Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
JCO Precis Oncol ; 5: 1677-1686, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34994652

RESUMEN

PURPOSE: Multigene panel testing (MGPT) identifies TP53 pathogenic or likely pathogenic (P/LP) variants in patients with diverse phenotypes, of which only one is classic Li-Fraumeni syndrome. Low variant allelic fraction (VAF) in TP53 found on germline testing may suggest aberrant clonal expansion or constitutional mosaicism. We evaluated TP53-positive probands seen in a cancer genetics program to determine germline versus somatic status. METHODS: We reviewed TP53-positive probands from 2012 to 2019 identified by MGPT on blood or saliva (N = 84). Available VAFs were collected. Probands with a familial variant, who met Li-Fraumeni syndrome testing criteria or who carried a founder variant, were considered germline. For those with uncertain germline status, TP53 variants were further examined using ancillary data of family members and somatic tissue. RESULTS: Of the 84 probands, 54.7% had germline variants with 33.3% meeting criteria for germline status and 21.4% confirmed through ancillary testing. Aberrant clonal expansion comprised 13.1% with clonal hematopoiesis of indeterminate potential and 2.4% with a hematologic malignancy. Constitutional mosaicism was confirmed in 8.3% probands. Definitive status could not be determined in 3.6% despite ancillary assessment, and 17.9% did not have ancillary testing. CONCLUSION: A TP53 P/LP variant found on peripheral blood or saliva MGPT does not always originate in the germline. In a clinical cancer genetics cohort, approximately half of the patients had TP53 P/LP germline variants; these patients plus those with constitutional mosaicism require intensified surveillance. A framework of multiple strategies enables discernment of germline from constitutional mosaic and acquired variants, which is essential for appropriate management.


Asunto(s)
Síndrome de Li-Fraumeni/sangre , Síndrome de Li-Fraumeni/genética , Saliva/química , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Pruebas Genéticas , Variación Genética , Células Germinativas , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Proteína p53 Supresora de Tumor/sangre , Adulto Joven
4.
J Med Genet ; 55(3): 173-180, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29070607

RESUMEN

BACKGROUND: Development of tumours such as adrenocortical carcinomas (ACC), choroid plexus tumours (CPT) or female breast cancers before age 31 or multiple primary cancers belonging to the Li-Fraumeni (LFS) spectrum is, independently of the familial history, highly suggestive of a germline TP53 mutation. The aim of this study was to determine the contribution of de novo and mosaic mutations to LFS. METHODS AND RESULTS: Among 328 unrelated patients harbouring a germline TP53 mutation identified by Sanger sequencing and/or QMPSF, we could show that the mutations had occurred de novo in 40 cases, without detectable parental age effect. Sanger sequencing revealed two mosaic mutations in a child with ACC and in an unaffected father of a child with medulloblastoma. Re-analysis of blood DNA by next-generation sequencing, performed at a depth above 500X, from 108 patients suggestive of LFS without detectable TP53 mutations, allowed us to identify 6 additional cases of mosaic TP53 mutations, in 2/49 children with ACC, 2/21 children with CPT, in 1/31 women with breast cancer before age 31 and in a patient who developed an osteosarcoma at age 12, a breast carcinoma and a breast sarcoma at age 35. CONCLUSIONS: This study performed on a large series of TP53 mutation carriers allows estimating the contribution to LFS of de novo mutations to at least 14% (48/336) and suggests that approximately one-fifth of these de novo mutations occur during embryonic development. Considering the medical impact of TP53 mutation identification, medical laboratories in charge of TP53 testing should ensure the detection of mosaic mutations.


Asunto(s)
Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Síndrome de Li-Fraumeni/genética , Proteína p53 Supresora de Tumor/genética , Carcinoma Corticosuprarrenal/sangre , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/patología , Adulto , Neoplasias de la Mama/sangre , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Niño , Neoplasias del Plexo Coroideo/sangre , Neoplasias del Plexo Coroideo/genética , Neoplasias del Plexo Coroideo/patología , Femenino , Mutación de Línea Germinal/genética , Humanos , Síndrome de Li-Fraumeni/sangre , Síndrome de Li-Fraumeni/patología , Masculino , Persona de Mediana Edad , Mosaicismo , Proteína p53 Supresora de Tumor/sangre , Adulto Joven
5.
Hum Mol Genet ; 26(14): 2591-2602, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28369373

RESUMEN

Li-Fraumeni Syndrome (LFS) results from heterozygous germline mutations of TP53, encoding a key transcriptional factor activated in response to DNA damage. We have recently shown, from a large LFS series, that dominant-negative missense mutations are the most clinically severe and, thanks to a new p53 functional assay in lymphocytes, that they alter the p53 transcriptional response to DNA damage more drastically than null mutations. In this study, we first confirmed this observation by performing the p53 functional assay in lymphocytes from 56 TP53 mutation carriers harbouring 35 distinct alterations. Then, to compare the impact of the different types of germline TP53 mutations on DNA binding, we performed chromatin immunoprecipitation-sequencing (ChIP-Seq) in lymphocytes exposed to doxorubicin. ChIP-Seq performed in wild-type TP53 control lymphocytes accurately mapped 1287 p53-binding sites. New p53-binding sites were validated using a functional assay in yeast. ChIP-Seq analysis of LFS lymphocytes carrying TP53 null mutations (p.P152Rfs*18 or complete deletion) or the low penetrant 'Brazilian' p.R337H mutation revealed a moderate decrease of p53-binding sites (949, 580 and 620, respectively) and of ChIP-Seq peak depths. In contrast, analysis of LFS lymphocytes with TP53 dominant-negative missense mutations p.R273H or p.R248W revealed only 310 and 143 p53-binding sites, respectively, and the depths of the corresponding peaks were drastically reduced. Altogether, our results show that TP53 mutation carriers exhibit a constitutive defect of the transcriptional response to DNA damage and that the clinical severity of TP53 dominant-negative missense mutations is explained by a massive and global alteration of p53 DNA binding.


Asunto(s)
ADN/metabolismo , Mutación de Línea Germinal , Síndrome de Li-Fraumeni/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Niño , Preescolar , Inmunoprecipitación de Cromatina , ADN/sangre , Daño del ADN , Femenino , Genes p53 , Predisposición Genética a la Enfermedad , Humanos , Lactante , Síndrome de Li-Fraumeni/sangre , Linfocitos/fisiología , Masculino , Persona de Mediana Edad , Transcripción Genética
6.
Mutat Res Genet Toxicol Environ Mutagen ; 789-790: 28-35, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26232255

RESUMEN

The p53 tumor suppressor protein has emerged as a universal sensor of genotoxic stress that regulates the transcription of numerous genes required for appropriate cellular response to DNA damage. Therefore, transcriptional induction of p53 target genes can be considered as a global and early indicator of genotoxic stress. By performing expression microarrays and RNA-Seq analysis on wild-type and mutant TP53 human lymphocytes respectively derived from controls and Li-Fraumeni patients and exposed to different classes of genotoxic agents, we first determined a common p53-dependent transcriptional signature of DNA damage. We then derived a simple and fast assay based on the exposure of wild-type TP53 lymphocytes to physical or chemical agents and on the quantitative measurement of selected p53 target gene transcriptional induction. The specificity of the p53 genotoxicity assay can easily be demonstrated by performing the same experiment in control lymphocytes with heterozygous TP53 mutations, which compromise responses to DNA damage. This assay allowed us to show that most of the drugs commonly used in cancer treatment, except the microtubule poisons, are highly genotoxic. The p53 genotoxicity assay should facilitate the measurement of the genotoxic effects of chemical and physical agents and the identification of drugs that are not genotoxic and do not expose patients to the risk of secondary malignancies, especially those with a constitutional defect in response to DNA damage, such as patients with Li-Fraumeni syndrome.


Asunto(s)
Linfocitos/metabolismo , Pruebas de Mutagenicidad/métodos , Transcriptoma/genética , Proteína p53 Supresora de Tumor/genética , Antineoplásicos/farmacología , Células Cultivadas , Cisplatino/farmacología , Daño del ADN , Doxorrubicina/farmacología , Fluorouracilo/farmacología , Humanos , Síndrome de Li-Fraumeni/sangre , Síndrome de Li-Fraumeni/genética , Linfocitos/efectos de los fármacos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reproducibilidad de los Resultados , Activación Transcripcional/efectos de los fármacos , Transcriptoma/efectos de los fármacos
7.
Cell Cycle ; 10(7): 1152-61, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21389785

RESUMEN

Ionizing radiation induces DNA Double-Strand Breaks (DSBs) which activate the ATM/CHEK2/p53 pathway leading to cell cycle arrest and apoptosis through transcription of genes including CDKN1A (p21) and BBC3 (PUMA). This pathway prevents genomic instability and tumorigenesis as demonstrated in heritable syndromes [e.g. Ataxia Telangiectasia (AT); Li-Fraumeni syndrome (LFS)]. Here, a simple assay based on gene expression in peripheral blood to measure accurately ATM/CHEK2/p53 pathway activity is described. The expression of p21, Puma and Sesn2 was determined in blood from mice with different gene copy numbers of Atm, Trp53 (p53), Chek2 or Arf and in human blood and mitogen stimulated T-lymphocyte (MSTL) cultures from AT, AT carriers, LFS patients, and controls, both before and after ex vivo ionizing irradiation. Mouse Atm/Chek2/p53 activity was highly dependent on the copy number of each gene except Arf. In human MSTL, an AT case, AT carriers and LFS patients showed responses distinct from healthy donors. The relationship between gene copy number and transcriptional induction upon radiation was linear for p21 and Puma and correlated well with cancer incidence in p53 variant mice. This reliable blood test provides an assay to determine ATM/CHEK2/p53 pathway activity and demonstrates the feasibility of assessing the activity of this essential cancer protection pathway in simple assays. These findings may have implications for the individualized prediction of cancer susceptibility.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Susceptibilidad a Enfermedades/metabolismo , Perfilación de la Expresión Génica/métodos , Neoplasias/diagnóstico , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/sangre , Ataxia Telangiectasia/sangre , Ataxia Telangiectasia/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada , Quinasa de Punto de Control 2 , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/sangre , Susceptibilidad a Enfermedades/sangre , Dosificación de Gen/genética , Regulación de la Expresión Génica/fisiología , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Síndrome de Li-Fraumeni/sangre , Síndrome de Li-Fraumeni/metabolismo , Ratones , Proteínas Nucleares/sangre , Proteínas Proto-Oncogénicas/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Linfocitos T/metabolismo
8.
Cancer ; 110(3): 694-702, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17567834

RESUMEN

BACKGROUND: A decrease in the age at cancer onset and increase in cancer incidence in successive generations in Li-Fraumeni syndrome (LFS) families with germline TP53 mutations have been previously described. In the current study a possible relation was analyzed between telomere length and cancer onset in TP53 mutation carriers. METHODS: Telomere length was measured using real-time quantitative polymerase chain reaction (PCR) in 20 carriers of germline TP53 mutations and in 83 unrelated healthy individuals. According to the age at blood sampling, patients and controls were divided into 2 age groups, children and adults. Telomere length was correlated to TP53 mutation status and telomere shortening in patients to the age at cancer onset. A t-test and linear regression were used to analyze the data. RESULTS: Compared with healthy controls, telomere length was significantly shorter both in the child (P = .001) and adult (P = .034) germline T53 mutation carriers. Although a statistically significant correlation between telomere shortening and the age at cancer onset was not observed, there was a trend of shorter telomeres in mutation carriers affected in childhood compared with those affected later in life. Neither cancer therapy nor sex differences were likely to affect the results. CONCLUSIONS: The findings suggest a possible link between the carriership of a germline TP53 mutation, telomere length, predisposition to early-onset cancer, and anticipation in LFS.


Asunto(s)
Mutación de Línea Germinal/genética , Síndrome de Li-Fraumeni/genética , Neoplasias/genética , Telómero/fisiología , Proteína p53 Supresora de Tumor/genética , Adulto , Edad de Inicio , Niño , Femenino , Tamización de Portadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Síndrome de Li-Fraumeni/sangre , Masculino , Neoplasias/sangre , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Cancer Res ; 67(4): 1415-8, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308077

RESUMEN

Li-Fraumeni syndrome (LFS) is a cancer predisposition syndrome frequently associated with germ line TP53 mutations. Unpredictable and disparate age of cancer onset is a major challenge in the management of LFS. Genetic modifiers, including the MDM2-SNP309 polymorphism, and genetic anticipation have been suggested as plausible explanations for young age of tumor onset, but the molecular mechanisms for these observations are unknown. We speculated that telomere attrition will increase genomic instability and cause earlier tumor onset in successive generations. We analyzed mean telomere length and MDM2-SNP309 polymorphism status in individuals from multiple LFS families and controls. A total of 45 peripheral blood lymphocyte samples were analyzed from 9 LFS families and 15 controls. High rate of MDM2-SNP309 was found in TP53 carriers (P = 0.0003). In children, telomere length was shorter in carriers affected with cancer than in nonaffected carriers and wild-type controls (P < 0.0001). The same pattern was seen in adults (P = 0.002). Within each family, telomere length was shorter in children with cancer than in their nonaffected siblings and their noncarrier parents. Telomere attrition between children and adults was faster in carriers than in controls. Our results support the role of MDM2-SNP309 as a genetic modifier in LFS. The novel finding of accelerated telomere attrition in LFS suggests that telomere length could explain earlier age of onset in successive generations of the same family with identical TP53/MDM2-SNP309 genotypes. Furthermore, telomere shortening could predict genetic anticipation observed in LFS and may serve as the first rational biological marker for clinical monitoring of these patients.


Asunto(s)
Síndrome de Li-Fraumeni/genética , Neoplasias/genética , Telómero/fisiología , Adolescente , Adulto , Factores de Edad , Niño , Preescolar , Femenino , Genes p53 , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Lactante , Síndrome de Li-Fraumeni/sangre , Linfocitos/ultraestructura , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Linaje , Polimorfismo de Nucleótido Simple
10.
Cancer Res ; 57(10): 1895-902, 1997 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9157982

RESUMEN

Lymphoblastoid cell lines (LCLs) with heterozygous p53 mutations at residues 286A, 133R, 282W, 132E, and 213ter were established from five independent Li-Fraumeni syndrome families. When cell cycle regulation in response to gamma-irradiation was studied, these LCLs showed an abnormal G1 checkpoint associated with defective inhibition of cyclin E/cyclin-dependent kinase 2 activity in all cases except for 282W LCL, which showed a normal G1 checkpoint. On the other hand, the control of S-phase-G2 as determined by cyclin A/cyclin-dependent kinase 2 activity was defective in all these LCLs. The mitotic checkpoint was also defective in the two LCLs analyzed as either competent or incompetent for G1 arrest. When radiation-induced apoptosis, which requires wild-type p53 function under optimal conditions, was studied, all of these LCLs showed significant failure compared to normal LCLs. These findings indicate that although p53-dependent transactivation and G1-S-phase cell cycle control are variably dysregulated, the induction of apoptosis and control of the cell cycle at S-phase-G2 and the mitotic checkpoint in response to DNA-damaging agents are consistently dysregulated in heterozygous mutant LCLs. This suggests that these dysfunctions underlie, at least in part, the susceptibility of Li-Fraumeni syndrome families to cancer. Furthermore, the approach presented is a potentially useful method for studying individual carriers of different germ-line p53 mutations and different biological features.


Asunto(s)
Apoptosis/fisiología , Quinasas CDC2-CDC28 , Daño del ADN , Genes p53 , Mutación de Línea Germinal , Síndrome de Li-Fraumeni/genética , Síndrome de Li-Fraumeni/patología , Adolescente , Adulto , Alelos , Apoptosis/efectos de la radiación , Ciclo Celular/fisiología , Muerte Celular/efectos de la radiación , Transformación Celular Viral , Preescolar , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/biosíntesis , Ciclinas/metabolismo , Susceptibilidad a Enfermedades , Femenino , Expresión Génica , Herpesvirus Humano 4 , Humanos , Síndrome de Li-Fraumeni/sangre , Linfocitos/citología , Linfocitos/fisiología , Linfocitos/efectos de la radiación , Masculino , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Activación Transcripcional , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/fisiología
11.
Oncogene ; 14(15): 1869-74, 1997 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-9150393

RESUMEN

Previous investigations of a Li - Fraumeni like family (Barnes et al., 1992) demonstrated that both the proband and her mother had elevated p53 protein levels in both tumour tissue and normal tissue at sites distant from the tumour, although no mutation was found in the p53 gene. In the present study two recently described functional assays for p53, an apoptotic assay and the functional assay for the separation of alleles in yeast (FASAY), have been employed to study the functional activity of p53 from this patient. The results of the apoptotic assay demonstrated that this patient had a p53 functional defect and the FASAY result suggested that this defect was in fact a germline mutation of the p53 gene. A point mutation of codon 337, which results in an amino acid substitution of a cysteine for an arginine, was demonstrated initially in cDNA and was confirmed by sequencing of genomic DNA. This is an unusual mutation as it is in the oligomerisation domain of p53, in contrast to the majority of p53 mutations which are in the core DNA binding domain. This mutation results in a protein which still retains partial transactivational activity in the FASAY. The mutation of codon 337 is only the second reported case of a germline missense mutation occurring in the oligomerisation domain of p53.


Asunto(s)
Genes p53 , Mutación de Línea Germinal , Síndrome de Li-Fraumeni/genética , Mutación Puntual , Proteína p53 Supresora de Tumor/genética , Alelos , Apoptosis/fisiología , Codón , ADN Complementario/sangre , ADN Complementario/genética , Humanos , Síndrome de Li-Fraumeni/sangre , Síndrome de Li-Fraumeni/patología , Linfocitos/fisiología , Estructura Terciaria de Proteína , Transformación Genética , Proteína p53 Supresora de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA