pair 16 - Abstract
Variations in the interleukin 4 receptor A (IL4RA) gene have been reported to be associated with atopy, asthma, and allergy, which may occur less frequently in subjects with type 1 diabetes (T1D). Since atopy shows a humoral immune reactivity pattern, and T1D results from a cellular (T lymphocyte) response, we hypothesised that alleles predisposing to atopy could be protective for T1D and transmitted less often than the expected 50% from heterozygous parents to offspring with T1D. We genotyped seven exonic single nucleotide polymorphisms (SNPs) and the -3223 C>T SNP in the putative promoter region of IL4RA in up to 3475 T1D families, including 1244 Finnish T1D families. Only the -3223 C>T SNP showed evidence of negative association (P=0.014). There was some evidence for an interaction between -3233 C>T and the T1D locus IDDM2 in the insulin gene region (P=0.001 in the combined and P=0.02 in the Finnish data set). We, therefore, cannot rule out a genetic effect of IL4RA in T1D, but it is not a major one.
- Published
- 2003
- Full Text
- View/download PDF
12. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease
- Author
-
Neil Walker, Constantin Ionescu-Tirgoviste, Kathleen M Gillespie, Cristian Guja, David A. Savage, Jane Rogers, Eva Tuomilehto-Wolf, Christopher Patterson, Dag E. Undlien, Laurence B. Peterson, H Ueda, Michael L. Metzker, Francesco Cucca, Jaakko Tuomilehto, C Bordin, R Nithiyananthan, Hywel Snook, Amit Allahabadia, Simon G. Gregory, Helen E. Rance, Jayne A. Franklyn, John A. Todd, A C Lam, F Payne, John S. Hulme, Laura Esposito, Polly J. Bingley, Anne Smith, Giselle Chamberlain, Dennis Carson, Gough Scl., Kjersti S. Rønningen, Deborah J. Smyth, G Di Genova, Barry C. Healy, Alexander P. Maxwell, Joanne M. Heward, Sarah Nutland, Howson Jmm., Christopher E. Lowe, Twells Rcj., Hunter Kmd., Sarah Howlett, Heather J. Cordell, Ingrid Dahlman, J F Hess, David Clayton, Linda S. Wicker, Mathias H. Herr, Vincent H. Everett, Costantino Motzo, Dan Rainbow, and Luc J. Smink
- Subjects
Immunoconjugates ,Genotype ,T-Lymphocytes ,T cell ,Population ,chemical and pharmacologic phenomena ,Biology ,Polymorphism, Single Nucleotide ,Autoimmune Diseases ,Abatacept ,PTPN22 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Hypothyroidism ,Gene mapping ,Antigens, CD ,medicine ,Animals ,Humans ,Protein Isoforms ,CTLA-4 Antigen ,Genetic Predisposition to Disease ,Allele ,General ,education ,Gene ,030304 developmental biology ,Regulator gene ,Genetics ,Autoimmune disease ,0303 health sciences ,education.field_of_study ,Multidisciplinary ,Base Sequence ,medicine.disease ,Antigens, Differentiation ,Graves Disease ,3. Good health ,Alternative Splicing ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,medicine.anatomical_structure ,Immunology ,030215 immunology - Abstract
Genes and mechanisms involved in common complex diseases, such as the autoimmune disorders that affect approximately 5% of the population, remain obscure. Here we identify polymorphisms of the cytotoxic T lymphocyte antigen 4 gene (CTLA4)--which encodes a vital negative regulatory molecule of the immune system--as candidates for primary determinants of risk of the common autoimmune disorders Graves' disease, autoimmune hypothyroidism and type 1 diabetes. In humans, disease susceptibility was mapped to a non-coding 6.1 kb 3' region of CTLA4, the common allelic variation of which was correlated with lower messenger RNA levels of the soluble alternative splice form of CTLA4. In the mouse model of type 1 diabetes, susceptibility was also associated with variation in CTLA-4 gene splicing with reduced production of a splice form encoding a molecule lacking the CD80/CD86 ligand-binding domain. Genetic mapping of variants conferring a small disease risk can identify pathways in complex disorders, as exemplified by our discovery of inherited, quantitative alterations of CTLA4 contributing to autoimmune tissue destruction.
- Published
- 2003
- Full Text
- View/download PDF
13. Reevaluating Human Gene Annotation: A Second-Generation Analysis of Chromosome 22
- Author
-
Sarah C. L. Knowles, Luc J. Smink, David Beare, Charlotte G. Cole, Ian Dunham, Jacqueline M. Bye, Melanie E. Goward, Elizabeth J. Huckle, and John E. Collins
- Subjects
Genetics ,Sequence analysis ,Chromosomes, Human, Pair 22 ,Pseudogene ,Molecular Sequence Data ,Chromosome Mapping ,Articles ,Gene Annotation ,Genome project ,Vertebrate and Genome Annotation Project ,Biology ,Mice ,Annotation ,Genes ,Chromosome 19 ,Animals ,Humans ,Human genome ,Genetics (clinical) - Abstract
We report a second-generation gene annotation of human chromosome 22. Using expressed sequence databases, comparative sequence analysis, and experimental verification, we have extended genes, fused previously fragmented structures, and identified new genes. The total length in exons of annotation was increased by 74% over our previously published annotation and includes 546 protein-coding genes and 234 pseudogenes. Thirty-two potential protein-coding annotations are partial copies of other genes, and may represent duplications on an evolutionary path to change or loss of function. We also identified 31 non-protein-coding transcripts, including 16 possible antisense RNAs. By extrapolation, we estimate the human genome contains 29,000–36,000 protein-coding genes, 21,300 pseudogenes, and 1500 antisense RNAs. We suggest that our revised annotation criteria provide a paradigm for future annotation of the human genome.[Supplemental material is available online at www.genome.org. The sequence data from this study have been submitted to GenBank under accession nos. AL009266, AL021682-3,AL021708, AL022729, AL035081-2, AL035364, AL035366, AL035545, AL049654,AL050253-8, AL050345-6, AL079310, AL096779-81, AL096879-81, AL096883,AL096886, AL138578, AL157851, AL159142-3, AL160111-2, AL160131-2,AL160311, AL355092, AL355192, AL355841, AL359401, AL359403, AL365511-5,AL442116, AL449243, AL449244, AL450314, AL589866-7, AL590120,AL590887-8, BU583989–BU585359. The following individuals kindly provided reagents, samples, or unpublished information as indicated in the paper: J. Seilhamer, L. Stuve, H. Roest-Crollius, A. Levine, G. Slater, and J. Kent.]
- Published
- 2002
- Full Text
- View/download PDF
14. A SNP Resource for Human Chromosome 22: Extracting Dense Clusters of SNPs From the Genomic Sequence
- Author
-
Kate Rice, Simon Livingston, David Bentley, Kazuhiko Kawasaki, Sarah E. Hunt, Luc J. Smink, Shinsei Minoshima, Bruce A. Roe, Rocky Ganske, Elisabeth Dawson, Mark Raymond Adams, Suzannah Bumpstead, Richard Bruskiewich, Yuan Chen, Adrienne Hunt, Ian Dunham, Pak C. Sham, and Nobuyoshi Shimizu
- Subjects
Genetics ,Chromosome 7 (human) ,Base Composition ,Genome, Human ,Chromosomes, Human, Pair 22 ,Chromosome Mapping ,Genetic Variation ,Reproducibility of Results ,Genome project ,Biology ,Polymorphism, Single Nucleotide ,Resources ,SNP genotyping ,Structural variation ,Chromosome 17 (human) ,Chromosome 19 ,DNA Transposable Elements ,Humans ,Chromosome Deletion ,Chromosome 21 ,Chromosome 22 ,Genetics (clinical) - Abstract
The recent publication of the complete sequence of human chromosome 22 provides a platform from which to investigate genomic sequence variation. We report the identification and characterization of 12,267 potential variants (SNPs and other small insertions/deletions) of human chromosome 22, discovered in the overlaps of 460 clones used for the chromosome sequencing. We found, on average, 1 potential variant every 1.07 kb and approximately 18% of the potential variants involve insertions/deletions. The SNPs have been positioned both relative to each other, and to genes, predicted genes, repeat sequences, other genetic markers, and the 2730 SNPs previously identified on the chromosome. A subset of the SNPs were verified experimentally using either PCR–RFLP or genomic Invader assays. These experiments confirmed 92% of the potential variants in a panel of 92 individuals. [Details of the SNPs and RFLP assays can be found at http://www.sanger.ac.uk and in dbSNP.]
- Published
- 2001
- Full Text
- View/download PDF
15. Assignment of the βB1 Crystallin Gene (CRYBB1) to Human Chromosome 22 and Mouse Chromosome 5
- Author
-
Luc J. Smink, Andries Westerveld, Gilles Thomas, Ian Dunham, Theo J. M. Hulsebos, Debra J. Gilbert, Neal G. Copeland, Olivier Delattre, Nancy A. Jenkins, and Other departments
- Subjects
Chromosomes, Human, Pair 22 ,Molecular Sequence Data ,Hybrid Cells ,Biology ,Mice ,Exon ,Species Specificity ,Crystallin ,Sequence Homology, Nucleic Acid ,Genetics ,Animals ,Humans ,Beta (finance) ,Gene ,Gene Library ,Base Sequence ,Somatic Cell Hybrids ,Genetic Complementation Test ,Chromosome Mapping ,Chromosome ,Exons ,Cosmids ,Crystallins ,Molecular biology ,Rats ,Beta-Crystallin ,Cattle ,Chromosome 22 - Abstract
By using primers complementary to the rat beta B1 crystallin gene sequence, we amplified exons 5 and 6 of the orthologous human gene (CRYBB1). The amplified human segments displayed greater than 88% sequence homology to the corresponding rat and bovine sequences. CRYBB1 was assigned to the group 5 region in 22q11.2-q12.1 by hybridizing the exon 6 PCR product to somatic cell hybrids containing defined portions of human chromosome 22. The exon 5 and exon 6 PCR products of CRYBB1 were used to localize, by interspecific backcross mapping, the mouse gene (Crybb1) to the central portion of chromosome 5. Three other beta crystallin genes (beta B2(-1), beta B3, and beta A4) have previously been mapped to the same regions in human and mouse. We demonstrate that the beta B1 and beta A4 crystallin genes are very closely linked in the two species. These assignments complete the mapping and identification of the human and mouse homologues of the major beta crystallins genes that are expressed in the bovine lens.
- Published
- 1995
- Full Text
- View/download PDF
16. NOD congenic strain analysis of autoimmune diabetes reveals genetic complexity of the Idd18 locus and identifies Vav3 as a candidate gene
- Author
-
Calliope A. Dendrou, Heather I. Fraser, Barry C. Healy, Laurence B. Peterson, Daniel B. Rainbow, Simon G. Gregory, Luc J. Smink, Linda S. Wicker, Sarah Howlett, Charles A. Steward, and John A. Todd
- Subjects
Candidate gene ,Immunology ,Congenic ,Single-nucleotide polymorphism ,Locus (genetics) ,Nerve Tissue Proteins ,Biology ,Article ,Exon ,Mice ,Mice, Congenic ,Gene mapping ,Mice, Inbred NOD ,Insulin-Secreting Cells ,Immunology and Allergy ,Animals ,Insulin ,Genetic Predisposition to Disease ,Proto-Oncogene Proteins c-vav ,Gene ,Alleles ,Crosses, Genetic ,NOD mice ,Genetics ,Exons ,Physical Chromosome Mapping ,Molecular biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,Genetic Loci ,Female ,Netrins - Abstract
We have used the public sequencing and annotation of the mouse genome to delimit the previously resolved type 1 diabetes (T1D) insulin-dependent diabetes (Idd)18 interval to a region on chromosome 3 that includes the immunologically relevant candidate gene, Vav3. To test the candidacy of Vav3, we developed a novel congenic strain that enabled the resolution of Idd18 to a 604-kb interval, designated Idd18.1, which contains only two annotated genes: the complete sequence of Vav3 and the last exon of the gene encoding NETRIN G1, Ntng1. Targeted sequencing of Idd18.1 in the NOD mouse strain revealed that allelic variation between NOD and C57BL/6J (B6) occurs in noncoding regions with 138 single nucleotide polymorphisms concentrated in the introns between exons 20 and 27 and immediately after the 3′ untranslated region. We observed differential expression of VAV3 RNA transcripts in thymocytes when comparing congenic mouse strains with B6 or NOD alleles at Idd18.1. The T1D protection associated with B6 alleles of Idd18.1/Vav3 requires the presence of B6 protective alleles at Idd3, which are correlated with increased IL-2 production and regulatory T cell function. In the absence of B6 protective alleles at Idd3, we detected a second T1D protective B6 locus, Idd18.3, which is closely linked to, but distinct from, Idd18.1. Therefore, genetic mapping, sequencing, and gene expression evidence indicate that alteration of VAV3 expression is an etiological factor in the development of autoimmune β-cell destruction in NOD mice. This study also demonstrates that a congenic strain mapping approach can isolate closely linked susceptibility genes.
- Published
- 2010
17. Natural Genetic Variants Influencing Type 1 Diabetes in Humans and in the NOD Mouse
- Author
-
Joanna M. M. Howson, Linda S. Wicker, Laurence B. Peterson, Luc J. Smink, Andrea Gonzalez-Munoz, Anne Marie Cumiskey, Barry C. Healy, Dan Rainbow, Simon G. Gregory, Valerie E S Garner, Amanda Kingsnorth, Carlos Penha-Gonçalves, Giselle Chamberlain, Carolyn Moule, Jan Clark, Kara Hunter, John A. Todd, Heather I. Fraser, Sarah Howlett, Paul A. Lyons, Jane Rogers, and Paul G. Tiffen
- Subjects
Genetics ,Type 1 diabetes ,Immune system ,medicine ,Human genome ,Disease ,Biology ,Central tolerance ,medicine.disease ,medicine.disease_cause ,Gene ,Phenotype ,Autoimmunity - Abstract
The understanding of the genetic basis of type 1 diabetes and other autoimmune diseases and the application of that knowledge to their treatment, cure and eventual prevention has been a difficult goal to reach. Cumulative progress in both mouse and human are finally giving way to some successes and significant insights have been made in the last few years. Investigators have identified key immune tolerance-associated phenotypes in convincingly reliable ways that are regulated by specific diabetes-associated chromosomal intervals. The combination of positional genetics and functional studies is a powerful approach to the identification of downstream molecular events that are causal in disease aetiology. In the case of type 1 diabetes, the availability of several animal models, especially the NOD mouse, has complemented the efforts to localize human genes causing diabetes and has shown that some of the same genes and pathways are associated with autoimmunity in both species. There is also growing evidence that the initiation or progression of many autoimmune diseases is likely to be influenced by some of the same genes.
- Published
- 2008
- Full Text
- View/download PDF
18. Robust associations of four new chromosome regions from genome-wide analyses of type 1 diabetes
- Author
-
Jason P. Hafler, Sarah F. Field, David Clayton, Neil Walker, Ellen C. Adlem, Meeta Maisuria, Adrian Vella, Stephen C. L. Gough, Jennie H M Yang, Helen Schuilenburg, David B. Dunger, Jeffrey S. Szeszko, Sergey Nejentsev, Nigel R. Ovington, Linda S. Wicker, William Meadows, Deborah J. Smyth, Gillian Coleman, Barry C. Healy, Sarah Nutland, Matthew J. Simmonds, Kate Downes, John A. Todd, Alex C. Lam, Lauren R. Zeitels, H. T. Leung, Oliver S. Burren, Chris Wallace, Felicity Payne, Constantin Ionescu-Tirgoviste, Jason D. Cooper, Luc J. Smink, Helen Stevens, Rebecca Bailey, Joanna M. M. Howson, James E. Allen, Vincent Plagnol, Cristian Guja, Joanne M. Heward, and Christopher E. Lowe
- Subjects
Autoimmune disease ,Genetics ,Type 1 diabetes ,Adolescent ,Genome, Human ,Case-control study ,Chromosome Mapping ,Single-nucleotide polymorphism ,Locus (genetics) ,CLEC16A ,Disease ,Biology ,medicine.disease ,Polymorphism, Single Nucleotide ,Article ,Diabetes Mellitus, Type 1 ,Chromosome regions ,Case-Control Studies ,medicine ,Humans ,Genetic Predisposition to Disease - Abstract
The Wellcome Trust Case Control Consortium (WTCCC) primary genome-wide association (GWA) scan1 on seven diseases, including the multifactorial, autoimmune disease, type 1 diabetes (T1D), shows significant association (P < 5 × 10−7 between T1D and six chromosome regions: 12q24, 12q13, 16p13, 18p11, 12p13 and 4q27. Here, we attempted to validate these and six other top findings in 4,000 individuals with T1D, 5,000 controls and 2,997 family trios that were independent of the WTCCC study. We confirmed unequivocally the associations of 12q24, 12q13, 16p13 and 18p11 (Pfollow-up ≤ 1.35 × 10−9; Poverall ≤ 1.15 × 10−14), leaving eight regions with small effects or false-positive associations with T1D. We also obtained evidence for chromosome 18q22 (Poverall = 1.38 × 10−8) from a genome-wide association study of nonsynonymous SNPs. Several regions, including 18q22 and 18p11, showed association with autoimmune thyroid disease. This study increases the number of T1D loci with compelling evidence from six to at least ten.
- Published
- 2007
19. New tools for defining the 'genetic background' of inbred mouse strains
- Author
-
Barry C. Healy, William M. Ridgway, Luc J. Smink, Linda S. Wicker, and Dan Rainbow
- Subjects
Genetics ,Transgene ,Immunology ,Congenic ,food and beverages ,Genetic Variation ,Mice, Inbred Strains ,Biology ,Phenotype ,chemistry.chemical_compound ,Mice ,Immune system ,chemistry ,Species Specificity ,Genetic variation ,Knockout mouse ,Immunology and Allergy ,Animals ,Gene ,DNA - Abstract
There is general appreciation that 'genetic background effects' can profoundly affect the immune phenotypes of congenic, transgenic and knockout mice. We suggest that attributing phenotypes to genetic background effects is outmoded and that new databases containing single-nucleotide polymorphisms obtained with a group of inbred mouse strains can be used to define the flanking DNA of nearly all mouse genes.
- Published
- 2007
20. Interaction analysis of the CBLB and CTLA4 genes in type 1 diabetes
- Author
-
Neil Walker, Jayne Hutchings, Helen Stevens, Alex C. Lam, Sarah Nutland, John A. Todd, Jason D. Cooper, Luc J. Smink, and Felicity Payne
- Subjects
Offspring ,Denmark ,Immunology ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Polymorphism (computer science) ,Antigens, CD ,Genotype ,Immunology and Allergy ,SNP ,Animals ,Humans ,CTLA-4 Antigen ,Genetic Predisposition to Disease ,Genetic Testing ,Proto-Oncogene Proteins c-cbl ,Allele ,Gene ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,Genetics ,0303 health sciences ,Cell Biology ,Antigens, Differentiation ,Rats ,Minor allele frequency ,Diabetes Mellitus, Type 1 ,Case-Control Studies ,CBLB ,030217 neurology & neurosurgery - Abstract
Gene-gene interaction analyses have been suggested as a potential strategy to help identify common disease susceptibility genes. Recently, evidence of a statistical interaction between polymorphisms in two negative immunoregulatory genes, CBLB and CTLA4, has been reported in type 1 diabetes (T1D). This study, in 480 Danish families, reported an association between T1D and a synonymous coding SNP in exon 12 of the CBLB gene (rs3772534 G>A; minor allele frequency, MAF=0.24; derived relative risk, RR for G allele=1.78; P=0.046). Furthermore, evidence of a statistical interaction with the known T1D susceptibility-associated CTLA4 polymorphism rs3087243 (laboratory name CT60, G>A) was reported (P
- Published
- 2007
21. Genome bioinformatic analysis of nonsynonymous SNPs
- Author
-
John A. Todd, Catherine L. Worth, Luc J. Smink, David F. Burke, Eva-Maria Priego, Tammy M. K. Cheng, Tom L. Blundell, Burke, David [0000-0001-8830-3951], Blundell, Tom [0000-0002-2708-8992], and Apollo - University of Cambridge Repository
- Subjects
Sequence analysis ,PREDICTION ,DATABASE ,DNA Mutational Analysis ,Single-nucleotide polymorphism ,BINDING SURFACES ,Biology ,STRUCTURE HOMOLOGY RECOGNITION ,AMINO-ACID SUBSTITUTION ,lcsh:Computer applications to medicine. Medical informatics ,Biochemistry ,Genome ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,Structural Biology ,Humans ,Molecular Biology ,Gene ,lcsh:QH301-705.5 ,030304 developmental biology ,Genetic association ,Genetics ,0303 health sciences ,Genome, Human ,Applied Mathematics ,Methodology Article ,030302 biochemistry & molecular biology ,LARGE-SCALE ,Chromosome Mapping ,Computational Biology ,Proteins ,Sequence Analysis, DNA ,Penetrance ,Computer Science Applications ,Minor allele frequency ,SUPPORT VECTOR MACHINES ,PROTEIN STABILITY CHANGES ,lcsh:Biology (General) ,NON-SYNONYMOUS SNPS ,SINGLE NUCLEOTIDE POLYMORPHISMS ,lcsh:R858-859.7 ,Human genome ,Algorithms - Abstract
Background Genome-wide association studies of common diseases for common, low penetrance causal variants are underway. A proportion of these will alter protein sequences, the most common of which is the non-synonymous single nucleotide polymorphism (nsSNP). It would be an advantage if the functional effects of an nsSNP on protein structure and function could be predicted, both for the final identification process of a causal variant in a disease-associated chromosome region, and in further functional analyses of the nsSNP and its disease-associated protein. Results In the present report we have compared and contrasted structure- and sequence-based methods of prediction to over 5500 genes carrying nearly 24,000 nsSNPs, by employing an automatic comparative modelling procedure to build models for the genes. The nsSNP information came from two sources, the OMIM database which are rare (minor allele frequency, MAF, < 0.01) and are known to cause penetrant, monogenic diseases. Secondly, nsSNP information came from dbSNP125, for which the vast majority of nsSNPs, mostly MAF > 0.05, have no known link to a disease. For over 40% of the nsSNPs, structure-based methods predicted which of these sequence changes are likely to either disrupt the structure of the protein or interfere with the function or interactions of the protein. For the remaining 60%, we generated sequence-based predictions. Conclusion We show that, in general, the prediction tools are able distinguish disease causing mutations from those mutations which are thought to have a neutral affect. We give examples of mutations in genes that are predicted to be deleterious and may have a role in disease. Contrary to previous reports, we also show that rare mutations are consistently predicted to be deleterious as often as commonly occurring nsSNPs.
- Published
- 2006
22. Discovery, linkage disequilibrium and association analyses of polymorphisms of the immune complement inhibitor, decay-accelerating factor gene (DAF/CD55) in type 1 diabetes
- Author
-
Deborah J. Smyth, Sarah Nutland, Alex C. Lam, Christopher E. Lowe, Jason D. Cooper, Barry C. Healy, Rebecca Bailey, Linda S. Wicker, Hidenori Taniguchi, John A. Todd, Luc J. Smink, Neil Walker, Oliver S. Burren, Burren, Oliver [0000-0002-3388-5760], and Apollo - University of Cambridge Repository
- Subjects
Linkage disequilibrium ,lcsh:QH426-470 ,ANTIGEN ,Single-nucleotide polymorphism ,DETERMINANTS ,SUSCEPTIBILITY ,Biology ,03 medical and health sciences ,Complement inhibitor ,LOCUS ,Genetics ,SNP ,International HapMap Project ,AUTOIMMUNE-DISEASE ,Decay-accelerating factor ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,CD46 ,Linkage Disequilibrium Mapping ,030305 genetics & heredity ,SINGLE-NUCLEOTIDE POLYMORPHISMS ,LYMPHOID TYROSINE PHOSPHATASE ,T-CELL IMMUNITY ,GENOME ,lcsh:Genetics ,CD55 ,Research Article - Abstract
BackgroundType 1 diabetes (T1D) is a common autoimmune disease resulting from T-cell mediated destruction of pancreatic beta cells. Decay accelerating factor (DAF, CD55), a glycosylphosphatidylinositol-anchored membrane protein, is a candidate for autoimmune disease susceptibility based on its role in restricting complement activation and evidence that DAF expression modulates the phenotype of mice models for autoimmune disease. In this study, we adopt a linkage disequilibrium (LD) mapping approach to test for an association between the DAF gene and T1D.ResultsInitially, we used HapMap II genotype data to examine LD across theDAFregion. Additional resequencing was required, identifying 16 novel polymorphisms. Combining both datasets, a LD mapping approach was adopted to test for association with T1D. Seven tag SNPs were selected and genotyped in case-control (3,523 cases and 3,817 controls) and family (725 families) collections.ConclusionWe obtained no evidence of association between T1D and theDAFregion in two independent collections. In addition, we assessed the impact of using only HapMap II genotypes for the selection of tag SNPs and, based on this study, found that HapMap II genotypes may require additional SNP discovery for comprehensive LD mapping of some genes in common disease.
- Published
- 2006
- Full Text
- View/download PDF
23. A genome-wide association study of nonsynonymous SNPs identifies a type 1 diabetes locus in the interferon-induced helicase (IFIH1) region
- Author
-
Luc J. Smink, Constantin Ionescu-Tirgoviste, Jason D. Cooper, Oliver S. Burren, Barry Widmer, Deborah J. Smyth, Neil Walker, David A. Savage, David B. Dunger, Cristian Guja, David Clayton, Sarah F. Field, John A. Todd, and Rebecca Bailey
- Subjects
Genetics ,IFIH1 Gene ,Interferon-Induced Helicase, IFIH1 ,Genotype ,Genome, Human ,Single-nucleotide polymorphism ,Locus (genetics) ,Genome-wide association study ,Biology ,Genome ,Polymorphism, Single Nucleotide ,Minor allele frequency ,DEAD-box RNA Helicases ,Diabetes Mellitus, Type 1 ,Chromosomes, Human, Pair 2 ,Humans ,Gene ,RNA Helicases - Abstract
In this study we report convincing statistical support for a sixth type 1 diabetes (T1D) locus in the innate immunity viral RNA receptor gene region IFIH1 (also known as mda-5 or Helicard) on chromosome 2q24.3. We found the association in an interim analysis of a genome-wide nonsynonymous SNP (nsSNP) scan, and we validated it in a case-control collection and replicated it in an independent family collection. In 4,253 cases, 5,842 controls and 2,134 parent-child trio genotypes, the risk ratio for the minor allele of the nsSNP rs1990760 A --G (A946T) was 0.86 (95% confidence interval = 0.82-0.90) at P = 1.42 x 10(-10).
- Published
- 2006
24. Type 1 diabetes genes and pathways shared by humans and NOD mice
- Author
-
Dan Rainbow, Laurence B. Peterson, Linda S. Wicker, Barry C. Healy, Kara Hunter, Jan Clark, John A. Todd, Andrea Gonzalez-Munoz, Valerie E S Garner, Luc J. Smink, Ray Rosa, Heather I. Fraser, and Sarah Howlett
- Subjects
Candidate gene ,Immunology ,Nod ,Protein tyrosine phosphatase ,Biology ,medicine.disease_cause ,Autoimmunity ,Mice ,Antigens, CD ,Mice, Inbred NOD ,medicine ,Immunology and Allergy ,Animals ,Humans ,CTLA-4 Antigen ,Genetic Predisposition to Disease ,Gene ,NOD mice ,Autoimmune disease ,Genetics ,Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,Protein Tyrosine Phosphatase, Non-Receptor Type 22 ,Receptors, Interleukin-2 ,medicine.disease ,Antigens, Differentiation ,Diabetes Mellitus, Type 1 ,Interleukin-2 ,Central tolerance ,Protein Tyrosine Phosphatases - Abstract
The identification of causative genes for the autoimmune disease type 1 diabetes (T1D) in humans and candidate genes in the NOD mouse has made significant progress in recent years. In addition to sharing structural aspects of the MHC class II molecules that confer susceptibility or resistance to T1D, genes and pathways contributing to autoimmune pathogenesis are held in common by the two species. There are data demonstrating a similar need to establish central tolerance to insulin. Gene variants for the interacting molecules IL2 and CD25, members of a pathway that is essential for immune homeostasis, are present in mice and humans, respectively. Variation of two molecules that negatively regulate T cells, CTLA-4 and the tyrosine phosphatase LYP/PEP, are associated with susceptibility to human and NOD T1D. These observations underscore the value of the NOD mouse model for mechanistic studies on human T1D-associated molecular and cellular pathways.
- Published
- 2005
25. Population structure, differential bias and genomic control in a large-scale, case-control association study
- Author
-
Rebecca Pask, Martin Moorhead, Lisa M. Maier, David Clayton, Joanna M. M. Howson, Paul Hardenbol, Deborah J. Smyth, Helen Stevens, John A. Todd, Malek Faham, Thomas D. Willis, Hywel B. Jones, Nigel R. Ovington, Alex C. Lam, Jason D. Cooper, Sarah Nutland, Neil Walker, Luc J. Smink, and Matthew Falkowski
- Subjects
Adolescent ,Genotype ,media_common.quotation_subject ,Population ,Genomics ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,Bias ,Statistics ,Genetics ,SNP ,Humans ,False Positive Reactions ,Lymphocytes ,education ,media_common ,Selection bias ,education.field_of_study ,Models, Genetic ,Confounding ,DNA ,United Kingdom ,Diabetes Mellitus, Type 1 ,Genetics, Population ,Causal inference ,Case-Control Studies - Abstract
The main problems in drawing causal inferences from epidemiological case-control studies are confounding by unmeasured extraneous factors, selection bias and differential misclassification of exposure. In genetics the first of these, in the form of population structure, has dominated recent debate. Population structure explained part of the significant +11.2% inflation of test statistics we observed in an analysis of 6,322 nonsynonymous SNPs in 816 cases of type 1 diabetes and 877 population-based controls from Great Britain. The remainder of the inflation resulted from differential bias in genotype scoring between case and control DNA samples, which originated from two laboratories, causing false-positive associations. To avoid excluding SNPs and losing valuable information, we extended the genomic control method by applying a variable downweighting to each SNP.
- Published
- 2005
26. Construction and analysis of tag single nucleotide polymorphism maps for six human-mouse orthologous candidate genes in type 1 diabetes
- Author
-
Lisa M, Maier, Deborah J, Smyth, Adrian, Vella, Felicity, Payne, Jason D, Cooper, Rebecca, Pask, Christopher, Lowe, John, Hulme, Luc J, Smink, Heather, Fraser, Carolyn, Moule, Kara M, Hunter, Giselle, Chamberlain, Neil, Walker, Sarah, Nutland, Dag E, Undlien, Kjersti S, Rønningen, Cristian, Guja, Constantin, Ionescu-Tîrgoviste, David A, Savage, David P, Strachan, Laurence B, Peterson, John A, Todd, Linda S, Wicker, Rebecca C, Twells, and Apollo - University of Cambridge Repository
- Subjects
Family Health ,Polymorphism, Genetic ,Genotype ,lcsh:QH426-470 ,Chromosome Mapping ,Sequence Analysis, DNA ,Polymorphism, Single Nucleotide ,White People ,Mice ,Open Reading Frames ,lcsh:Genetics ,Diabetes Mellitus, Type 1 ,Mice, Inbred NOD ,Untranslated Regions ,Case-Control Studies ,Animals ,Humans ,Genetic Predisposition to Disease ,Research Article - Abstract
Background One strategy to help identify susceptibility genes for complex, multifactorial diseases is to map disease loci in a representative animal model of the disorder. The nonobese diabetic (NOD) mouse is a model for human type 1 diabetes. Linkage and congenic strain analyses have identified several NOD mouse Idd (insulin dependent diabetes) loci, which have been mapped to small chromosome intervals, for which the orthologous regions in the human genome can be identified. Here, we have conducted re-sequencing and association analysis of six orthologous genes identified in NOD Idd loci: NRAMP1/SLC11A1 (orthologous to Nramp1/Slc11a1 in Idd5.2), FRAP1 (orthologous to Frap1 in Idd9.2), 4-1BB/CD137/TNFRSF9 (orthologous to 4-1bb/Cd137/Tnrfrsf9 in Idd9.3), CD101/IGSF2 (orthologous to Cd101/Igsf2 in Idd10), B2M (orthologous to B2m in Idd13) and VAV3 (orthologous to Vav3 in Idd18). Results Re-sequencing of a total of 110 kb of DNA from 32 or 96 type 1 diabetes cases yielded 220 single nucleotide polymorphisms (SNPs). Sixty-five SNPs, including 54 informative tag SNPs, and a microsatellite were selected and genotyped in up to 1,632 type 1 diabetes families and 1,709 cases and 1,829 controls. Conclusion None of the candidate regions showed evidence of association with type 1 diabetes (P values > 0.2), indicating that common variation in these key candidate genes does not play a major role in type 1 diabetes susceptibility in the European ancestry populations studied.
- Published
- 2005
27. Identification of a structurally distinct CD101 molecule encoded in the 950-kb Idd10 region of NOD mice
- Author
-
Christopher J. Lord, Carlos Penha-Gonçalves, Laurence B. Peterson, Paul A. Lyons, Simon G. Gregory, John A. Todd, Joseph J. Suttie, Linda S. Wicker, Jane Rogers, Carolyn Moule, Joanna M. M. Howson, and Luc J. Smink
- Subjects
Genetic Markers ,Subfamily ,Endocrinology, Diabetes and Metabolism ,Pseudogene ,Molecular Sequence Data ,Congenic ,Nod ,Biology ,Polymorphism, Single Nucleotide ,Mice ,Antigens, CD ,Mice, Inbred NOD ,Internal Medicine ,Animals ,Genetic Predisposition to Disease ,Amino Acid Sequence ,Gene ,NOD mice ,Genetics ,Membrane Glycoproteins ,Contig ,Base Sequence ,Chromosome Mapping ,Genetic Variation ,Exons ,Diabetes Mellitus, Type 1 ,Chromosome 3 ,Amino Acid Substitution ,Pseudogenes - Abstract
Genes affecting autoimmune type 1 diabetes susceptibility in the nonobese diabetic (NOD) mouse (Idd loci) have been mapped using a congenic strain breeding strategy. In the present study, we used a combination of BAC clone contig construction, polymorphism analysis of DNA from congenic strains, and sequence mining of the human orthologous region to generate an integrated map of the Idd10 region on mouse chromosome 3. We found seven genes and one pseudogene in the 950-kb Idd10 region. Although all seven genes in the interval are Idd10 candidates, we suggest the gene encoding the EWI immunoglobulin subfamily member EWI-101 (Cd101) as the most likely Idd10 candidate because of the previously reported immune-associated properties of the human CD101 molecule. Additional support for the candidacy of Cd101 is the presence of 17 exonic single-neucleotide polymorphisms that differ between the NOD and B6 sequences, 10 causing amino acid substitutions in the predicted CD101 protein. Four of these 10 substitutions are nonconservative, 2 of which could potentially alter N-linked glycosylation. Considering our results together with those previous reports that antibodies recognizing human CD101 modulate human T-cell and dendritic cell function, there is now justification to test whether the alteration of CD101 function affects autoimmune islet destruction.
- Published
- 2003
28. The physical maps for sequencing human chromosomes 1, 6, 9, 10, 13, 20 and X
- Author
-
Nigel P. Carter, Ian Dunham, A. L. Atkinson, Jaime Hughes, M. Izmajlowicz, Richard Durbin, Mark T. Ross, Soumi Joseph, Adam Butler, G. L. Harper, Louise McDonald, Rohan Taylor, W. D. Burrill, Graeme Bethel, S. R. Prathalingam, David R. Bentley, Theologia Sarafidou, Vassos Neocleous, John Sulston, F. L. Dearden, C. M. Rice, E. C. Sotheran, Carol A. Edwards, S. M. Clegg, N. Brady, T. E. Wilmer, B. L. Hopkins, G. L. Maslen, Simon G. Gregory, Owen T. McCann, Andrew J. Mungall, M.A. Leversha, Elisabeth Dawson, K. J. Phillips, R Evans, A. A. Thorpe, C M Clee, Cordelia Langford, E. J. Huckle, Helen E. Steingruber, Carol Scott, Y. H. Ramsey, John E. Collins, Gareth R. Howell, Adam Whittaker, M. E. Earthrowl, M. H. Lehvaslaiho, Pamela Whittaker, G. Laird, J. C. Brook, D. J. Scott, K. J. Ashcroft, S. H. Williams, Georgina Warry, Nicholas K. Moschonas, D. M. Pearson, K. S. Halls, C. L. Wright, R. W. Heathcott, C. Carder, Jane L. Holden, D. C. Burford, S. A. Ranby, P. J. Howard, K. Aubin, K. M. Porter, E. Holloway, R. A. Cooper, A. J. Coffey, David Beare, J. J. Catanese, C. A. Jones, J. S. Conquer, J. Ghori, E. J. Tinsley, Lisa French, Charlotte G. Cole, P. D. Dhami, K. M. Culley, Christopher J. Gillson, Sarah E. Hunt, Rhian Gwilliam, Panagiotis Deloukas, V. Cobley, Carol Soderlund, A. Taylor, G. J. Sharp, Luc J. Smink, S. Hammond, Andrew Dunham, L. J. Rogers, D. Mistry, Richard Wooster, P. J. De Jong, Jane Rogers, P. J. Hunt, L D Green, C. J. Shaw-Smith, Jennifer McDowall, C. Burrows, and Sean Humphray
- Subjects
Genetics ,Multidisciplinary ,X Chromosome ,Contig ,Gene map ,Chromosomes, Human, Pair 13 ,Chromosomes, Human, Pair 10 ,Genome, Human ,Chromosomes, Human, Pair 20 ,food and beverages ,Chromosome ,Computational biology ,Biology ,Genome ,Contig Mapping ,Gene mapping ,Chromosomes, Human, Pair 1 ,Humans ,Chromosomes, Human, Pair 6 ,X chromosome ,Genomic organization - Abstract
We constructed maps for eight chromosomes (1, 6, 9, 10, 13, 20, X and (previously) 22), representing one-third of the genome, by building landmark maps, isolating bacterial clones and assembling contigs. By this approach, we could establish the long-range organization of the maps early in the project, and all contig extension, gap closure and problem-solving was simplified by containment within local regions. The maps currently represent more than 94% of the euchromatic (gene-containing) regions of these chromosomes in 176 contigs, and contain 96% of the chromosome-specific markers in the human gene map. By measuring the remaining gaps, we can assess chromosome length and coverage in sequenced clones.
- Published
- 2001
29. Mechanism of spreading of the highly related neurofibromatosis type 1 (NF1) pseudogenes on chromosomes 2, 14 and 22
- Author
-
Ian Dunham, Bruce A. Roe, Theo J. M. Hulsebos, Blaine T Smith, Yingping Wang, M. Luijten, Luc J. Smink, Andries Westerveld, and Other departments
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Sequence analysis ,Pseudogene ,Chromosomes, Human, Pair 22 ,Molecular Sequence Data ,Biology ,Polymerase Chain Reaction ,Homology (biology) ,Evolution, Molecular ,Sequence Homology, Nucleic Acid ,Gene duplication ,Genetics ,Humans ,Gene ,neoplasms ,Genetics (clinical) ,Phylogeny ,Chromosomes, Human, Pair 14 ,Neurofibromin 1 ,Base Sequence ,Nucleic acid sequence ,Chromosome ,Chromosome Mapping ,Nucleic Acid Hybridization ,Proteins ,DNA ,nervous system diseases ,Chromosomes, Human, Pair 2 ,Chromosome 22 ,Pseudogenes - Abstract
Neurofibromatosis type 1 (NF1) is a frequent hereditary disorder that involves tissues derived from the embryonic neural crest. Besides the functional gene on chromosome arm 17q, NF1-related sequences (pseudogenes) are present on a number of chromosomes including 2, 12, 14, 15, 18, 21, and 22. We elucidated the complete nucleotide sequence of the NF1 pseudogene on chromosome 22. Only the middle part of the functional gene but not exons 21-27a, encoding the functionally important GAP-related domain of the NF1 protein, is presented in this pseudogene. In addition to the two known NF1 pseudogenes on chromosome 14 we identified two novel variants. A phylogenetic tree was constructed, from which we concluded that the NF1 pseudogenes on chromosomes 2, 14, and 22 are closely related to each other. Clones containing one of these pseudogenes cross-hybridised with the other pseudogenes in this subset, but did not reveal any in situ hybridisation with the functional NF1 gene or with NF1 pseudogenes on other chromosomes. This suggests that their hybridisation specificity is mainly determined by homologous sequences flanking the pseudogenes. Strong support for this concept was obtained by sequence analysis of the flanking regions, which revealed more than 95% homology. We hypothesise that during evolution this subset of NF1 pseudogenes initially arose by duplication and transposition of the middle part of the functional NF1 gene to chromosome 2. Subsequently, a much larger fragment, including flanking sequences, was duplicated and gave rise to the current NF1 pseudogene copies on chromosomes 14 and 22.
- Published
- 2000
30. The DNA sequence of human chromosome 22
- Author
-
L. Song, D. M. Lloyd, R. M. Swann, Ian F Korf, Lucinda Fulton, Carol Soderlund, I. D. Martyn, A. King, W Burrill, H. Wu, Y. Ramsey, Tracy Rohlfing, Mark T. Ross, Robert S. Fulton, L. Spragon, Darek Kedra, Laurens G. Wilming, Lisa Edelmann, James G. R. Gilbert, L. Williams, L. Chu, K. Fleming, J. Burgess, S. Shaull, M. N. Whiteley, Phil Latreille, Y. Qian, Ian Dunham, Dan Layman, Jennifer Lewis, A. C.C. Wong, Nobuyoshi Shimizu, Noriaki Aoki, Melanie M. Wall, Margaret A. Leversha, Ingegerd Fransson, M. Vaudin, Takashi Sasaki, Bernice E. Morrow, Graeme T Clark, S. Lewis, S. M. Clegg, H. Ramsay, A M Kimberley, S. J. Dodsworth, Melvin I. Simon, Stephan Beck, D. Conroy, Joseph A. Murray, Michele Clamp, Jan P. Dumanski, Christine Lloyd, Joseph L. McClay, P. Hu, Genwei Zhang, Adrienne Hunt, Steve Kenton, Antony V. Cox, Tina Graves, T. Nguyen, Lesley J. Rogers, Kazuhiko Kawasaki, Luc J. Smink, C. Dockree, J. M. Fey, J. C. Davis, U. J. Kim, Nigel P. Carter, Philip Ozersky, R. W. Heathcott, Richard Durbin, Ai Shintani, J Bailey, S. Bourne, Feng Chen, Harminder Sehra, Sulagna C. Saitta, G. Hall-Tamlyn, Charmain L. Wright, A. A. Garner, T. Do, Jane Rogers, Rebekah Hall, Joseph A. Bedell, Shuichi Asakawa, K Bates, J P Almeida, C. Hall, R. Pavitt, Charlotte G. Cole, K. Hinds, N Corby, V. Cobley, D. Pearson, Beverly S. Emanuel, C. Odell, Carl E.G. Bruder, Darren Grafham, Hiroki Kurahashi, Cordelia Langford, Dave Willey, T. E. Wilmer, David R. Bentley, I. Tapia, Hiroaki Shizuya, Myriam Peyrard, Tamim H. Shaikh, J K Kershaw, F. Fang, LaDeana W. Hillier, P. Loh, C L Bagguley, Tim Hubbard, John Sulston, Z. Wang, Kazunori Shibuya, R. E. Collier, Melanie E. Goward, K F Barlow, Richard Bruskiewich, M. L. Budarf, Yuan Chen, Kathryn L. Evans, Sarah E. Hunt, Judy S. Crabtree, Benjamin Phillimore, Stuart McLaren, M Mashreghi-Mohammadi, S. Chissoe, D. Willingham, J. Hawkins, Huaqin Pan, Q. Wang, Michelle Smith, H. Bradshaw, C. Walker, C. D. Skuce, Jim White, Amanda McMurray, Lucy Matthews, John Burton, Patricia Wohldmann, G. Bemis, O. Beasley, Robert H. Waterston, David W. Johnson, Elaine R. Mardis, H. Williamson, D. Buck, Yuhang Wang, Andrew D. Ellington, Zijin Du, Eyal Seroussi, Susumu Mitsuyama, A. Wamsley, Joanne O. Nelson, Y. Yoshizaki, K. P. O'Brien, H. I. Lao, R. Connor, S. Smalley, Anne Bridgeman, R Ainscough, Matthew Jones, Elisabeth Dawson, Joanna Collins, Pawandeep Dhami, S. Holmes, S. Phan, L. Ray, Angela Dorman, O. T. McCann, Christine P. Bird, Sarah Milne, Q. Ren, B. J. Mortimore, Carol Scott, Lisa French, Shuk-Mei Ho, G. J. Coville, Richard K. Wilson, Patrick Minx, Ziyun Yao, Jun Kudoh, David Beare, Charles A. Steward, Hongshing Lai, Alexander Johnson, Scott M. Williams, Robert W. Plumb, M. Zhan, Y. Fu, A. V. Pearce, S. Blakey, D. Goela, Gavin K. Laird, N. Miller, Matt Cordes, Kymberlie H. Pepin, Sam Phillips, David Bentley, Stéphane Deschamps, A. Do, Shaoping Lin, Shinsei Minoshima, Bruce A. Roe, Axin Hua, S. Qi, C Carder, Paul Scheet, Mark Griffiths, A K Babbage, J. M. Wallis, Heather E. McDermid, Eda Malaj, D. Sloan, and K. Kemp
- Subjects
Multidisciplinary ,Sequence analysis ,Chromosomes, Human, Pair 22 ,Molecular Sequence Data ,Nucleic acid sequence ,Gene Dosage ,Chromosome Mapping ,Computational biology ,DNA ,Sequence Analysis, DNA ,Biology ,ENCODE ,Genome ,Complete sequence ,Mice ,Species Specificity ,Human Genome Project ,Animals ,Humans ,Human genome ,Sequence (medicine) ,Genomic organization ,Repetitive Sequences, Nucleic Acid - Abstract
Knowledge of the complete genomic DNA sequence of an organism allows a systematic approach to defining its genetic components. The genomic sequence provides access to the complete structures of all genes, including those without known function, their control elements, and, by inference, the proteins they encode, as well as all other biologically important sequences. Furthermore, the sequence is a rich and permanent source of information for the design of further biological studies of the organism and for the study of evolution through cross-species sequence comparison. The power of this approach has been amply demonstrated by the determination of the sequences of a number of microbial and model organisms. The next step is to obtain the complete sequence of the entire human genome. Here we report the sequence of the euchromatic part of human chromosome 22. The sequence obtained consists of 12 contiguous segments spanning 33.4 megabases, contains at least 545 genes and 134 pseudogenes, and provides the first view of the complex chromosomal landscapes that will be found in the rest of the genome.
- Published
- 1999
31. Identification and characterization of NF1-related loci on human chromosomes 22, 14 and 2
- Author
-
Peter Riegman, Engelien H. Bijleveld, Ian Dunham, Theo J. M. Hulsebos, Luc J. Smink, and Other departments
- Subjects
Mutation rate ,congenital, hereditary, and neonatal diseases and abnormalities ,Pseudogene ,Chromosomes, Human, Pair 22 ,Restriction Mapping ,Gene Conversion ,Locus (genetics) ,Biology ,Gene mapping ,Genes, Neurofibromatosis 1 ,Genetics ,Humans ,neoplasms ,Chromosomes, Artificial, Yeast ,Genetics (clinical) ,In Situ Hybridization, Fluorescence ,Genes, Dominant ,Chromosomes, Human, Pair 14 ,Chromosome ,Chromosome Mapping ,Cosmids ,nervous system diseases ,Blotting, Southern ,Chromosome Arm ,Chromosomes, Human, Pair 2 ,Mutation ,Human genome ,DNA Probes ,Chromosome 22 ,Pseudogenes - Abstract
Neurofibromatosis type 1 (NF1) is a frequent hereditary disorder. The disease is characterized by a very high mutation rate (up to 1/10000 gametes per generation). NF1-related loci in the human genome have been implicated in the high mutation rate by hypothesizing that these carry disease-causing mutations, which can be transferred to the functional NF1 gene on chromosome arm 17q by interchromosomal gene conversion. To test this hypothesis, we want to identify and characterize the NF1-related loci in the human genome. In this study, we have localized an NF1-related locus in the most centromeric region of the long arm of chromosome 22. We demonstrate that this locus contains sequences homologous to cDNAs that include the GAP-related domain of the functional NF1 gene. However, the GAP-related domain itself is not represented in this locus. In addition, cosmids specific to this locus reveal, by in situ hybridization, NF1-related loci in the pericentromeric region of chromosome arm 14q and in chromosomal band 2q21. These cosmids will enable us to determine whether identified disease-causing mutations are present at the chromosome 22-associated NF1-related locus.
- Published
- 1996
32. A bacterial artificial chromosome-based framework contig map of human chromosome 22q
- Author
-
Ung Jin Kim, Hyung Lyun Kang, Melvin I. Simon, Julie R. Korenberg, Hiroaki Shizuya, Ian Dunham, Bruce W. Birren, Sun Shim Choi, Luc J. Smink, and Charmain L. Garrett
- Subjects
Genetics ,Yeast artificial chromosome ,Bacterial artificial chromosome ,Genomic Library ,Multidisciplinary ,Contig ,Gene map ,Chromosomes, Human, Pair 22 ,food and beverages ,Chromosome Mapping ,Genome project ,Human artificial chromosome ,Biology ,Chromosomes, Bacterial ,Cell Line ,Chromosome 19 ,Humans ,Chromosome 21 ,Caltech Library Services ,Research Article - Abstract
We have constructed a physical map of human chromosome 22q using bacterial artificial chromosome (BAC) clones. The map consists of 613 chromosome 22-specific BAC clones that have been localized and assembled into contigs using 452 landmarks, 346 of which were previously ordered and mapped to specific regions of the q arm of the chromosome by means of chromosome 22-specific yeast artificial chromosome clones. The BAC-based map provides immediate access to clones that are stable and convenient for direct genome analysis. The approach to rapidly developing marker-specific BAC contigs is relatively straightforward and can be extended to generate scaffold BAC contig maps of the rest of the chromosomes. These contigs will provide substrates for sequencing the entire human genome. We discuss how to efficiently close contig gaps using the end sequences of BAC clone inserts.
- Published
- 1996
33. Sequencing and association analysis of the type 1 diabetes – linked region on chromosome 10p12-q11
- Author
-
Felicity Payne, Sarah Nutland, Oliver S. Burren, Neil Walker, Lisa Godfrey, Ian Dunham, Christopher E. Lowe, John A. Todd, Luc J. Smink, Jennifer Masters, Yuan Chen, Alex C. Lam, Charmain L. Wright, Helen Stevens, Jane Rogers, Sergey Nejentsev, Panagiotis Deloukas, Lisa French, Rebecca C.J. Twells, Rebecca Bailey, Deborah J. Smyth, Bryan J. Barratt, Anne Smith, Nezhentsev, Sergey [0000-0002-7528-4461], Burren, Oliver [0000-0002-3388-5760], and Apollo - University of Cambridge Repository
- Subjects
Genetic Markers ,Male ,Candidate gene ,lcsh:QH426-470 ,Genotype ,onset ,Single-nucleotide polymorphism ,Biology ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,il-2 ,Gene Frequency ,SEARCH ,Genetics ,Polymorphic Microsatellite Marker ,HUMAN GENOME ,Humans ,Genetics(clinical) ,SYSTEMIC-LUPUS-ERYTHEMATOSUS ,Genetics (clinical) ,030304 developmental biology ,Genetic association ,DNA Primers ,0303 health sciences ,Contig ,Chromosomes, Human, Pair 10 ,families ,Physical Chromosome Mapping ,3. Good health ,lcsh:Genetics ,Diabetes Mellitus, Type 1 ,Genetic marker ,chemokine gene variant ,Case-Control Studies ,Chromosomal region ,T-CELLS ,Microsatellite ,Female ,linkage ,030215 immunology ,Research Article - Abstract
Background In an effort to locate susceptibility genes for type 1 diabetes (T1D) several genome-wide linkage scans have been undertaken. A chromosomal region designated IDDM10 retained genome-wide significance in a combined analysis of the main linkage scans. Here, we studied sequence polymorphisms in 23 Mb on chromosome 10p12-q11, including the putative IDDM10 region, to identify genes associated with T1D. Results Initially, we resequenced the functional candidate genes, CREM and SDF1, located in this region, genotyped 13 tag single nucleotide polymorphisms (SNPs) and found no association with T1D. We then undertook analysis of the whole 23 Mb region. We constructed and sequenced a contig tile path from two bacterial artificial clone libraries. By comparison with a clone library from an unrelated person used in the Human Genome Project, we identified 12,058 SNPs. We genotyped 303 SNPs and 25 polymorphic microsatellite markers in 765 multiplex T1D families and followed up 22 associated polymorphisms in up to 2,857 families. We found nominal evidence of association in six loci (P = 0.05 – 0.0026), located near the PAPD1 gene. Therefore, we resequenced 38.8 kb in this region, found 147 SNPs and genotyped 84 of them in the T1D families. We also tested 13 polymorphisms in the PAPD1 gene and in five other loci in 1,612 T1D patients and 1,828 controls from the UK. Overall, only the D10S193 microsatellite marker located 28 kb downstream of PAPD1 showed nominal evidence of association in both T1D families and in the case-control sample (P = 0.037 and 0.03, respectively). Conclusion We conclude that polymorphisms in the CREM and SDF1 genes have no major effect on T1D. The weak T1D association that we detected in the association scan near the PAPD1 gene may be either false or due to a small genuine effect, and cannot explain linkage at the IDDM10 region.
- Published
- 2007
34. [Untitled]
- Author
-
Rebecca Pask, Neil Walker, Constantin Ionescu-Tirgoviste, John S. Hulme, Sarah Nutland, David A. Savage, Dag E. Undlien, Laurence B. Peterson, Lisa M. Maier, Heather I. Fraser, John A. Todd, Kara Hunter, Felicity Payne, Giselle Chamberlain, Cristian Guja, Deborah J. Smyth, Kjersti S. Rønningen, Christopher E. Lowe, Linda S. Wicker, Luc J. Smink, Adrian Vella, David P. Strachan, Rebecca C.J. Twells, Jason D. Cooper, and Carolyn Moule
- Subjects
2. Zero hunger ,Genetics ,0303 health sciences ,Candidate gene ,Sequence analysis ,Single-nucleotide polymorphism ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Polymorphism (computer science) ,Genotype ,Microsatellite ,Human genome ,Genetics (clinical) ,030304 developmental biology ,030215 immunology ,Genetic association - Abstract
One strategy to help identify susceptibility genes for complex, multifactorial diseases is to map disease loci in a representative animal model of the disorder. The nonobese diabetic (NOD) mouse is a model for human type 1 diabetes. Linkage and congenic strain analyses have identified several NOD mouse Idd (insulin dependent diabetes) loci, which have been mapped to small chromosome intervals, for which the orthologous regions in the human genome can be identified. Here, we have conducted re-sequencing and association analysis of six orthologous genes identified in NOD Idd loci: NRAMP1/SLC11A1 (orthologous to Nramp1/Slc11a1 in Idd5.2), FRAP1 (orthologous to Frap1 in Idd9.2), 4-1BB/CD137/TNFRSF9 (orthologous to 4-1bb/Cd137/Tnrfrsf9 in Idd9.3), CD101/IGSF2 (orthologous to Cd101/Igsf2 in Idd10), B2M (orthologous to B2m in Idd13) and VAV3 (orthologous to Vav3 in Idd18). Re-sequencing of a total of 110 kb of DNA from 32 or 96 type 1 diabetes cases yielded 220 single nucleotide polymorphisms (SNPs). Sixty-five SNPs, including 54 informative tag SNPs, and a microsatellite were selected and genotyped in up to 1,632 type 1 diabetes families and 1,709 cases and 1,829 controls. None of the candidate regions showed evidence of association with type 1 diabetes (P values > 0.2), indicating that common variation in these key candidate genes does not play a major role in type 1 diabetes susceptibility in the European ancestry populations studied.
- Published
- 2005
- Full Text
- View/download PDF
35. [Untitled]
- Author
-
Sarah Nutland, John A. Todd, Luc J. Smink, Deborah J. Smyth, Rebecca C.J. Twells, Alex C. Lam, Meera Sebastian, Rebecca Pask, Helen E. Rance, Anne Smith, Neil Walker, and Bryan J. Barratt
- Subjects
Genetics ,Whole Genome Amplification ,0303 health sciences ,Multiple displacement amplification ,Nucleic acid amplification technique ,Biology ,Molecular Inversion Probe ,SNP genotyping ,03 medical and health sciences ,genomic DNA ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Human genome ,Genotyping ,030304 developmental biology ,Biotechnology - Abstract
Sustainable DNA resources and reliable high-throughput genotyping methods are required for large-scale, long-term genetic association studies. In the genetic dissection of common disease it is now recognised that thousands of samples and hundreds of thousands of markers, mostly single nucleotide polymorphisms (SNPs), will have to be analysed. In order to achieve these aims, both an ability to boost quantities of archived DNA and to genotype at low costs are highly desirable. We have investigated Φ29 polymerase Multiple Displacement Amplification (MDA)-generated DNA product (MDA product), in combination with highly multiplexed BeadArray™ genotyping technology. As part of a large-scale BeadArray genotyping experiment we made a direct comparison of genotyping data generated from MDA product with that from genomic DNA (gDNA) templates. Eighty-six MDA product and the corresponding 86 gDNA samples were genotyped at 345 SNPs and a concordance rate of 98.8% was achieved. The BeadArray sample exclusion rate, blind to sample type, was 10.5% for MDA product compared to 5.8% for gDNA. We conclude that the BeadArray technology successfully produces high quality genotyping data from MDA product. The combination of these technologies improves the feasibility and efficiency of mapping common disease susceptibility genes despite limited stocks of gDNA samples.
- Published
- 2004
- Full Text
- View/download PDF
36. Development of an integrated genome informatics, data management and workflow infrastructure: A toolbox for the study of complex disease genetics
- Author
-
Neil Walker, Felicity Payne, Daniel B. Rainbow, Vincent H. Everett, Helen Schuilenburg, Oliver S. Burren, Barry C. Healy, John A. Todd, Davide Laneri, Sarah Nutland, Luc J. Smink, Helen E. Rance, Geoffrey E. Dolman, Linda S. Wicker, Christopher E. Lowe, Alex C. Lam, Bryan J. Barratt, Rebecca C.J. Twells, Deborah J. Smyth, Burren, Oliver [0000-0002-3388-5760], and Apollo - University of Cambridge Repository
- Subjects
Quality Control ,Candidate gene ,Informatics ,Databases, Factual ,lcsh:QH426-470 ,Genetic Linkage ,type 1 diabetes ,Information Storage and Retrieval ,lcsh:Medicine ,Genomics ,Biology ,Models, Biological ,Polymorphism, Single Nucleotide ,Genome ,03 medical and health sciences ,Drug Discovery ,Genetics ,Animals ,Chromosomes, Human ,Humans ,Ensembl ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Biological data ,Models, Genetic ,Genome, Human ,030306 microbiology ,complex disease ,lcsh:R ,Genetic Diseases, Inborn ,Chromosome Mapping ,Computational Biology ,Sequence Analysis, DNA ,Gene Annotation ,genome informatics ,Disease Models, Animal ,lcsh:Genetics ,Diabetes Mellitus, Type 1 ,Database Management Systems ,Molecular Medicine ,Human genome ,data management ,Primary Research ,Information Systems - Abstract
The genetic dissection of complex disease remains a significant challenge. Sample-tracking and the recording, processing and storage of high-throughput laboratory data with public domain data, require integration of databases, genome informatics and genetic analyses in an easily updated and scaleable format. To find genes involved in multifactorial diseases such as type 1 diabetes (T1D), chromosome regions are defined based on functional candidate gene content, linkage information from humans and animal model mapping information. For each region, genomic information is extracted from Ensembl, converted and loaded into ACeDB for manual gene annotation. Homology information is examined using ACeDB tools and the gene structure verified. Manually curated genes are extracted from ACeDB and read into the feature database, which holds relevant local genomic feature data and an audit trail of laboratory investigations. Public domain information, manually curated genes, polymorphisms, primers, linkage and association analyses, with links to our genotyping database, are shown in Gbrowse. This system scales to include genetic, statistical, quality control (QC) and biological data such as expression analyses of RNA or protein, all linked from a genomics integrative display. Our system is applicable to any genetic study of complex disease, of either large or small scale.
- Published
- 2004
- Full Text
- View/download PDF
37. Transcriptional regulation by p53: more than meets the eye?
- Author
-
Luc J Smink
- Subjects
DNA binding site ,Chromatin binding ,Nucleosome ,ChIP-on-chip ,Biology ,Molecular Biology ,Biochemistry ,Molecular biology ,ChIA-PET ,Chromatin remodeling ,Chromatin ,Cell biology ,ChIP-sequencing - Abstract
The tumour-suppressor protein p53 has the ability to regulate such diverse and crucial processes as cell-cycle progression and apoptosis. It is also mutated in about half of all human tumours. However, little is known about the actual mechanism of interaction of p53 with its target genes and how it regulates their expression. Espinosa and Emerson [1xTranscriptional regulation by p53 through intrinsic DNA/chromatin binding and site-directed cofactor recruitment. Espinosa, J.M. and Emerson, B.M. Mol. Cell. 2001; 8: 57–59Abstract | Full Text | Full Text PDF | PubMed | Scopus (332)See all References][1] have begun to address this issue by developing a p53-dependent in vitro transcription system. This system exploits the p21 promoter, which contains two consensus p53-binding sites at 2.3 kb (5′) and 1.4 kb (3′).In electronic mobility shift assays, p53 was mainly inactive for specific binding of oligonucleotides, 25 bp in length and containing the p21 5′ promoter site. This binding activity was dramatically improved by p300-mediated acetylation. The authors also showed that transcriptional activation of a chromatin-assembled p21-reporter gene construct, in which the p21 promoter drives transcription of a luciferase reporter gene, requires p53 (acting at a distance of ≥1.4 kb) in combination with p300.DNase I chromatin footprinting assays were used to study the mechanism by which p300 mediates p53-dependent transcriptional activation. Surprisingly, p53 alone created a footprinting pattern at the 5′ and the 3′ binding sites in the chromatin-assembled p21 promoter, indicating occupancy of both sites. Addition of p300 did not alter the binding significantly, but did enhance transcription from the p21 promoter.In explanation of these results, footprinting experiments with chromatin samples lacking ATP-dependent nucleosome remodeling activities, ATP or acetyl-CoA, and the majority of chromatin assembly proteins, revealed that p53 has an almost undetectable binding on short oligonucleotides. By contrast, it binds efficiently to the same sites when assembled into chromatin, in the absence of chromatin modifying or remodeling activities. The addition of p300 does not increase the binding of p53 to p21 in chromatin but does increase the affinity to short oligonucleotides containing the p21-binding sites.Further experiments showed that putative activating modifications, binding of the p53 antibody PAb421 or acetylation, do not improve p53-binding activity. The authors also showed that affinity of p53 for the 5′ promoter improves as the target DNA size increases. These results suggest the involvement of secondary structures in the binding sites.The data presented by Espinosa and Emerson suggest that nucleosomes in chromatin-assembled p21-promoter-containing plasmids are acetylated by p300 in the presence of p53. Acetylation is highest when p300 is recruited to the 3′ promoter, and then presumably spreads to encompass the TATA box. These experiments offer a valuable first insight into p53-mediated activation. The ability of p53 to act as both an activator and a repressor, and its ability to recruit different cofactors, might be reliant on its binding to DNA and/or chromatin with specific topologies.
- Published
- 2001
- Full Text
- View/download PDF
38. Acetylation can regulate cell-cycle progression
- Author
-
Luc J. Smink
- Subjects
biology ,Cyclin-dependent kinase 2 ,Molecular biology ,Fusion protein ,Histone ,Acetylation ,Cyclin-dependent kinase ,biology.protein ,Molecular Medicine ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,CREB-binding protein ,E2F ,Molecular Biology - Abstract
The retinoblastoma tumour suppressor (pRb) is crucial for the negative control of cell proliferation. The protein has a major role in G1- to S-phase transition by controlling key transcription factors such as the E2F family. In tumour cells, pRb is sequestered by viral oncoproteins (E1A) and regulated by phosphorylation through G1 cyclin-dependent kinases (CDKs),primarily cyclinD/CDK4 and cyclinE/Cdk2,which sequentially phosphorylate pRb as the cells move towards S-phase.In a new study, Chan et al. 1xAcetylation control of the retinoblastoma tumour-suppressor protein. Chan, H.M. et al. Nat. Cell Biol. 2001; 3: 667–674CrossRef | PubMed | Scopus (194)See all References1 have identified acetylation as a new type of regulator of pRb function. The acetylation is mediated by p300 through adenovirus E1A recruitment of pRb and p300/CREB binding protein (CBP) in a multimeric complex. Both pRb and p300/CBP are targets for viral oncoproteins, such as the adenovirus E1A.In an elegant series of experiments, the authors tested the acetylation of pRb by the histone acetyl transferase (HAT) activity of p300 using a glutathione S-transferase (GST)–pRb fusion protein in vitro. This is influenced by the integrity of a pocket domain at the C-terminus of the pRb protein. Two pocket mutants, isolated from tumour cells, showed reduced acetylation. To establish whether this also happened in vivo, the authors used an antibody against acetylated lysines in pRB and showed the presence of endogenous acetylated pRb. Under similar circumstances, the pocket mutant was not acetylated. Use of mutant derivatives showed that the main site of acetylation was located between amino-acid residues 794 and 880. There are eight lysine residues in this region, and Chan et al. focused on the five lysines between residues 830 and 884, systematically altering each lysine to arginine and identifying lysines 873 and 874 as being acetylated. Having done this, the authors assessed whether acetylation might influence pRb phosphorylation, by using constructs where the lysines at residues 873 and 874 were replaced by arginines or glutamines (873/874RR and 873/874QQ, respectively). A change to glutamine mimics the acetylation status, whereas a change to arginine prevents acetylation. Experiments using these constructs showed that wild-type pRb and 873/874 RR were phosphorylated to a similar extent, but the 873/874 QQ construct showed a significant reduction in phosphorylation. The p300-dependent acetylation of pRb is increased by adenovirus E1A, through recruitment of p300 and pRb into the same complex. This process is dependent on E1A concentrations: at high E1A concentrations the acetylation of pRb is lost. To assess the function of acetylation, the authors looked at proteins that interact with the C-terminus of the pRb protein. For example, the MDM2 oncoprotein bound more efficiently to pRb in its acytelated form, both in vitro and in vivo. Similar experiments with E2F failed to detect any differences.These recent advances suggest acetylation as a new control mechanism in regulating pRb activity and a new mechanism through which viral oncoproteins can affect tumour-suppressor activity.
- Published
- 2001
- Full Text
- View/download PDF
39. Senescence bypass screen identifies novel immortalizing genes
- Author
-
Luc J. Smink
- Subjects
Senescence ,cDNA library ,Biology ,Molecular biology ,BMI1 ,CDKN2A ,Complementary DNA ,Cancer research ,Molecular Medicine ,E2F1 ,neoplasms ,Molecular Biology ,Gene ,Genetic screen - Abstract
The lifespan of normal cells is limited by several control mechanisms, and ends either in replicative senescence or apoptosis. In cancerous cells these control mechanisms are bypassed. Jacobs et al.1xSenescence bypass screen identifies TBX2, which represses Cdkn2a (p19ARF) and is amplified in a subset of human breast cancers. Jacobs, J.J.L et al. Nat. Genet. 2000; 26: 291–299Crossref | PubMed | Scopus (252)See all References1 have identified TBX2 as a new immortalising gene, using a genetic screen which identifies genes able to bypass the senescence arrest of Bmi1 oncogene deficient primary fibroblasts.Absence of Bmi1 causes accumulation of Cdkn2a (p16INK4a) and Cdkn2a (p19ARF), leading to premature senescence and proliferation defects in lymphoid organs and cerebellum. Using Bmi1 deficient mouse embryo fibroblasts (MEFs) in a retroviral cDNA library screen, the authors expected to find strong inhibitors of the Cdkn2a pathway. In five experiments the MEFs were infected with a replication-deficient retroviral cDNA library. After two to three weeks colonies started to arise. To distinguish between clones that escaped senescence spontaneously and those that contained cDNA inserts, clones were infected with wild-type replication-competent MoMuLV – a tool to mobilize retroviral cDNA inserts. The viruses produced were used in a second Bmi-/- infection. The complete cDNA for human TBX2 – a member of the T-box family of transcription factors – was identified several times. Recloning TBX2 cDNA into retroviral vectors and infection of Bmi-/- MEFs showed that TBX2 prevents premature senescence and immortalises MEFs.Analysis of Cdkn2a (p16INK4a) and Cdkn2a (p19ARF) concentrations after infection with TBX2-containing retroviruses showed a strong downregulation of Cdkn2a (p19ARF) and a relatively small reduction of Cdkn2a (p16INK4a). This appears to be sufficient for full immortalization since the TBX2-immoratilised MEFs do not display the frequent disruption of the Cdkn2a locus or Trp53 loss of function, which is most often observed in spontaneous immortalization of wild-type MEFs. TBX2 also attenuates E2F1, Myc or HRAS-mediated induction of Cdkn2a. These results show – possibly for the first time – the association of overexpression of a T-box gene with regulation of the Cdkn2a checkpoint and the onset of cancer.The TBX2 gene resides on human chromosome 17q23, a region frequently amplified in human breast cancers. Jacob and colleagues showed amplification of TBX2 in a subset of both sporadic and familial human breast cancers. The authors are currently pursuing the question whether TBX2 expression can be correlated to certain clinical or prognostic features of breast cancer.
- Published
- 2001
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.