Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Fam Cancer ; 15(4): 563-70, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26880076

RESUMO

Familial adenomatous polyposis (FAP) is a dominantly inherited syndrome caused by germline mutations in the APC gene and characterized by the development of multiple colorectal adenomas and a high risk of developing colorectal cancer (CRC). The severity of polyposis is correlated with the site of the APC mutation. However, there is also phenotypic variability within families with the same underlying APC mutation, suggesting that additional factors influence the severity of polyposis. Genome-wide association studies identified several single nucleotide polymorphisms (SNPs) that are associated with CRC. We assessed whether these SNPs are associated with polyp multiplicity in proven APC mutation carriers. Sixteen CRC-associated SNPs were analysed in a cohort of 419 APC germline mutation carriers from 182 families. Clinical data were retrieved from the Dutch Polyposis Registry. Allele frequencies of the SNPs were compared for patients with <100 colorectal adenomas versus patients with ≥100 adenomas, using generalized estimating equations with the APC genotype as a covariate. We found a trend of association of two of the tested SNPs with the ≥100 adenoma phenotype: the C alleles of rs16892766 at 8q23.3 (OR 1.71, 95 % CI 1.05-2.76, p = 0.03, dominant model) and rs3802842 at 11q23.1 (OR 1.51, 95 % CI 1.03-2.22, p = 0.04, dominant model). We identified two risk variants that are associated with a more severe phenotype in APC mutation carriers. These risk variants may partly explain the phenotypic variability in families with the same APC gene defect. Further studies with a larger sample size are recommended to evaluate and confirm the phenotypic effect of these SNPs in FAP.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Cromossomos Humanos Par 11 , Cromossomos Humanos Par 8 , Neoplasias Colorretais/genética , Adenoma/genética , Polipose Adenomatosa do Colo/genética , Adulto , Frequência do Gene , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos , Masculino , Mutação , Polimorfismo de Nucleotídeo Único
2.
Oncogene ; 25(13): 1841-51, 2006 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-16288217

RESUMO

The Smad4(+/E6sad) mouse carries a null mutation in the endogenous Smad4 gene resulting in serrated adenomas and mixed polyposis of the upper gastrointestinal (GI) tract with 100% penetrance. Here, we show by loss of heterozygosity (LOH) analysis and immunohistochemistry (IHC) that, although the majority of the tumors appear at 9 months of age, somatic loss of the wild-type Smad4 allele occurs only at later stages of tumor progression. Hence, haploinsufficiency underlies Smad4-driven tumor initiation in the GI tract. As both the Apc and Smad4 tumor suppressor genes map to mouse chromosome 18, we have bred Smad4(+/E6sad) with the Apc(+/1638N) model to generate two distinct compound heterozygous lines carrying both mutations either in cis (CAS) or in trans (TAS). Strikingly, both models show increased tumor multiplicities when compared with the single mutant littermates, although CAS mice are more severely affected and became moribund at only 5-6 weeks of age. Phenotypic and molecular analyses indicate that Smad4 haploinsufficiency is sufficient to significantly affect tumor initiation and progression both prior to and upon loss of Apc function. Moreover, complete loss of Smad4 strongly enhances Apc-driven tumor formation.


Assuntos
Genes APC , Neoplasias Intestinais/genética , Perda de Heterozigosidade , Proteína Smad4/genética , Idade de Início , Animais , Análise Mutacional de DNA , Progressão da Doença , Imuno-Histoquímica , Neoplasias Intestinais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Transdução de Sinais , Fator de Crescimento Transformador beta/fisiologia , Proteínas Wnt/fisiologia
3.
Oncogene ; 20(4): 451-60, 2001 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-11313976

RESUMO

Aggressive fibromatosis is a locally invasive soft tissue lesion. Seventy-five per cent of cases harbor a somatic mutation in either the APC or beta-catenin genes, resulting in beta-catenin protein stabilization. Cyclooxygenase-2 (COX-2) is an enzyme involved in prostaglandin synthesis that modulates the formation of colonic neoplasia, especially in cases due to mutations resulting in beta-catenin stabilization. Human aggressive fibromatoses and lesions from the Apc+/Apc1638N mouse (a murine model for Apc-driven fibromatosis) demonstrated elevated COX-2 levels. COX-2 blockade either by the selective agent DFU or by non-selective COX blocking agents results in reduced proliferation in human tumor cell cultures. Breeding mice with Cox-2-/- mice resulted in no difference in number of aggressive fibromatoses formed, but in a smaller tumor size, while there was a decrease in number of GI lesions by 50%. Mice fed various COX blocking agents also showed a decline in tumor size. COX-2 expression was regulated by tcf-dependent transcription in this lesion. COX-2 partially regulates proliferation due to beta-catenin stabilization in aggressive fibromatosis. Although COX blockade alone does not cause tumor regression, this data suggests that it may have a role as an adjuvant therapy to slow tumor growth in this lesion.


Assuntos
Fibromatose Agressiva/etiologia , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Transativadores , Animais , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Proteínas do Citoesqueleto/isolamento & purificação , Fibromatose Agressiva/patologia , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Prostaglandina-Endoperóxido Sintases/genética , Receptores Citoplasmáticos e Nucleares/isolamento & purificação , Fatores de Transcrição/isolamento & purificação , beta Catenina
4.
Gastroenterology ; 119(4): 1045-53, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11040191

RESUMO

BACKGROUND & AIMS: Inactivation of the adenomatous polyposis coli (APC) gene is observed at early stages of intestinal tumor formation, whereas loss of E-cadherin is usually associated with tumor progression. Because both proteins compete for the binding to beta-catenin, an essential component of the Wnt signaling pathway, reduction of E-cadherin levels in an Apc mouse model could influence both tumor initiation and progression. In addition, loss or haploinsufficiency of E-cadherin may affect tumorigenesis by altering its cell-adhesive and associated functions. METHODS: Apc1638N mice were bred with animals carrying a targeted E-cadherin knockout mutation. RESULTS: Double heterozygous animals showed a significant 9-fold and 5-fold increase of intestinal and gastric tumor numbers, respectively, compared with Apc1638N animals. The intestinal tumors of both groups showed no significant differences in grading and staging. Loss of heterozygosity analysis at the Apc and E-cadherin loci in both intestinal and gastric Apc(+/1638N)/E-cad(+/-) tumors revealed loss of the wild-type Apc allele in most cases, whereas the wild-type E-cadherin allele was always retained. This was supported by a positive, although reduced, staining for E-cadherin of intestinal tumor sections. CONCLUSIONS: Introduction of the E-cadherin mutation in Apc1638N animals enhances Apc-driven tumor initiation without clearly affecting tumor progression.


Assuntos
Caderinas/genética , Proteínas do Citoesqueleto/genética , Mucosa Gástrica/patologia , Genes APC , Mucosa Intestinal/patologia , Neoplasias Intestinais/genética , Perda de Heterozigosidade , Neoplasias Gástricas/genética , Proteína da Polipose Adenomatosa do Colo , Alelos , Animais , Caderinas/análise , Caderinas/fisiologia , Mapeamento Cromossômico , Proteínas do Citoesqueleto/deficiência , Proteínas do Citoesqueleto/fisiologia , Modelos Animais de Doenças , Feminino , Heterozigoto , Neoplasias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Neoplasias Gástricas/patologia
5.
Genes Chromosomes Cancer ; 29(3): 229-39, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10992298

RESUMO

The APC gene, originally identified as the gene for familial adenomatous polyposis (FAP), is now considered as the true "gatekeeper" of colonic epithelial proliferation. Its main tumor suppressing activity seems to reside in the capacity to properly regulate intracellular beta-catenin signaling. Most somatic APC mutations are detected between codons 1286 and 1513, the mutation cluster region (MCR). This clustering can be explained either by the presence of mutation-prone sequences within the MCR, or by the selective advantage provided by the resulting truncated polypeptides. Here, a Msh2-deficient mouse model (Msh2(delta 7N) ) was generated and bred with Apc(1638N) and Apc(Min) that allowed the comparison of the somatic mutation spectra along the Apc gene in the different allelic combinations. Mutations identified in Msh2(delta 7N/delta 7N) tumors are predominantly dinucleotide deletions at simple sequence repeats leading to truncated Apc polypeptides that partially retain the 20 a.a. beta-catenin downregulating motifs. In contrast, the somatic mutations identified in the wild type Apc allele of Msh2(delta 7N/delta 7N) /Apc(+/1638N) and Msh2(delta 7N/delta 7N) /Apc(+/Min) tumors are clustered more to the 5' end, thereby completely inactivating the beta-catenin downregulating activity of APC. These results indicate that somatic Apc mutations are selected during intestinal tumorigenesis and that inactivation of the beta-catenin downregulating function of APC is likely to represent the main selective factor.


Assuntos
Polipose Adenomatosa do Colo/genética , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA , Regulação para Baixo/genética , Genes APC/genética , Mutação/genética , Transativadores , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , beta Catenina
6.
Genes Dev ; 13(10): 1309-21, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10346819

RESUMO

The adenomatous polyposis coli (APC) gene is considered as the true gatekeeper of colonic epithelial proliferation: It is mutated in the majority of colorectal tumors, and mutations occur at early stages of tumor development in mouse and man. These mutant proteins lack most of the seven 20-amino-acid repeats and all SAMP motifs that have been associated with down-regulation of intracellular beta-catenin levels. In addition, they lack the carboxy-terminal domains that bind to DLG, EB1, and microtubulin. APC also appears to be essential in development because homozygosity for mouse Apc mutations invariably results in early embryonic lethality. Here, we describe the generation of a mouse model carrying a targeted mutation at codon 1638 of the mouse Apc gene, Apc1638T, resulting in a truncated Apc protein encompassing three of the seven 20 amino acid repeats and one SAMP motif, but missing all of the carboxy-terminal domains thought to be associated with tumorigenesis. Surprisingly, homozygosity for the Apc1638T mutation is compatible with postnatal life. However, homozygous mutant animals are characterized by growth retardation, a reduced postnatal viability on the B6 genetic background, the absence of preputial glands, and the formation of nipple-associated cysts. Most importantly, Apc1638T/1638T animals that survive to adulthood are tumor free. Although the full complement of Apc1638T is sufficient for proper beta-catenin signaling, dosage reductions of the truncated protein result in increasingly severe defects in beta-catenin regulation. The SAMP motif retained in Apc1638T also appears to be important for this function as shown by analysis of the Apc1572T protein in which its targeted deletion results in a further reduction in the ability of properly controlling beta-catenin/Tcf signaling. These results indicate that the association with DLG, EB1, and microtubulin is less critical for the maintenance of homeostasis by APC than has been suggested previously, and that proper beta-catenin regulation by APC appears to be required for normal embryonic development and tumor suppression.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Proteínas de Drosophila , Neoplasias Experimentais/etiologia , Transativadores , Proteínas Supressoras de Tumor , Proteína da Polipose Adenomatosa do Colo , Fatores Etários , Animais , Peso Corporal , Cistos/genética , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/química , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Genitália Masculina/anatomia & histologia , Genitália Masculina/embriologia , Proteínas de Insetos/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Mutagênese Insercional , Fenótipo , Glândulas Sebáceas/fisiologia , Dermatopatias/genética , Células-Tronco/metabolismo , Tubulina (Proteína)/análise , beta Catenina
7.
Oncogene ; 18(2): 543-50, 1999 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-9927211

RESUMO

The inv(16) and related t(16;16) are found in 10% of all cases with de novo acute myeloid leukemia. In these rearrangements the core binding factor beta (CBFB) gene on 16q22 is fused to the smooth muscle myosin heavy chain gene (MYH11) on 16p13. To gain insight into the mechanisms causing the inv(16) we have analysed 24 genomic CBFB-MYH11 breakpoints. All breakpoints in CBFB are located in a 15-Kb intron. More than 50% of the sequenced 6.2 Kb of this intron consists of human repetitive elements. Twenty-one of the 24 breakpoints in MYH11 are located in a 370-bp intron. The remaining three breakpoints in MYH11 are located more upstream. The localization of three breakpoints adjacent to a V(D)J recombinase signal sequence in MYH11 suggests a V(D)J recombinase-mediated rearrangement in these cases. V(D)J recombinase-associated characteristics (small nucleotide deletions and insertions of random nucleotides) were detected in six other cases. CBFB and MYH11 duplications were detected in four of six cases tested.


Assuntos
Inversão Cromossômica , Cromossomos Humanos Par 16 , Leucemia Mieloide/genética , Doença Aguda , Sequência de Bases , Clonagem Molecular , Subunidade beta de Fator de Ligação ao Core , DNA Complementar , Proteínas de Ligação a DNA/genética , Humanos , Íntrons , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Fator de Transcrição AP-2 , Fatores de Transcrição/genética
8.
Gastroenterology ; 114(2): 275-83, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9453487

RESUMO

BACKGROUND & AIMS: Germline mutations in the adenomatous polyposis coli (APC) gene are responsible for familial adenomatous polyposis (FAP), an autosomal dominant predisposition to the formation of multiple colorectal adenomas. Moreover, patients with FAP are at high risk of developing several extracolonic manifestations, including desmoids, cutaneous cysts, and tumors of the upper gastrointestinal tract. Although by definition desmoids are nonmalignant, because of their aggressive invasion of local structures, they represent one of the major causes of morbidity and mortality among patients with FAP. METHODS: This study describes the histopathologic and molecular characterization of Apc1638N, a mouse model for the broad spectrum of extracolonic manifestations characteristic of FAP. RESULTS: Heterozygous Apc+/Apc1638N animals develop fully penetrant and multifocal cutaneous follicular cysts and desmoid tumors in addition to attenuated polyposis of the upper gastrointestinal tract. Moreover, breeding of Apc+/Apc1638N mice in a p53-deficient background results in a dramatic seven-fold increase of the desmoid multiplicity. CONCLUSIONS: Because of the attenuated nature of their intestinal phenotype, these mice survive longer than other murine models for Apc-driven tumorigenesis. Therefore, Apc1638N represents an ideal laboratory tool to test various therapeutic intervention strategies for the management of intestinal as well as extraintestinal tumors.


Assuntos
Polipose Adenomatosa do Colo/complicações , Cistos/etiologia , Modelos Animais de Doenças , Fibromatose Agressiva/etiologia , Dermatopatias/etiologia , Polipose Adenomatosa do Colo/genética , Idade de Início , Animais , Cistos/patologia , Feminino , Fibromatose Agressiva/patologia , Genes APC/genética , Genes p53/genética , Perda de Heterozigosidade , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mutação/fisiologia , Neoplasias Primárias Múltiplas/complicações , Fenótipo , Distribuição por Sexo , Dermatopatias/patologia
9.
Carcinogenesis ; 18(2): 321-7, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9054624

RESUMO

The Apc1638N mouse carries a targeted mutant allele at the endogenous adenomatous polyposis coli (Apc) gene and represents a unique in vivo model to study intestinal tumor formation and progression. Heterozygous Apc+/Apc1638N mice progressively develop 5-6 adenomas and adenocarcinomas of the small intestine within the first 6 months of life following a histologic sequence similar to that observed in human intestinal tumors. Here, we present the somatic mutation analysis of a total of 57 tumors. The results indicate that in > or = 75% of the lesions tested the wild type copy of the Apc gene is lost and that this LOH event extends to the entire mouse chromosome 18. Unexpectedly, mutations at the K-, N- and H-ras genes have not been found in these tumors. Immunohistochemical analysis of the Apc1638N tumors failed to detect accumulation of the Tp53 protein. Also, no mutations have been found in exons 7 and 8 of the Tp53 gene. These results indicate that, although the genetic inactivation of Apc is involved in the initiating event of the human as well as murine intestinal tumorigenesis, tumor growth and progression follow different mutational pathways in these two species.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 18 , Genes APC , Genes p53/genética , Genes ras/genética , Neoplasias Intestinais/genética , Intestino Delgado , Adenocarcinoma/genética , Adenoma/genética , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA