Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 58(11): 760-766, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-32994281

RESUMEN

BACKGROUND: Familial clustering of melanoma suggests a shared genetic predisposition among family members, but only 10%-40% of familial cases carry a pathogenic variant in a known high-risk melanoma susceptibility gene. We investigated whether a melanoma-specific Polygenic Risk Score (PRS) is associated with melanoma risk in patients with genetically unexplained familial melanoma. METHODS: Dutch familial melanoma cases (n=418) were genotyped for 46 SNPs previously identified as independently associated with melanoma risk. The 46-SNP PRS was calculated and standardised to 3423 healthy controls (sPRS) and the association between PRS and melanoma risk was modelled using logistic regression. Within the case series, possible differences were further explored by investigating the PRS in relation to (1) the number of primary melanomas in a patient and (2) the extent of familial clustering of melanoma. RESULTS: The PRS was significantly associated with melanoma risk, with a per-SD OR of 2.12 (95% CI 1.90 to 2.35, p<0.001), corresponding to a 5.70-fold increased risk (95% CI 3.93 to 8.28) when comparing the top 90th to the middle 40-60th PRS percentiles. The mean PRS was significantly higher in cases with multiple primary melanomas than in cases with a single melanoma (sPRS 1.17 vs 0.71, p=0.001). Conversely, cases from high-density melanoma families had a lower (but non-significant) mean PRS than cases from low-density families (sPRS 0.60 vs 0.94, p=0.204). CONCLUSION: Our work underlines the significance of a PRS in determining melanoma susceptibility and encourages further exploration of the diagnostic value of a PRS in genetically unexplained melanoma families.


Asunto(s)
Melanoma/genética , Polimorfismo de Nucleótido Simple , Neoplasias Cutáneas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Factor de Transcripción Asociado a Microftalmía/genética , Persona de Mediana Edad , Países Bajos , Receptor de Melanocortina Tipo 1/genética , Adulto Joven , Melanoma Cutáneo Maligno
2.
J Med Genet ; 57(3): 203-210, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31704778

RESUMEN

BACKGROUND: A proportion of patients diagnosed with cutaneous melanoma reports a positive family history. Inherited variants in CDKN2A and several other genes have been shown to predispose to melanoma; however, the genetic basis of familial melanoma remains unknown in most cases. The objective of this study was to provide insight into the genetic basis of familial melanoma. METHODS: In order to identify novel melanoma susceptibility genes, whole exome sequencing (WES) analysis was applied in a Dutch family with melanoma. The causality of a candidate variant was characterised by performing cosegregation analysis in five affected family members using patient-derived tissues and digital droplet PCR analysis to accurately quantify mutant allele frequency. Functional in-vitro studies were performed to assess the pathogenicity of the candidate variant. RESULTS: Application of WES identified a rare, nonsense variant in the NEK11 gene (c.1120C>T, p.Arg374Ter), cosegregating in all five affected members of a Dutch family. NEK11 (NIMA-related Kinase 11) is involved in the DNA damage response, enforcing the G2/M cell cycle checkpoint. In a melanoma from a variant carrier, somatic loss of the wildtype allele of this putative tumour suppressor gene was demonstrated. Functional analyses showed that the NEK11 p.Arg374Ter mutation results in strongly reduced expression of the truncated protein caused by proteasomal degradation. CONCLUSION: The NEK11 p.Arg374Ter variant identified in this family leads to loss-of-function through protein instability. Collectively, these findings support NEK11 as a melanoma susceptibility gene.


Asunto(s)
Predisposición Genética a la Enfermedad , Melanoma/genética , Quinasas Relacionadas con NIMA/genética , Neoplasias de la Úvea/genética , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Melanoma/fisiopatología , Persona de Mediana Edad , Mutación , Linaje , Penetrancia , Neoplasias de la Úvea/fisiopatología , Secuenciación del Exoma
3.
BMC Public Health ; 21(1): 692, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33888076

RESUMEN

BACKGROUND: Individuals from melanoma-prone families have similar or reduced sun-protective behaviors compared to the general population. Studies on trends in sun-related behaviors have been temporally and geographically limited. METHODS: Individuals from an international consortium of melanoma-prone families (GenoMEL) were retrospectively asked about sunscreen use, sun exposure (time spent outside), sunburns, and sunbed use at several timepoints over their lifetime. Generalized linear mixed models were used to examine the association between these outcomes and birth cohort defined by decade spans, after adjusting for covariates. RESULTS: A total of 2407 participants from 547 families across 17 centers were analyzed. Sunscreen use increased across subsequent birth cohorts, and although the likelihood of sunburns increased until the 1950s birth cohort, it decreased thereafter. Average sun exposure did not change across the birth cohorts, and the likelihood of sunbed use increased in more recent birth cohorts. We generally did not find any differences in sun-related behavior when comparing melanoma cases to non-cases. Melanoma cases had increased sunscreen use, decreased sun exposure, and decreased odds of sunburn and sunbed use after melanoma diagnosis compared to before diagnosis. CONCLUSIONS: Although sunscreen use has increased and the likelihood of sunburns has decreased in more recent birth cohorts, individuals in melanoma-prone families have not reduced their overall sun exposure and had an increased likelihood of sunbed use in more recent birth cohorts. These observations demonstrate partial improvements in melanoma prevention and suggest that additional intervention strategies may be needed to achieve optimal sun-protective behavior in melanoma-prone families.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Quemadura Solar , Humanos , Melanoma/epidemiología , Melanoma/prevención & control , Estudios Retrospectivos , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/prevención & control , Quemadura Solar/epidemiología , Quemadura Solar/prevención & control , Protectores Solares/uso terapéutico
4.
Genes Chromosomes Cancer ; 59(6): 366-374, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32017278

RESUMEN

Melanoma demonstrates altered patterns of DNA methylation that are associated with genetic instability and transcriptional repression of numerous genes. Active DNA demethylation is mediated by TET enzymes that catalyze conversion of 5-methylcytosine (mC) to 5-hydroxymethylcytosine (hmC). Loss of hmC occurs in melanoma and correlates with disease progression. Here we analyzed the genomic distribution of hmC along with mC in nevus and melanoma using oxidative bisulfite chemistry combined with high-density arrays. HmC was enriched relative to mC at enhancers, 5'UTR regions and CpG shores in nevus and melanoma samples, pointing to specific TET enzyme activity. The proportion of interrogated CpG sites with high hmC levels was lower in melanoma (0.54%) than in nevus (2.0%). Depletion of hmC in melanoma was evident across all chromosomes and intragenic regions, being more pronounced in metastatic than in non-metastatic tumors. The patterns of hmC distribution in melanoma samples differed significantly from those in nevus samples, exceeding differences in mC patterns. We identified specific CpG sites and regions with significantly lower hmC levels in melanoma than in nevus that might serve as diagnostic markers. Differentially hydroxymethylated regions localized to cancer-related genes, including the PTEN gene promoter, suggesting that deregulated DNA hydroxymethylation may contribute to melanoma pathogenesis.


Asunto(s)
5-Metilcitosina/análogos & derivados , Biomarcadores de Tumor/genética , Metilación de ADN , Melanoma/genética , Nevo/genética , Regiones no Traducidas 5' , 5-Metilcitosina/análisis , Adulto , Islas de CpG , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Nevo/patología
5.
Int J Cancer ; 144(10): 2453-2464, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30414346

RESUMEN

Germline mutations in the major melanoma susceptibility gene CDKN2A explain genetic predisposition in only 10-40% of melanoma-prone families. In our study we comprehensively characterized 488 melanoma cases from 451 non-CDKN2A/CDK4 families for mutations in 30 established and candidate melanoma susceptibility genes using a custom-designed targeted gene panel approach. We identified (likely) pathogenic variants in established melanoma susceptibility genes in 18 families (n = 3 BAP1, n = 15 MITF p.E318K; diagnostic yield 4.0%). Among the three identified BAP1-families, there were no reported diagnoses of uveal melanoma or malignant mesothelioma. We additionally identified two potentially deleterious missense variants in the telomere maintenance genes ACD and TERF2IP, but none in the POT1 gene. MC1R risk variants were strongly enriched in our familial melanoma cohort compared to healthy controls (R variants: OR 3.67, 95% CI 2.88-4.68, p <0.001). Several variants of interest were also identified in candidate melanoma susceptibility genes, in particular rare (pathogenic) variants in the albinism gene OCA2 were repeatedly found. We conclude that multigene panel testing for familial melanoma is appropriate considering the additional 4% diagnostic yield in non-CDKN2A/CDK4 families. Our study shows that BAP1 and MITF are important genes to be included in such a diagnostic test.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Predisposición Genética a la Enfermedad/genética , Mutación de Línea Germinal/genética , Melanoma/genética , Estudios de Cohortes , Femenino , Humanos , Neoplasias Pulmonares/genética , Masculino , Mesotelioma/genética , Mesotelioma Maligno , Factor de Transcripción Asociado a Microftalmía/genética , Ubiquitina Tiolesterasa/genética
6.
J Am Acad Dermatol ; 81(2): 386-394, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30731170

RESUMEN

BACKGROUND: Although rare in the general population, highly penetrant germline mutations in CDKN2A are responsible for 5%-40% of melanoma cases reported in melanoma-prone families. We sought to determine whether MELPREDICT was generalizable to a global series of families with melanoma and whether performance improvements can be achieved. METHODS: In total, 2116 familial melanoma cases were ascertained by the international GenoMEL Consortium. We recapitulated the MELPREDICT model within our data (GenoMELPREDICT) to assess performance improvements by adding phenotypic risk factors and history of pancreatic cancer. We report areas under the curve (AUC) with 95% confidence intervals (CIs) along with net reclassification indices (NRIs) as performance metrics. RESULTS: MELPREDICT performed well (AUC 0.752, 95% CI 0.730-0.775), and GenoMELPREDICT performance was similar (AUC 0.748, 95% CI 0.726-0.771). Adding a reported history of pancreatic cancer yielded discriminatory improvement (P < .0001) in GenoMELPREDICT (AUC 0.772, 95% CI 0.750-0.793, NRI 0.40). Including phenotypic risk factors did not improve performance. CONCLUSION: The MELPREDICT model functioned well in a global data set of familial melanoma cases. Adding pancreatic cancer history improved model prediction. GenoMELPREDICT is a simple tool for predicting CDKN2A mutational status among melanoma patients from melanoma-prone families and can aid in directing these patients to receive genetic testing or cancer risk counseling.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Predisposición Genética a la Enfermedad , Modelos Logísticos , Melanoma/genética , Neoplasias Pancreáticas , Neoplasias Cutáneas/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Niño , Pruebas Genéticas , Mutación de Línea Germinal , Heterocigoto , Humanos , Internacionalidad , Persona de Mediana Edad , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/genética , Fenotipo , Valor Predictivo de las Pruebas , Probabilidad , Curva ROC , Factores de Riesgo , Adulto Joven
7.
J Med Genet ; 55(10): 661-668, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29661971

RESUMEN

BACKGROUND: Several factors have been reported that influence the probability of a germline CDKN2A mutation in a melanoma family. Our goal was to create a scoring system to estimate this probability, based on a set of clinical features present in the patient and his or her family. METHODS: Five clinical features and their association with CDKN2A mutations were investigated in a training cohort of 1227 Dutch melanoma families (13.7% with CDKN2A mutation) using multivariate logistic regression. Predefined features included number of family members with melanoma and with multiple primary melanomas, median age at diagnosis and presence of pancreatic cancer or upper airway cancer in a family member. Based on these five features, a scoring system (CDKN2A Mutation(CM)-Score) was developed and subsequently validated in a combined Swedish and Dutch familial melanoma cohort (n=421 families; 9.0% with CDKN2A mutation). RESULTS: All five features were significantly associated (p<0.05) with a CDKN2A mutation. At a CM-Score of 16 out of 49 possible points, the threshold of 10% mutation probability is approximated (9.9%; 95% CI 9.8 to 10.1). This probability further increased to >90% for families with ≥36 points. A CM-Score under 16 points was associated with a low mutation probability (≤4%). CM-Score performed well in both the training cohort (area under the curve (AUC) 0.89; 95% CI 0.86 to 0.92) and the external validation cohort (AUC 0.94; 95% CI 0.90 to 0.98). CONCLUSION: We developed a practical scoring system to predict CDKN2A mutation status among melanoma-prone families. We suggest that CDKN2A analysis should be recommended to families with a CM-Score of ≥16 points.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Melanoma/genética , Neoplasias Pancreáticas/genética , Adulto , Estudios de Cohortes , Europa (Continente) , Femenino , Mutación de Línea Germinal , Humanos , Modelos Logísticos , Masculino , Melanoma/diagnóstico , Persona de Mediana Edad , Países Bajos , Neoplasias Pancreáticas/diagnóstico , Proyectos de Investigación , Suecia , Adulto Joven
8.
J Med Genet ; 54(9): 607-612, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28592523

RESUMEN

BACKGROUND: Sarcomas are rare mesenchymal malignancies whose pathogenesis is poorly understood; both environmental and genetic risk factors could contribute to their aetiology. METHODS AND RESULTS: We performed whole-exome sequencing (WES) in a familial aggregation of three individuals affected with soft-tissue sarcoma (STS) without TP53 mutation (Li-Fraumeni-like, LFL) and found a shared pathogenic mutation in CDKN2A tumour suppressor gene. We searched for individuals with sarcoma among 474 melanoma-prone families with a CDKN2A-/+ genotype and for CDKN2A mutations in 190 TP53-negative LFL families where the index case was a sarcoma. Including the initial family, eight independent sarcoma cases carried a germline mutation in the CDKN2A/p16INK4A gene. In five out of seven formalin-fixed paraffin-embedded sarcomas, heterozygosity was lost at germline CDKN2A mutations sites demonstrating complete loss of function. As sarcomas are rare in CDKN2A/p16INK4A carriers, we searched in constitutional WES of nine carriers for potential modifying rare variants and identified three in platelet-derived growth factor receptor (PDGFRA) gene. Molecular modelling showed that two never-described variants could impact the PDGFRA extracellular domain structure. CONCLUSION: Germline mutations in CDKN2A/P16INK4A, a gene known to predispose to hereditary melanoma, pancreatic cancer and tobacco-related cancers, account also for a subset of hereditary sarcoma. In addition, we identified PDGFRA as a candidate modifier gene.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Genes p16 , Mutación de Línea Germinal , Sarcoma/genética , Neoplasias de los Tejidos Blandos/genética , Femenino , Determinismo Genético , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Masculino , Linaje , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Secuenciación del Exoma
9.
Hum Genet ; 135(11): 1241-1249, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27449771

RESUMEN

The risk of pancreatic cancer (PC) is increased in melanoma-prone families but the causal relationship between germline CDKN2A mutations and PC risk is uncertain, suggesting the existence of non-CDKN2A factors. One genetic possibility involves patients having mutations in multiple high-risk PC-related genes; however, no systematic examination has yet been conducted. We used next-generation sequencing data to examine 24 putative PC-related genes in 43 PC patients with and 23 PC patients without germline CDKN2A mutations and 1001 controls. For each gene and the four pathways in which they occurred, we tested whether PC patients (overall or CDKN2A+ and CDKN2A- cases separately) had an increased number of rare nonsynonymous variants. Overall, we identified 35 missense variants in PC patients, 14 in CDKN2A+ and 21 in CDKN2A- PC cases. We found nominally significant associations for mismatch repair genes (MLH1, MSH2, MSH6, PMS2) in all PC patients and for ATM, CPA1, and PMS2 in CDKN2A- PC patients. Further, nine CDKN2A+ and four CDKN2A- PC patients had rare potentially deleterious variants in multiple PC-related genes. Loss-of-function variants were only observed in CDKN2A- PC patients, with ATM having the most pathogenic variants. Also, ATM variants (n = 5) were only observed in CDKN2A- PC patients with a family history that included digestive system tumors. Our results suggest that a subset of PC patients may have increased risk because of germline mutations in multiple PC-related genes.


Asunto(s)
Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Melanoma/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal/genética , Humanos , Masculino , Melanoma/patología , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/patología , Linaje , Factores de Riesgo , Transducción de Señal/genética , Neoplasias Cutáneas , Melanoma Cutáneo Maligno
10.
Int J Cancer ; 136(3): 618-31, 2015 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24917043

RESUMEN

The MC1R gene is a key regulator of skin pigmentation. We aimed to evaluate the association between MC1R variants and the risk of sporadic cutaneous melanoma (CM) within the M-SKIP project, an international pooled-analysis on MC1R, skin cancer and phenotypic characteristics. Data included 5,160 cases and 12,119 controls from 17 studies. We calculated a summary odds ratio (SOR) for the association of each of the nine most studied MC1R variants and of variants combined with CM by using random-effects models. Stratified analysis by phenotypic characteristics were also performed. Melanoma risk increased with presence of any of the main MC1R variants: the SOR for each variant ranged from 1.47 (95%CI: 1.17-1.84) for V60L to 2.74 (1.53-4.89) for D84E. Carriers of any MC1R variant had a 66% higher risk of developing melanoma compared with wild-type subjects (SOR; 95%CI: 1.66; 1.41-1.96) and the risk attributable to MC1R variants was 28%. When taking into account phenotypic characteristics, we found that MC1R-associated melanoma risk increased only for darker-pigmented Caucasians: SOR (95%CI) was 3.14 (2.06-4.80) for subjects with no freckles, no red hair and skin Type III/IV. Our study documents the important role of all the main MC1R variants in sporadic CM and suggests that they have a direct effect on melanoma risk, independently on the phenotypic characteristics of carriers. This is of particular importance for assessing preventive strategies, which may be directed to darker-pigmented Caucasians with MC1R variants as well as to lightly pigmented, fair-skinned subjects.


Asunto(s)
Predisposición Genética a la Enfermedad , Melanoma/genética , Receptor de Melanocortina Tipo 1/genética , Neoplasias Cutáneas/genética , Humanos , Melanoma/etiología , Persona de Mediana Edad , Fenotipo , Riesgo , Neoplasias Cutáneas/etiología , Pigmentación de la Piel , Población Blanca
11.
Int J Cancer ; 136(6): 1351-60, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25077817

RESUMEN

At least 17 genomic regions are established as harboring melanoma susceptibility variants, in most instances with genome-wide levels of significance and replication in independent samples. Based on genome-wide single nucleotide polymorphism (SNP) data augmented by imputation to the 1,000 Genomes reference panel, we have fine mapped these regions in over 5,000 individuals with melanoma (mainly from the GenoMEL consortium) and over 7,000 ethnically matched controls. A penalized regression approach was used to discover those SNP markers that most parsimoniously explain the observed association in each genomic region. For the majority of the regions, the signal is best explained by a single SNP, which sometimes, as in the tyrosinase region, is a known functional variant. However in five regions the explanation is more complex. At the CDKN2A locus, for example, there is strong evidence that not only multiple SNPs but also multiple genes are involved. Our results illustrate the variability in the biology underlying genome-wide susceptibility loci and make steps toward accounting for some of the "missing heritability."


Asunto(s)
Predisposición Genética a la Enfermedad , Melanoma/genética , Polimorfismo de Nucleótido Simple , Mapeo Cromosómico , Ciclina D1/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Sitios Genéticos , Humanos , Telomerasa/genética
12.
J Am Acad Dermatol ; 72(3): 496-507.e7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25592620

RESUMEN

BACKGROUND: Inherited susceptibility genes have been associated with histopathologic characteristics of tumors. OBJECTIVE: We sought to identify associations between histology of melanomas and CDKN2A genotype. METHODS: This was a case-control study design comparing 28 histopathologic tumor features among individuals with sporadic melanomas (N = 81) and cases from melanoma families with (N = 123) and without (N = 120) CDKN2A germline mutations. RESULTS: Compared with CDKN2A(-) cases, mutation carriers tended to have histologic features of superficial spreading melanoma subtype including higher pigmentation (Ptrend = .02) and increased pagetoid scatter (Ptrend = .07) after adjusting for age at diagnosis, sex, and American Joint Committee on Cancer thickness category. Similar associations were observed when comparing mutation carriers with a combined group of CDKN2A(-) (wild type) and sporadic melanomas. The presence of spindle cell morphology in the vertical growth phase was also an important predictor of genotype. Of the 15 cases with this phenotype, none were observed to harbor a CDKN2A mutation. LIMITATIONS: Our study examined rare mutations and may have been underpowered to detect small, but biologically significant associations between histology and genotype. CONCLUSION: Familial melanomas with CDKN2A mutations preferentially express a histologic phenotype of dense pigmentation, high pagetoid scatter, and a non-spindle cell morphology in the vertical growth phase.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Melanoma/genética , Melanoma/patología , Mutación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Adulto , Estudios de Casos y Controles , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad
13.
Nat Genet ; 38(1): 3-5, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16468121

RESUMEN

Networks of investigators have begun sharing best practices, tools and methods for analysis of associations between genetic variation and common diseases. A Network of Investigator Networks has been set up to drive the process, sponsored by the Human Genome Epidemiology Network. A workshop is planned to develop consensus guidelines for reporting results of genetic association studies. Published literature databases will be integrated, and unpublished data, including 'negative' studies, will be captured by online journals and through investigator networks. Systematic reviews will be expanded to include more meta-analyses of individual-level data and prospective meta-analyses. Field synopses will offer regularly updated overviews.


Asunto(s)
Métodos Epidemiológicos , Genoma Humano , Bases de Datos Factuales , Predisposición Genética a la Enfermedad , Proyecto Genoma Humano , Humanos , MEDLINE , Proyectos de Investigación
14.
Differentiation ; 85(4-5): 173-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23817083

RESUMEN

It is generally thought that class III ß-tubulin expression is limited to cells of the neural lineage and is therefore often used to identify neurons amongst other cell types, both in vivo and in vitro. Melanocytes are derived from the neural crest and share both morphological features and functional characteristics with peripheral neurons. Here, we show that these similarities extend to class III ß-tubulin (TUBB3) expression, and that human melanocytes express this protein both in vivo and in vitro. In addition, we studied the expression of class III ß-tubulin in two murine melanogenic cell lines and show that expression of this protein starts as melanoblasts mature into melanocytes. Melanin bleaching experiments revealed close proximity between melanin and TUBB3 proteins. In vitro stimulation of primary human melanocytes by α-MSH indicated separate regulatory mechanisms for melanogenesis and to TUBB3 expression. Together, these observations imply that human melanocytes express TUBB3 and that this protein should be recognized as a wider marker for multiple neural crest-derived cells.


Asunto(s)
Linaje de la Célula , Melanocitos/metabolismo , Tubulina (Proteína)/metabolismo , Anciano , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Melaninas/metabolismo , Melanocitos/citología , Ratones , Cresta Neural/citología , Neuronas/metabolismo , Factores de Transcripción/metabolismo , alfa-MSH/metabolismo
15.
Gastroenterology ; 140(3): 850-6, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21129377

RESUMEN

BACKGROUND & AIMS: Surveillance of high-risk groups for pancreatic cancer might increase early detection and treatment outcomes. Individuals with germline mutations in p16-Leiden have a lifetime risk of 15% to 20% of developing pancreatic cancer. We assessed the feasibility of detecting pancreatic cancer at an early stage and investigated the outcomes of patients with neoplastic lesions. METHODS: Individuals with germline mutations in p16-Leiden (N = 79; 31 male; mean age, 56 years; range, 39-72 years) were offered annual surveillance by magnetic resonance imaging (MRI) and magnetic resonance cholangiopancreatography (MRCP). Those found to have neoplastic lesions were offered options for surgery or intensive follow-up. Individuals found to have possible neoplastic lesions were examined again by MRI/MRCP within 2 to 4 months. RESULTS: After a median follow-up period of 4 years (range, 0-10 years), pancreatic cancer was diagnosed in 7 patients (9%). The mean age at diagnosis was 59 years (range, 49-72 years). Three of the tumors were present at the first examination, and 4 were detected after a negative result in the initial examination. All 7 patients had a resectable lesion; 5 underwent surgery, 3 had an R0 resection, and 2 had lymph node metastases. Possible precursor lesions (ie, duct ectasias, based on MRCP) were found in 9 individuals (11%). CONCLUSIONS: MRI/MRCP detects small, solid pancreatic tumors and small duct ectasias. Although surveillance increases the rate of resectability, carriers of a p16-Leiden mutation develop aggressive tumors.


Asunto(s)
Adenocarcinoma/diagnóstico , Pancreatocolangiografía por Resonancia Magnética , Genes p16 , Mutación de Línea Germinal , Imagen por Resonancia Magnética , Tamizaje Masivo/métodos , Páncreas/patología , Neoplasias Pancreáticas/diagnóstico , Lesiones Precancerosas/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma/cirugía , Adulto , Anciano , Detección Precoz del Cáncer , Estudios de Factibilidad , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Países Bajos , Páncreas/cirugía , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/cirugía , Fenotipo , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Lesiones Precancerosas/cirugía , Valor Predictivo de las Pruebas , Pronóstico , Sistema de Registros , Factores de Tiempo
16.
BMC Med Res Methodol ; 12: 116, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22862891

RESUMEN

BACKGROUND: For complex diseases like cancer, pooled-analysis of individual data represents a powerful tool to investigate the joint contribution of genetic, phenotypic and environmental factors to the development of a disease. Pooled-analysis of epidemiological studies has many advantages over meta-analysis, and preliminary results may be obtained faster and with lower costs than with prospective consortia. DESIGN AND METHODS: Based on our experience with the study design of the Melanocortin-1 receptor (MC1R) gene, SKin cancer and Phenotypic characteristics (M-SKIP) project, we describe the most important steps in planning and conducting a pooled-analysis of genetic epidemiological studies. We then present the statistical analysis plan that we are going to apply, giving particular attention to methods of analysis recently proposed to account for between-study heterogeneity and to explore the joint contribution of genetic, phenotypic and environmental factors in the development of a disease. Within the M-SKIP project, data on 10,959 skin cancer cases and 14,785 controls from 31 international investigators were checked for quality and recoded for standardization. We first proposed to fit the aggregated data with random-effects logistic regression models. However, for the M-SKIP project, a two-stage analysis will be preferred to overcome the problem regarding the availability of different study covariates. The joint contribution of MC1R variants and phenotypic characteristics to skin cancer development will be studied via logic regression modeling. DISCUSSION: Methodological guidelines to correctly design and conduct pooled-analyses are needed to facilitate application of such methods, thus providing a better summary of the actual findings on specific fields.


Asunto(s)
Diseño de Investigaciones Epidemiológicas , Predisposición Genética a la Enfermedad , Receptor de Melanocortina Tipo 1 , Neoplasias Cutáneas/genética , Luz Solar/efectos adversos , Adulto , Estudios de Casos y Controles , Recolección de Datos/normas , Interpretación Estadística de Datos , Femenino , Hospitalización , Humanos , Modelos Logísticos , Masculino , Metaanálisis como Asunto , Fenotipo , Neoplasias Cutáneas/fisiopatología , Neoplasias Cutáneas/secundario , Fumar
17.
Acta Derm Venereol ; 92(6): 587-92, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22965007

RESUMEN

Individuals with two red hair colour (RHC)-MC1R genetic variants have light skin and blond/reddish hair and, in comparison with those without such alleles, are at an increased risk of developing melanoma. Our study investigated the association of RHC variants and the Total Dermo-scopy Score (TDS), and the items that make up the TDS, in those with atypical naevi and melanomas from high risk melanoma patients. Eight hundred and seventy-six atypical naevi and 21 melanomas were scored according to the TDS system and MC1R polymorphisms were determined. Analyses revealed that several TDS items including pigment network, dark-brown colour and streaks were more frequently observed in atypical naevi from individuals without RHC variants, while structureless areas were more often observed in individuals with two RHC variants. Finally, no significant difference in TDS was detected in atypical naevi from individuals with two RHC variants compared to those without RHC. Clinicians should be aware of a different dermoscopic naevus pheno-type in patients with light blond or RHC MC1R variants.


Asunto(s)
Dermoscopía , Melanoma/genética , Nevo Pigmentado/genética , Polimorfismo Genético , Receptor de Melanocortina Tipo 1/genética , Neoplasias Cutáneas/genética , Piel/patología , Distribución de Chi-Cuadrado , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Predisposición Genética a la Enfermedad , Color del Cabello/genética , Humanos , Modelos Lineales , Modelos Logísticos , Melanoma/patología , Mutación , Nevo Pigmentado/patología , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Neoplasias Cutáneas/patología , Pigmentación de la Piel/genética
18.
J Mol Diagn ; 24(1): 88-100, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34775028

RESUMEN

An accurate T-cell quantification is prognostically and therapeutically relevant in various malignancies. We previously developed a digital PCR-based approach offering a precise T-cell enumeration in small amounts of DNA. However, it may be challenging to apply this method in malignant specimens, as genetic instability can disturb the underlying mathematical model. For example, approximately 24% of the tumors from The Cancer Genome Atlas pan-cancer data set carried a copy number alteration affecting the TRB gene T-cell marker, which would cause an underestimation or overestimation of the T-cell fraction. In this study, we introduce a multiplex digital PCR experimental setup to quantify T cells in copy number unstable DNA samples. By implementing a so-called regional corrector, genetic alterations involving the T-cell marker locus can be recognized and corrected for. This novel setup is evaluated mathematically in silico and validated in vitro by measuring T-cell presence in various samples with a known T-cell fraction. The utility of the approach is further demonstrated in copy number altered cutaneous melanomas. Our novel multiplex setup provides a simple, but accurate, DNA-based T-cell quantification in both copy number stable and unstable specimens. This approach has potential clinical and diagnostic applications, as it does not depend on availability of T-cell epitopes, has low requirements for sample quantity and quality, and can be performed in a relatively easy experiment.


Asunto(s)
Variaciones en el Número de Copia de ADN , Linfocitos T , ADN/genética , Variaciones en el Número de Copia de ADN/genética , Humanos , Reacción en Cadena de la Polimerasa Multiplex/métodos
19.
JCO Precis Oncol ; 6: e2200145, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36409970

RESUMEN

PURPOSE: Pancreatic ductal adenocarcinoma (PDAC) is a component of familial melanoma due to germline pathogenic variants (GPVs) in CDKN2A. However, it is unclear what role this gene or other genes play in its etiology. MATERIALS AND METHODS: We analyzed 189 cancer predisposition genes using parametric rare-variant association (RVA) tests and nonparametric permutation tests to identify gene-level associations in PDAC for patients with (CDKN2A+) and without (CDKN2A-) GPV. Exome sequencing was performed on 84 patients with PDAC, 47 CDKN2A+ and 37 CDKN2A-. After variant filtering, various RVA tests and permutation tests were run separately by CDKN2A status. Genes with the strongest nominal associations were evaluated in patients with PDAC from The Cancer Genome Atlas and the UK Biobank (UKB). A secondary analysis including only GPV from UKB was also performed. RESULTS: In RVA tests, ERCC4 and RET showed the most compelling evidence as plausible PDAC candidate genes for CDKN2A+ patients. In contrast, the findings in CDKN2A- patients provided evidence for HMBS, EPCAM, and MRE11 as potential new candidate genes and confirmed ATM, BRCA2, and PALB2 as PDAC genes, consistent with findings in The Cancer Genome Atlas and the UKB. As expected, CDKN2A- patients were more likely to harbor GPVs from the 189 genes investigated. When including only GPVs from UKB, significant associations with PDAC were seen for ATM, BRCA2, and CDKN2A. CONCLUSION: These results suggest that variants in other genes likely play a role in PDAC in all patients and that PDAC in CDKN2A+ patients has a distinct etiology from PDAC in CDKN2A- patients.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Predisposición Genética a la Enfermedad/genética , Neoplasias Pancreáticas/genética , Carcinoma Ductal Pancreático/genética , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Células Germinativas/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA