Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Clin Genet ; 93(1): 52-59, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28589637

RESUMEN

To establish whether existing mutation prediction models can identify which male breast cancer (MBC) patients should be offered BRCA1 and BRCA2 diagnostic DNA screening, we compared the performance of BOADICEA (Breast and Ovarian Analysis of Disease Incidence and Carrier Estimation Algorithm), BRCAPRO (BRCA probability) and the Myriad prevalence table ("Myriad"). These models were evaluated using the family data of 307 Dutch MBC probands tested for BRCA1/2, 58 (19%) of whom were carriers. We compared the numbers of observed vs predicted carriers and assessed the Area Under the Receiver Operating Characteristic (ROC) Curve (AUC) for each model. BOADICEA predicted the total number of BRCA1/2 mutation carriers quite accurately (observed/predicted ratio: 0.94). When a cut-off of 10% and 20% prior probability was used, BRCAPRO showed a non-significant better performance (observed/predicted ratio BOADICEA: 0.81, 95% confidence interval [CI]: [0.60-1.09] and 0.79, 95% CI: [0.57-1.09], vs. BRCAPRO: 1.02, 95% CI: [0.75-1.38] and 0.94, 95% CI: [0.68-1.31], respectively). Myriad underestimated the number of carriers in up to 69% of the cases. BRCAPRO showed a non-significant, higher AUC than BOADICEA (0.798 vs 0.776). Myriad showed a significantly lower AUC (0.671). BRCAPRO and BOADICEA can efficiently identify MBC patients as BRCA1/2 mutation carriers. Besides their general applicability, these tools will be of particular value in countries with limited healthcare resources.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Neoplasias de la Mama Masculina/genética , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas/métodos , Mutación , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Neoplasias de la Mama Masculina/diagnóstico , Estudios de Cohortes , Femenino , Frecuencia de los Genes , Heterocigoto , Humanos , Masculino , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/genética , Curva ROC
2.
Fam Cancer ; 15(4): 563-70, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26880076

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 8 , Neoplasias Colorrectales/genética , Adenoma/genética , Poliposis Adenomatosa del Colon/genética , Adulto , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple
3.
Breast Cancer Res Treat ; 133(1): 393-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22370629

RESUMEN

Despite extensive analysis of the BRCA1 and BRCA2 genes, germline mutations are detected in <20% of families with a presumed genetic predisposition for breast and ovarian cancer. Recent literature reported RAD51C as a new breast cancer susceptibility gene. In this study, we report the analysis of 410 patients from 351 unrelated pedigrees. All were referred for genetic testing and we selected families with at least one reported case of ovarian cancer in which BRCA1&2 mutations were previously ruled out. We analyzed the coding exons, intron-exons boundaries, and UTRs of RAD51C. Our mutation analysis did not reveal any unequivocal deleterious mutation. In total 12 unique sequence variations were identified of which two were novel. Our study and others suggest a low prevalence of RAD51C mutations with an exception for some founder populations. This observation is in favor of the rare allele hypothesis in the debate over the nature of the genetic contribution to individual susceptibility to breast and ovarian cancer and further genome-wide studies in high risk families are warranted.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Humanos , Masculino , Mutación Missense , Linaje , Polimorfismo de Nucleótido Simple
4.
Clin Genet ; 81(2): 179-84, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21204799

RESUMEN

Founder mutations in BRCA1 and BRCA2 have been reported in many different populations. We studied 105 Coloured and 16 Black Xhosa women residing in the Western Cape of South Africa diagnosed with breast cancer. We screened these patients using our standard panel of six previously reported SA Afrikaner and Ashkenazi Jewish BRCA1/2 mutations and identified only two Afrikaner mutations. Further screening by the protein truncation test (BRCA1 exon 11, and BRCA2 exons 10 and 11) revealed an additional four deleterious mutations (BRCA1 c.1504_ 1508del,p.Leu502AlafsX2, BRCA2 c.2826_2829del,p.Ile943LysfsX16, c.6447_6448dup,p.Lys2150IlefsX19 and c.5771_5774del,p.Ile1924Argfs X38). The latter, also known in Breast Cancer Information Core nomenclature as 5999del4, was identified in 4 of 105 (3.8%) Coloureds and 4 of 16 (25%) Xhosa women, which makes it a frequent founder mutation in the Western Cape Province. Although this mutation was previously reported to occur in the Netherlands, haplotype analysis indicated two distinct origins for the Dutch and South African mutations, excluding the possibility of a common Dutch ancestor and suggesting gene flow from the indigenous tribes such as the Xhosa to the Coloured population instead. Further studies to determine the carrier rate of this variant in the Xhosa and other SA populations are warranted.


Asunto(s)
Proteína BRCA2/genética , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Efecto Fundador , Mutación , Adulto , Anciano , Proteína BRCA1/genética , Neoplasias de la Mama/patología , Exones , Femenino , Haplotipos , Humanos , Persona de Mediana Edad , Estadificación de Neoplasias , Prevalencia , Sudáfrica/epidemiología , Sudáfrica/etnología
5.
Clin Genet ; 80(3): 243-55, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21261604

RESUMEN

Childhood brain tumours may be due to germline bi-allelic mismatch repair (MMR) gene mutations in MLH1, MSH2, MSH6 or PMS2. These mutations can also lead to colorectal neoplasia and haematological malignancies. Here, we review this syndrome and present siblings with early-onset rectal adenoma and papillary glioneural brain tumour, respectively, due to novel germline bi-allelic PMS2 mutations. Identification of MMR protein defects can lead to early diagnosis of this condition. In addition, assays for these defects may help to classify brain tumours for research protocols aimed at targeted therapies.


Asunto(s)
Adenoma/genética , Adenosina Trifosfatasas , Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , Enzimas Reparadoras del ADN , Proteínas de Unión al ADN , Mutación de Línea Germinal , Glioma/genética , Adenoma/diagnóstico , Adenoma/patología , Adenosina Trifosfatasas/genética , Edad de Inicio , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Niño , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Glioma/diagnóstico , Glioma/patología , Heterocigoto , Humanos , Masculino , Repeticiones de Microsatélite , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Linaje , Hermanos , Síndrome , Adulto Joven
6.
Leukemia ; 24(8): 1462-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20535148

RESUMEN

Relapse of pediatric acute lymphoblastic leukemia (ALL) remains the main cause of treatment failure after allogeneic stem cell transplantation (alloSCT). A high level of minimal residual disease (MRD) before alloSCT has been shown to predict these relapses. Patients at risk might benefit from a preemptive alloimmune intervention. In this first prospective, MRD-guided intervention study, 48 patients were stratified according to pre-SCT MRD level. Eighteen children with MRD level >or=1 x 10(-4) were eligible for intervention, consisting of early cyclosporine A tapering followed by consecutive, incremental donor lymphocyte infusions (n=1-4). The intervention was associated with graft versus host disease >or=grade II in only 23% of patients. Event-free survival in the intervention group was 19%. However, in contrast with the usual early recurrence of leukemia, relapses were delayed up to 3 years after SCT. In addition, several relapses presented at unusual extramedullary sites suggesting that the immune intervention may have altered the pattern of leukemia recurrence. In 8 out of 11 evaluable patients, relapse was preceded by MRD recurrence (median 9 weeks, range 0-30). We conclude that in children with high-risk ALL, immunotherapy-based regimens after SCT are feasible and may need to be further intensified to achieve total eradication of residual leukemic cells.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Neoplasia Residual , Leucemia-Linfoma Linfoblástico de Células Precursoras/cirugía , Niño , Preescolar , Femenino , Humanos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Riesgo
7.
Gut ; 59(7): 975-86, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581245

RESUMEN

Peutz-Jeghers syndrome (PJS, MIM175200) is an autosomal dominant condition defined by the development of characteristic polyps throughout the gastrointestinal tract and mucocutaneous pigmentation. The majority of patients that meet the clinical diagnostic criteria have a causative mutation in the STK11 gene, which is located at 19p13.3. The cancer risks in this condition are substantial, particularly for breast and gastrointestinal cancer, although ascertainment and publication bias may have led to overestimates in some publications. Current surveillance protocols are controversial and not evidence-based, due to the relative rarity of the condition. Initially, endoscopies are more likely to be done to detect polyps that may be a risk for future intussusception or obstruction rather than cancers, but surveillance for the various cancers for which these patients are susceptible is an important part of their later management. This review assesses the current literature on the clinical features and management of the condition, genotype-phenotype studies, and suggested guidelines for surveillance and management of individuals with PJS. The proposed guidelines contained in this article have been produced as a consensus statement on behalf of a group of European experts who met in Mallorca in 2007 and who have produced guidelines on the clinical management of Lynch syndrome and familial adenomatous polyposis.


Asunto(s)
Síndrome de Peutz-Jeghers/diagnóstico , Adulto , Anciano , Neoplasias de la Mama/diagnóstico , Niño , Preescolar , Endoscopía Gastrointestinal , Medicina Basada en la Evidencia/métodos , Femenino , Neoplasias Gastrointestinales/diagnóstico , Neoplasias de los Genitales Femeninos/diagnóstico , Genotipo , Humanos , Cuidados a Largo Plazo/métodos , Masculino , Tamizaje Masivo/métodos , Persona de Mediana Edad , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/terapia , Fenotipo , Vigilancia de la Población/métodos , Adulto Joven
8.
Br J Cancer ; 102(2): 447-54, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-19920828

RESUMEN

It is now recognised that a part of the inherited risk of colorectal cancer (CRC) can be explained by the co-inheritance of low-penetrance genetic variants. The accumulated experience to date in identifying these variants has served to highlight difficulties in conducting statistically and methodologically rigorous studies and follow-up analyses. The COGENT (COlorectal cancer GENeTics) consortium includes 20 research groups in Europe, Australia, the Americas, China and Japan. The overarching goal of COGENT is to identify and characterise low-penetrance susceptibility variants for CRC through association-based analyses. In this study, we review the rationale for identifying low-penetrance variants for CRC and our proposed strategy for establishing COGENT.


Asunto(s)
Neoplasias Colorrectales/genética , Polimorfismo Genético , Predisposición Genética a la Enfermedad , Humanos , Penetrancia , Pronóstico , Riesgo , Factores de Riesgo
9.
Gut ; 57(1): 71-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17604324

RESUMEN

BACKGROUND: The patient with 10 or more adenomas in the colon poses a diagnostic challenge. Beside germline mutations in the APC and MUTYH genes, only four cases of mosaic APC mutations have been reported. AIM: Given the relatively high frequency of de novo APC mutations in familial adenomatous polyposis (FAP), an investigation was carried out into whether the proportion of somatic mosaic APC mutations is currently underestimated. METHODS: Between 1 January 1994 and 31 December 2005 germline mutation analysis was performed in 599 consecutive index patients with polyposis coli referred for diagnostic APC scanning using a combination of denaturing gradient gel electrophoresis (DGGE) and protein truncation test (PTT). Variants were analysed by direct sequencing with primers flanking those used for DGGE and PTT, and quantified using pyrosequencing. RESULTS: Scrutinizing the molecular genetic results and family data of 242 index patients with pathogenic APC mutations led to the identification of 10 mosaic cases (4%). C>T transitions were observed in CGA sites in four of the 10 cases with somatic mosaicism, which is significantly more than 26 of the 232 non-mosaic cases (p = 0.02). Phenotypes of patients with somatic mosaicism ranged from an attenuated form of polyposis coli to florid polyposis with major extracolonic manifestations. CONCLUSIONS: Mosaicism occurs in a significant number of APC mutations and it is estimated that one-fifth of the de novo cases of FAP are mosaic. Clinically, the severity of manifestations in offspring and the recurrence risk for siblings of apparently sporadic polyposis patients may be underestimated due to parental APC mosaicism.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Genes APC , Mosaicismo , Adulto , Anciano , Estudios de Cohortes , Femenino , Genotipo , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo
10.
Clin Genet ; 71(5): 427-33, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17489848

RESUMEN

A small fraction of families with familial adenomatous polyposis (FAP) display an attenuated form of FAP (AFAP). We aimed to assess the presence of germline mutations in the MUTYH and adenomatous polyposis coli (APC) genes in AFAP families and to compare the clinical features between the two causative genes. Families with clinical AFAP were selected from the Dutch Polyposis Registry according to the following criteria: (a) at least two patients with 10-99 adenomas diagnosed at age >30 years or (b) one patient with 10-99 adenomas at age >30 years and a first-degree relative with colorectal cancer (CRC) with a few adenomas, and, applying for both criteria, no family members with more than 100 polyps before the age of 30 years. All probands were screened for germline mutations in the APC and MUTYH genes. Twenty-five of 315 Dutch families with FAP (8%) met our criteria for AFAP. These families included 146 patients with adenomas and/or CRC. Germline APC mutations were identified in nine families and biallelic MUTYH mutations in another nine families. CRC was identified at a mean age of 54 years (range 24-83 years) in families with APC and at 50 years (range 39-70 years) in families with MUTYH (p = 0.29). APC and biallelic MUTYH mutations are responsible for the majority of AFAP families. Based on our results and those reported in the literature, we recommend colonoscopy once every 2 years in AFAP families, starting surveillance from the late teens in APC mutation carriers and from age 20-25 years in biallelic MUTYH mutation carriers.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , ADN Glicosilasas/genética , Genes APC , Mutación de Línea Germinal , Poliposis Adenomatosa del Colon/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Mutación , Países Bajos , Sistema de Registros
11.
J Med Genet ; 42(9): e54, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16140997

RESUMEN

OBJECTIVE: To investigate the contribution of MYH associated polyposis coli (MAP) among polyposis families in the Netherlands, and the prevalence of colonic and extracolonic manifestations in MAP patients. METHODS: 170 patients with polyposis coli, who previously tested negative for APC mutations, were screened by denaturing gradient gel electrophoresis and direct sequencing to identify MYH germline mutations. RESULTS: Homozygous and compound heterozygous MYH mutations were identified in 40 patients (24%). No difference was found in the percentage of biallelic mutation carriers between patients with 10-99 polyps or 100-1000 polyps (29% in both groups). Colorectal cancer was found in 26 of the 40 patients with MAP (65%) within the age range 21 to 67 years (median 45). Complete endoscopic reports were available for 16 MAP patients and revealed five cases with gastro-duodenal polyps (31%), one of whom also presented with a duodenal carcinoma. Breast cancer occurred in 18% of female MAP patients, significantly more than expected from national statistics (standardised morbidity ratio = 3.75). CONCLUSIONS: Polyp numbers in MAP patients were equally associated with the attenuated and classical polyposis coli phenotypes. Two thirds of the MAP patients had colorectal cancer, 95% of whom were older than 35 years, and one third of a subset of patients had upper gastrointestinal lesions. Endoscopic screening of the whole intestine should be carried out every two years for all MAP patients, starting from age 25-30 years. The frequent occurrence of additional extraintestinal manifestations, such as breast cancer among female MAP patients, should be thoroughly investigated.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , ADN Glicosilasas/genética , Adolescente , Adulto , Anciano , Niño , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Mutación de Línea Germinal , Humanos , Patrón de Herencia/genética , Masculino , Persona de Mediana Edad , Países Bajos , Fenotipo , Riesgo
13.
Oncogene ; 20(43): 6241-4, 2001 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-11593433

RESUMEN

Recently, we and others reported instability in the (C)8 repeat in exon 5 of MSH6 as a preferential target for somatic mutations in tumours from MSH6 germline mutation carriers. Here, we report that in 45% of tumours from MLH1, MSH2 and MSH6 germline mutation carriers no sequence change in the (C)8 repeat of MSH6 was found upon DNA sequencing analysis of PCR products with a shift in electrophoresis mobility. Using "standard" PCR primers a high frequency of instability (50-86%) of the (C)8 repeat was found, but using a modified PCR reverse primer, accomplishing modulation of non-templated addition of adenine during in vitro PCR amplification by the Taq polymerase, a markedly lower frequency of instability was found in tumours from MLH1, MSH2 and MSH6 mutation carriers (6, 13 and 40%, respectively). Furthermore, a significant difference of the frequency of instability of the (C)8 repeat in tumours from MSH6 mutation carriers was found compared to MLH1, MSH2 mutation carriers. These results might have important implications for the detection of instability of other short mononucleotide repeats, e.g. TGFbetaRII, BAX, IGFRII, PTEN, BRCA2.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Fúngicas/genética , Proteínas de Saccharomyces cerevisiae , Expansión de Repetición de Trinucleótido , Proteínas Adaptadoras Transductoras de Señales , Sesgo , Proteínas Portadoras , Análisis Mutacional de ADN , Cartilla de ADN/metabolismo , Exones , Eliminación de Gen , Humanos , Repeticiones de Microsatélite , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
14.
J Clin Oncol ; 19(20): 4074-80, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11600610

RESUMEN

PURPOSE: Hereditary nonpolyposis colorectal cancer (HNPCC) is an autosomal dominant disease characterized by the clustering of colorectal cancer, endometrial cancer, and various other cancers. The disease is caused by mutations in DNA-mismatch-repair (MMR) genes, most frequently in MLH1, MSH2, and MSH6. The aims of the present study were to compare the risk of developing colorectal, endometrial, and other cancers between families with the various MMR-gene mutations. PATIENTS AND METHODS: Clinical and pathologic data were collected from 138 families with HNPCC. Mutation analyses were performed for all families. Survival analysis was used to calculate the cumulative risk of developing cancer in the various subsets of relatives. RESULTS: Mutations were identified in 79 families: 34 in MLH1, 40 in MSH2, and five in MSH6. The lifetime risk of developing cancer at any site was significantly higher for MSH2 mutation carriers than for MLH1 mutation carriers (P < .01). The risk of developing colorectal or endometrial cancer was higher in MSH2 mutation carriers than in MLH1 mutation carriers, but the difference was not significant (P = .13 and P = .057, respectively). MSH2 mutation carriers were found to have a significantly higher risk of developing cancer of the urinary tract (P < .05). The risk of developing cancer of the ovaries, stomach, and brain was also higher in the MSH2 mutation carriers than in the MLH1 mutation carriers, but the difference was not statistically significant. CONCLUSION: Pending large prospective studies, the extension of the current surveillance program in MSH2 mutation carriers with the inclusion of the urinary tract should be considered.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Proteínas de Unión al ADN , Mutación , Proteínas de Neoplasias/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Disparidad de Par Base/genética , Proteínas Portadoras , Niño , Preescolar , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Reparación del ADN/genética , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Países Bajos/epidemiología , Proteínas Nucleares , Sistema de Registros , Factores de Riesgo , Factores Sexuales
15.
Ned Tijdschr Geneeskd ; 145(16): 780-2, 2001 Apr 21.
Artículo en Holandés | MEDLINE | ID: mdl-11346916

RESUMEN

Hereditary non-polyposis colorectal cancer (HNPCC), also known as Lynch syndrome, is the most common autosomal dominant condition associated with early-onset colorectal cancer and the occurrence of cancer at other anatomical sites, i.e. endometrium, stomach, small intestine, urinary tract and ovaries, at an early age. Germline mutations in one of five DNA mismatch repair genes: MSH2, MLH1, PMS1, PMS2, and MSH6, predispose to HNPCC. Tumours of HNPCC patients display a high level of genomic instability, usually observed as changes in repeat numbers of simple repetitive sequences (microsatellite instability), which is a reflection of the malfunction of the DNA mismatch repair machinery.


Asunto(s)
Disparidad de Par Base/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación del ADN/genética , ADN de Neoplasias/genética , Mutación de Línea Germinal/genética , Edad de Inicio , Neoplasias del Colon/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Comorbilidad , Neoplasias Endometriales/genética , Femenino , Humanos , Masculino , Repeticiones de Microsatélite/genética , Países Bajos/epidemiología , Neoplasias Ováricas/genética , Neoplasias del Recto/genética , Neoplasias Gástricas/genética , Neoplasias Urológicas/genética
16.
Fam Cancer ; 1(3-4): 169-73, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-14574174

RESUMEN

BACKGROUND AND AIMS: Hereditary non-polyposis colorectal cancer (HNPCC) may be caused by mutations in the mismatch repair (MMR) genes MLH1, MSH2 or MSH6. Family history (Amsterdam criteria) has traditionally been used to select patients for mutation testing. It has been demonstrated that germline mutations in the MMR genes are associated with lack of the corresponding gene product as assessed with immunohistochemistry (IHC) in tumour specimens. The aim of the study was to assess the value of the Amsterdam criteria II and IHC in predicting germline mutations. METHODS: Fifty-six families that were previously tested for MLH1, MSH2 and MSH6 mutations were selected for this study. All pedigrees were extended and verified and the families were scored according to the original (I) and the revised Amsterdam criteria (II). The probabilities for MLH1 and MSH2 mutations were calculated by logistic regression. In addition, all available tumour material from indexed family members was examined by IHC for the presence of the three gene products. RESULTS: Three out of seven (39%) families where the mutation could be identified complied with the Amsterdam criteria I, while all seven (100%) met the Amsterdam criteria II. All families carrying a MLH1 or MSH2 mutation had > 15% calculated probability of finding a mutation. Tumours from all seven mutation carriers lacked the immunohistochemical expression of the corresponding MMR gene. CONCLUSION: The results indicate that the Amsterdam criteria II in combination with immunohistochemistry of the mismatch repair proteins in tumours may be a cost-effective approach to select families for mutation analysis.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/metabolismo , Proteínas de Unión al ADN/metabolismo , Pruebas Genéticas , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adulto , Disparidad de Par Base , Proteínas Portadoras , Reparación del ADN , ADN de Neoplasias , Femenino , Mutación de Línea Germinal , Humanos , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Países Bajos , Proteínas Nucleares , Valor Predictivo de las Pruebas , Sensibilidad y Especificidad
17.
J Pathol ; 192(3): 328-35, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11054716

RESUMEN

Instability of microsatellite repeat sequences has been observed in colorectal carcinomas and in extracolonic malignancies, predominantly endometrial tumours, occurring in the context of hereditary non-polyposis colorectal cancer (HNPCC). Microsatellite instability (MSI) as a feature of human DNA mismatch repair (MMR)-driven tumourigenesis of the uterine mucosa has been studied primarily in sporadic tumours showing predominantly somatic hypermethylation of MLH1. The present study shows that all endometrial carcinomas (n=12) from carriers of MLH1 and MSH2 germline mutations demonstrate an MSI-high phenotype involving all types of repeat markers, while in endometrial carcinomas from MSH6 mutation carriers, only 36% (4 out of 11) demonstrate an MSI-high phenotype. Interestingly, an MSI-high phenotype was found in endometrial hyperplasias from MSH2 mutation carriers, in contrast to hyperplasias from MLH1 mutation carriers, which exhibited an MSI-stable phenotype. Instability of only mononucleotide repeat markers was found in both endometrial carcinomas and hyperplasias from MSH6 mutation carriers. In 29 out of 31 (94%) endometrial tumour foci, combined MSI and immunohistochemical analysis of MLH1, MSH2, and MSH6 could predict the identified germline mutation. The observation of MSI in endometrial hyperplasia and of altered protein staining for the MMR genes supports the idea that inactivation of MMR genes is an early event in endometrial tumourigenesis. A correlation was found between the variation in the extent and level of MSI and the age of onset of carcinoma, suggesting differences in the rate of tumour progression. A high frequency of MSI in hyperplasias, found only in MSH2 mutation carriers, might indicate a more rapid tumour progression, correlating with an earlier age of onset of carcinoma. The present study indicates that assessment of altered protein staining combined with MSI analysis of endometrial tumours might direct the mutational analysis of MMR genes.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación del ADN/genética , Hiperplasia Endometrial/genética , Neoplasias Endometriales/genética , Repeticiones de Microsatélite/genética , Adulto , Edad de Inicio , Neoplasias Colorrectales Hereditarias sin Poliposis/complicaciones , Neoplasias Colorrectales Hereditarias sin Poliposis/metabolismo , Progresión de la Enfermedad , Hiperplasia Endometrial/etiología , Hiperplasia Endometrial/metabolismo , Neoplasias Endometriales/etiología , Neoplasias Endometriales/metabolismo , Femenino , Mutación de Línea Germinal , Humanos , Persona de Mediana Edad , Fenotipo , Valor Predictivo de las Pruebas
18.
Blood ; 94(9): 2955-62, 1999 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10556177

RESUMEN

We have investigated the blood cells from a woman with a low degree of chronic nonspherocytic hemolytic anemia and frequent bacterial infections accompanied by icterus and anemia. The activity of glucose 6-phosphate dehydrogenase (G6PD) in her red blood cells (RBCs) was below detection level, and in her leukocytes less than 3% of normal. In cultured skin fibroblasts, G6PD activity was approximately 15% of normal, with 4- to 5-fold increased Michaelis constant (Km) for NADP and for glucose 6-phosphate. Activated neutrophils showed a decreased respiratory burst. Family studies showed normal G6PD activity in the RBCs from all family members, including both parents and the 2 daughters of the patient. Sequencing of polymerase chain reaction (PCR)-amplified genomic DNA showed a novel, heterozygous 514C-->T mutation, predicting a Pro172-->Ser replacement. Analysis of G6PD RNA from the patient's leukocytes and fibroblasts showed only transcripts with the 514C-->T mutation. This was explained by the pattern of X-chromosome inactivation, studied by means of the human androgen receptor (HUMARA) assay, which proved to be skewed in the patient, her mother, and one of the patient's daughters. Thus, the patient has inherited a de novo mutation in G6PD from her father and an X-chromosome inactivation determinant from her mother, causing exclusive expression of the mutated G6PD allele. Purified mutant protein from an Escherichia coli expression system showed strongly decreased specific activity, increased Km for NADP and for glucose 6-phosphate, and increased heat lability, which indicates that the defective phenotype is due to 2 synergistic molecular dysfunctions: decreased catalytic efficiency and protein instability.


Asunto(s)
Anemia Hemolítica/genética , Glucosafosfato Deshidrogenasa/genética , Granulocitos/fisiología , Adulto , Anemia Hemolítica/complicaciones , Anemia Hemolítica/enzimología , Anemia Hemolítica/fisiopatología , Enfermedad Crónica , Enfermedades Transmisibles/etiología , Enfermedades Transmisibles/genética , Activación Enzimática , Femenino , Predisposición Genética a la Enfermedad , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Mutación , Linaje , Reacción en Cadena de la Polimerasa
19.
Int J Cancer ; 82(4): 516-9, 1999 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-10404064

RESUMEN

Hereditary nonpolyposis colorectal cancer (Lynch syndrome) is an autosomal dominant disease caused by mutations in the mismatch repair genes in particular in MLH1, MSH2 and MSH6. The disease is characterized by the development of colorectal, endometrial cancer and several other cancers. There is evidence that the clinical expression of the disease varies from one country to another. This variation might affect not only the application of criteria proposed to identify families but also clinical risk factors reported to predict the outcome of genetic testing. Data on site of the cancer, age at diagnosis and pathology were collected from 155 families with suspected HNPCC known at the Korean and Dutch HNPCC registries. DGGE, SSCP and DNA-sequencing were performed to identify MSH2, MLH1 and MSH6 mutations. A total of 33 Korean and 42 Dutch families met the clinical criteria for HNPCC. Germline mutations in the MMR-genes were found in 23 Korean and 24 Dutch families. In families that met the Amsterdam criteria, and also in those associated with MLH1 mutations, more cancers of the stomach and pancreas were observed in the Korean families than in the Dutch HNPCC families; in relative terms, the incidence of cancers of the endometrium in the Korean families was lower. Multivariate analysis showed that an early age at diagnosis, and the occurrence of pancreatic cancer were independent predictive factors of germline mutations in MLH1, MSH2 and MSH6 in the Korean subset of families.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Proteínas de Unión al ADN/genética , Mutación de Línea Germinal , Proteínas de Neoplasias/genética , Proteínas/genética , Proteínas Proto-Oncogénicas/genética , Adulto , Análisis de Varianza , Neoplasias Colorrectales Hereditarias sin Poliposis/etnología , Femenino , Humanos , Corea (Geográfico)/etnología , Masculino , Persona de Mediana Edad , Proteína 2 Homóloga a MutS , Países Bajos/etnología
20.
Ned Tijdschr Geneeskd ; 143(23): 1201-6, 1999 Jun 05.
Artículo en Holandés | MEDLINE | ID: mdl-10389534

RESUMEN

About 5% of colorectal cancer cases are due to an autosomal dominant genetic predisposition with high penetrance. In this condition, the patient is carrier of a pathogenic gene mutation present in all body cells which can be transmitted to descendants, a so-called germ line mutation. The mutation is usually present in a tumour suppressor gene. Three subgroups of hereditary colorectal cancer can be distinguished on the basis of the clinical characteristics: (a) syndromes without polyposis (mostly hereditary non-polyposis colorectal carcinoma; HNPCC), (b) syndromes with adenomatous polyposis (mostly familial adenomatous polyposis; FAP) and (c) syndromes with hamartomatous polyposis. Recently, the main gene defects which underlie these syndromes were identified. Consequently, it is possible in approximately half the families with HNPCC or FAP in patients with colorectal cancer to demonstrate the causative gene defect and subsequently, by blood testing of healthy relatives to determine who is and is not a carrier of this hereditary condition. Thus, preventive measures can be directed toward family members with a demonstrable high risk of large bowel cancer.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Primarias Múltiples/genética , Síndrome de Peutz-Jeghers/genética , Poliposis Adenomatosa del Colon/epidemiología , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/cirugía , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/genética , Niño , Preescolar , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Femenino , Genes APC/genética , Tamización de Portadores Genéticos , Mutación de Línea Germinal/genética , Síndrome de Hamartoma Múltiple/epidemiología , Síndrome de Hamartoma Múltiple/genética , Humanos , Masculino , Persona de Mediana Edad , Países Bajos/epidemiología , Síndrome de Peutz-Jeghers/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...