Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 411(6834): 199-204, 2001 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11346797

RESUMO

With the availability of a dense genome-wide map of single nucleotide polymorphisms (SNPs), a central issue in human genetics is whether it is now possible to use linkage disequilibrium (LD) to map genes that cause disease. LD refers to correlations among neighbouring alleles, reflecting 'haplotypes' descended from single, ancestral chromosomes. The size of LD blocks has been the subject of considerable debate. Computer simulations and empirical data have suggested that LD extends only a few kilobases (kb) around common SNPs, whereas other data have suggested that it can extend much further, in some cases greater than 100 kb. It has been difficult to obtain a systematic picture of LD because past studies have been based on only a few (1-3) loci and different populations. Here, we report a large-scale experiment using a uniform protocol to examine 19 randomly selected genomic regions. LD in a United States population of north-European descent typically extends 60 kb from common alleles, implying that LD mapping is likely to be practical in this population. By contrast, LD in a Nigerian population extends markedly less far. The results illuminate human history, suggesting that LD in northern Europeans is shaped by a marked demographic event about 27,000-53,000 years ago.


Assuntos
Mapeamento Cromossômico/métodos , Genoma Humano , Desequilíbrio de Ligação/genética , Polimorfismo de Nucleotídeo Único/genética , Alelos , Viés , Simulação por Computador , Europa (Continente)/etnologia , Efeito Fundador , Doenças Genéticas Inatas/genética , Haplótipos/genética , Heterozigoto , Humanos , Modelos Genéticos , Nigéria , Filogenia , Grupos Raciais/genética , Recombinação Genética/genética , Reprodutibilidade dos Testes , Seleção Genética , Fatores de Tempo , Estados Unidos
3.
Proc Natl Acad Sci U S A ; 97(22): 12164-9, 2000 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-11035790

RESUMO

Generating human single-nucleotide polymorphisms (SNPs) is no longer a rate-limiting step for genetic studies of disease. The number of SNPs in public databases already exceeds 200,000, and the total is expected to exceed 1,000,000 within a year. Rather, progress is limited by the inability to genotype large numbers of SNPs. Current genotyping methods are suitable for studying individual loci or at most a handful at a time. Here, we describe a method for parallel genotyping of SNPs, called single base extension-tag array on glass slides, SBE-TAGS. The principle is as follows. SNPs are genotyped by single base extension (SBE), using bifunctional primers carrying a unique sequence tag in addition to a locus-specific sequence. Because each locus has a distinct tag, the genotyping reactions can be performed in a highly multiplexed fashion, and the resulting product can then be "demultiplexed" by hybridization to the reverse complements of the sequence tags arrayed on a glass slide. SBE-TAGS is simple and inexpensive because of the high degree of multiplexing and the use of an easily generated, generic tag array. The method is also highly accurate: we genotyped over 100 SNPs, obtaining over 5, 000 genotypes, with approximately 99% accuracy.


Assuntos
Perfilação da Expressão Gênica , Polimorfismo de Nucleotídeo Único , Animais , Primers do DNA , Genótipo , Humanos , Camundongos , Reação em Cadeia da Polimerase
4.
Nat Genet ; 26(1): 76-80, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10973253

RESUMO

Genetic association studies are viewed as problematic and plagued by irreproducibility. Many associations have been reported for type 2 diabetes, but none have been confirmed in multiple samples and with comprehensive controls. We evaluated 16 published genetic associations to type 2 diabetes and related sub-phenotypes using a family-based design to control for population stratification, and replication samples to increase power. We were able to confirm only one association, that of the common Pro12Ala polymorphism in peroxisome proliferator-activated receptor-gamma(PPARgamma) with type 2 diabetes. By analysing over 3,000 individuals, we found a modest (1.25-fold) but significant (P=0.002) increase in diabetes risk associated with the more common proline allele (85% frequency). Moreover, our results resolve a controversy about common variation in PPARgamma. An initial study found a threefold effect, but four of five subsequent publications failed to confirm the association. All six studies are consistent with the odds ratio we describe. The data implicate inherited variation in PPARgamma in the pathogenesis of type 2 diabetes. Because the risk allele occurs at such high frequency, its modest effect translates into a large population attributable risk-influencing as much as 25% of type 2 diabetes in the general population.


Assuntos
Diabetes Mellitus Tipo 2/genética , Polimorfismo Genético , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Adulto , Idade de Início , Idoso , Alanina/genética , Alelos , Glicemia/genética , Pressão Sanguínea/genética , Índice de Massa Corporal , Colesterol/genética , Saúde da Família , Pai , Feminino , Genótipo , Humanos , Desequilíbrio de Ligação , Lipoproteínas HDL/genética , Masculino , Pessoa de Meia-Idade , Modelos Genéticos , Mães , Fenótipo , Prolina/genética , Fatores de Risco
5.
Proc Natl Acad Sci U S A ; 97(1): 268-73, 2000 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-10618407

RESUMO

Reduced penetrance in genetic disorders may be either dependent or independent of the genetic background of gene carriers. Hirschsprung disease (HSCR) demonstrates a complex pattern of inheritance with approximately 50% of familial cases being heterozygous for mutations in the receptor tyrosine kinase RET. Even when identified, the penetrance of RET mutations is only 50-70%, gender-dependent, and varies with the extent of aganglionosis. We searched for additional susceptibility genes which, in conjunction with RET, lead to phenotypic expression by studying 12 multiplex HSCR families. Haplotype analysis and extensive mutation screening demonstrated three types of families: six families harboring severe RET mutations (group I); and the six remaining families, five of which are RET-linked families with no sequence alterations and one RET-unlinked family (group II). Although the presence of RET mutations in group I families is sufficient to explain HSCR inheritance, a genome scan reveals a new susceptibility locus on 9q31 exclusively in group II families. As such, the gene at 9q31 is a modifier of HSCR penetrance. These observations imply that identification of new susceptibility factors in a complex disease may depend on classification of families by mutational type at known susceptibility genes.


Assuntos
Cromossomos Humanos Par 9/genética , Proteínas de Drosophila , Doença de Hirschsprung/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Feminino , Ligação Genética , Marcadores Genéticos , Haplótipos , Humanos , Escore Lod , Masculino , Modelos Genéticos , Mutação , Linhagem , Penetrância , Fenótipo , Proteínas Proto-Oncogênicas c-ret , Splicing de RNA/genética , Estatísticas não Paramétricas
7.
Genomics ; 48(3): 354-62, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9545641

RESUMO

Congenital aganglionic megacolon, commonly known as Hirschsprung disease (HSCR), is the most frequent cause of congenital bowel obstruction. Germline mutations in the RET receptor tyrosine kinase have been shown to cause HSCR. Knockout mice for RET and for its ligand, glial cell line-derived neurotrophic factor (GDNF), exhibit both complete intestinal aganglionosis and renal defects. Recently, GDNF and GFRA1 (GDNF family receptor, also known as GDNFR-alpha), its GPI-linked coreceptor, were demonstrated to be components of a functional ligand for RET. Moreover, GDNF has been implicated in rare cases of HSCR. We have mapped GFRA1 to human chromosome 10q25, isolated human and mouse genomic clones, determined the gene's intron-exon boundaries, isolated a highly polymorphic microsatellite marker adjacent to exon 7, and scanned for GFRA1 mutations in a large panel of HSCR patients. No evidence of linkage was detected in HSCR kindreds, and no sequence variants were found to be in significant excess in patients. These data suggest that GFRA1'S role in enteric neurogenesis in humans remains to be elucidated and that RET signaling in the gut may take place via alternate pathways, such as the recently described GDNF-related molecule neurturin and its GFRA1-like coreceptor, GFRA2.


Assuntos
Proteínas de Drosophila , Doença de Hirschsprung/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Sequência de Aminoácidos , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Humanos Par 10 , Clonagem Molecular , DNA Complementar/isolamento & purificação , Éxons , Marcadores Genéticos , Predisposição Genética para Doença , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Doença de Hirschsprung/metabolismo , Humanos , Íntrons , Ligantes , Repetições de Microssatélites , Dados de Sequência Molecular , Mutação , Proteínas Proto-Oncogênicas c-ret , Receptores Proteína Tirosina Quinases/metabolismo , Alinhamento de Sequência , Análise de Sequência de DNA
8.
Oncogene ; 17(23): 3065-70, 1998 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-9881709

RESUMO

Hirschsprung disease (HSCR), or congenital aganglionic megacolon, is the most frequent cause of congenital bowel obstruction. Germline mutations in the RET receptor tyrosine kinase have been shown to cause HSCR. Mice that carry null alleles for RET or for its ligand, glial cell line-derived neurotrophic factor (GDNF), both exhibit complete intestinal aganglionosis and renal defects. Recently, the Src homology 2 (SH2) domain-containing protein Grb10 has been shown to interact with RET in vitro and in vivo, early in development. We have confirmed the map location of GRB10 on human chromosome 7, isolated human BACs containing the gene, elucidated its genomic structure, isolated a highly polymorphic microsatellite marker adjacent to exon 14 and scanned the gene for mutations in a large panel of HSCR patients. No evidence of linkage was detected in HSCR kindreds and no mutations were found in patients. These data suggest that while GRB10 may be important for signal transduction in developing embryos, it does not play an obvious role in HSCR.


Assuntos
Proteínas Sanguíneas , Doença de Hirschsprung/genética , Fosfoproteínas , Proteínas/genética , Proteínas/fisiologia , Domínios de Homologia de src , Proteína Adaptadora GRB10 , Humanos
9.
Nat Genet ; 14(3): 341-4, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8896568

RESUMO

Hirschsprung disease (HSCR), or congenital aganglionic megacolon, is the most common cause of congenital bowel obstruction with an incidence of 1 in 5000 live births. HSCR may be inherited as a single gene disorder with reduced penetrance or as a multigenic trait. HSCR mutations have been identified in the RET receptor tyrosine kinase, endothelin-B receptor (EDNRB) and its physiological ligand, endothelin 3 (EDN3). Although RET's ligand has remained elusive, it is expected to be an extracellular neurotrophic molecule expressed in the developing gut and kidney mesenchyme, based on the phenotypes of intestinal aganglionosis and renal agenesis observed in homozygous RET knockout (Ret -/-) mice. The glial cell line-derived neurotrophic factor (GDNF) is such a molecule. Recently, mice carrying two null alleles for Gdnf were shown to exhibit phenotypes remarkably similar to Ret-/- animals. We screened 106 unrelated HSCR patients for mutations in GDNF by direct sequencing. We identified one familial mutation in a HSCR patient with a known de novo RET mutation and malrotation of the gut. No haplotype sharing was evident in any of 36 HSCR kindreds typed for microsatellite markers surrounding GDNF on human chromosome 5p. Our data suggest that GDNF is a minor contributor to human HSCR susceptibility and that loss of its function in enteric neurogenesis may be compensated for by other neurotrophic factors or via other pathways. However, it may be that in rare instances, RET and GDNF mutations act in concert to produce an enteric phenotype.


Assuntos
Proteínas de Drosophila , Mutação em Linhagem Germinativa , Doença de Hirschsprung/genética , Fatores de Crescimento Neural , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Animais , Mapeamento Cromossômico , Cromossomos Humanos Par 5 , Feminino , Marcadores Genéticos , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Heterozigoto , Humanos , Masculino , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-ret , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico
11.
Am J Med Genet ; 63(4): 603-9, 1996 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-8826440

RESUMO

Congenital central hypoventilation syndrome (CCHS) usually occurs as an isolated phenotype. However, 16% of the index cases are also affected with Hirschsprung disease (HSCR). Complex segregation analysis suggests that CCHS is familial and has the same inheritance pattern with or without HSCR. We postulate that alteration of normal function of the receptor tyrosine kinase, RET, may contribute to CCHS based on RET's expression pattern and the identification of RET mutations in HSCR patients. To further explore the nature of the inheritance of CCHS, we have undertaken two main routes of investigation: cytogenetic analysis and mutation detection. Cytogenetic analysis of metaphase chromosomes showed normal karyotypes in 13 of the 14 evaluated index cases; one index case carried a familial pericentric inversion on chromosome 2. Mutation analysis showed no sequence changes unique to index cases, as compared to control individuals, and as studied by single strand conformational polymorphism (SSCP) analysis of the coding region of RET. We conclude that point mutations in the RET coding region cannot account for a substantial fraction of CCHS in this patient population, and that other candidate genes involved in neural crest cell differentiation and development must be considered.


Assuntos
Proteínas de Drosophila , Hipoventilação/enzimologia , Hipoventilação/genética , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Síndromes da Apneia do Sono/congênito , Síndromes da Apneia do Sono/genética , Criança , Humanos , Hipoventilação/congênito , Cariotipagem , Mutação , Polimorfismo Conformacional de Fita Simples , Proteínas Proto-Oncogênicas c-ret , Síndromes da Apneia do Sono/enzimologia
12.
Hum Mol Genet ; 4(5): 821-30, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7633441

RESUMO

Hirschsprung disease (HSCR), or congenital aganglionic megacolon, is the most common cause of congenital bowel obstruction with an incidence of 1 in 5000 live births. Recently, linkage of an incompletely penetrant, dominant form of HSCR was reported, followed by identification of mutations in the RET receptor tyrosine kinase. To determine the frequency of RET mutations in HSCR and correlate genotype with phenotype, we have screened for mutations among 80 HSCR probands representing a wide range of phenotypes and family structures. Polymerase chain reaction (PCR) and single-strand conformation polymorphism (SSCP) analysis of RET's 20 exons for mutations among probands revealed eight putative mutations (10%). Sequence changes, which included missense, frameshift and complex mutations, were detected in both familial and isolated cases, among patients with both long- and short-segment HSCR and in three kindreds with other phenotypes (maternal deafness, talipes and malrotation of the gut, respectively). Two mutations (C609Y and C620R) we identified have previously been associated with multiple endocrine neoplasia type 2A (MEN2A), medullary thyroid carcinoma (MTC) and, on rare occasions, HSCR. Thus, while HSCR family members may be at risk for developing neuroendocrine tumors, it follows that identical mutations in RET may be able to participate in the pathogenesis of distinct phenotypes. Our data suggest that: (i) the overall frequency of RET mutations in HSCR patients is low and therefore, other genetic and/or environmental determinants contribute to the majority of HSCR susceptibility, and (ii) at present, there is no obvious relationship between RET genotype and HSCR phenotype.


Assuntos
Proteínas de Drosophila , Doença de Hirschsprung/enzimologia , Doença de Hirschsprung/genética , Mutação , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Sequência de Bases , DNA/genética , Análise Mutacional de DNA , Éxons , Feminino , Genótipo , Doença de Hirschsprung/etiologia , Humanos , Masculino , Dados de Sequência Molecular , Neoplasia Endócrina Múltipla Tipo 2a/genética , Linhagem , Fenótipo , Reação em Cadeia da Polimerase , Polimorfismo Genético , Proteínas Proto-Oncogênicas c-ret
13.
Hum Mol Genet ; 3(8): 1217-25, 1994 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7987295

RESUMO

Hirschsprung disease (HSCR) is a congenital disorder of unknown etiology characterized by the absence of enteric ganglia in the distal colon. We have ascertained a large, inbred, Mennonite kindred which demonstrates a high incidence of Hirschsprung disease (HSCR). Genealogical analysis of all kinship relationships identified a single common ancestral couple for all parents of affected offspring. Segregation analysis yielded a segregation ratio of 10.67% for males and 5.45% for females. We searched for locations of the gene(s) responsible for HSCR in this pedigree by genotyping three small multicase families and locating genomic regions demonstrating identity-by-descent followed by linkage disequilibrium analysis of 28 additional nuclear families. Based on this novel strategy, we report the mapping of a new locus for HSCR to chromosome 13q22. Nine microsatellite markers spanning 10 cM in this region were genotyped on thirty-one nuclear families. Significant nonrandom association was detected with alleles at markers D13S162, D13S160, D13S170, and AFM240zg9. In addition, our studies reveal preliminary evidence for a genetic modifier of HSCR in this kindred on chromosome 21q22.


Assuntos
Mapeamento Cromossômico , Cromossomos Humanos Par 13 , Genes Recessivos/genética , Doença de Hirschsprung/genética , Feminino , Haplótipos , Humanos , Masculino , Linhagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA