Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Br J Dermatol ; 180(1): 172-180, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30141192

RESUMEN

BACKGROUND: Data on dermatological manifestations of cardiofaciocutaneous syndrome (CFCS) remain heterogeneous and almost without expert dermatological classification. OBJECTIVES: To describe the dermatological manifestations of CFCS; to compare them with the literature findings; to assess those discriminating CFCS from other RASopathies, including Noonan syndrome (NS) and Costello syndrome (CS); and to test for dermatological phenotype-genotype correlations. METHODS: We performed a 4-year, large, prospective, multicentric, collaborative dermatological and genetic study. RESULTS: Forty-five patients were enrolled. Hair abnormalities were ubiquitous, including scarcity or absence of eyebrows and wavy or curly hair in 73% and 69% of patients, respectively. Keratosis pilaris (KP), ulerythema ophryogenes (UO), palmoplantar hyperkeratosis (PPHK) and multiple melanocytic naevi (MMN; over 50 naevi) were noted in 82%, 44%, 27% and 29% of patients, respectively. Scarcity or absence of eyebrows, association of UO and PPHK, diffuse KP and MMN best differentiated CFCS from NS and CS. Oral acitretin may be highly beneficial for therapeutic management of PPHK, whereas treatment of UO by topical sirolimus 1% failed. No significant dermatological phenotype-genotype correlation was determined. CONCLUSIONS: A thorough knowledge of CFCS skin manifestations would help in making a positive diagnosis and differentiating CFCS from CS and NS.


Asunto(s)
Displasia Ectodérmica/diagnóstico , Insuficiencia de Crecimiento/diagnóstico , Cardiopatías Congénitas/diagnóstico , Acitretina/administración & dosificación , Administración Cutánea , Administración Oral , Adolescente , Niño , Preescolar , Síndrome de Costello/diagnóstico , Diagnóstico Diferencial , Displasia Ectodérmica/tratamiento farmacológico , Displasia Ectodérmica/genética , Facies , Insuficiencia de Crecimiento/tratamiento farmacológico , Insuficiencia de Crecimiento/genética , Femenino , Francia , Estudios de Asociación Genética , Cardiopatías Congénitas/tratamiento farmacológico , Cardiopatías Congénitas/genética , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 2/genética , Masculino , Mutación , Síndrome de Noonan/diagnóstico , Estudios Prospectivos , Proteínas Proto-Oncogénicas B-raf/genética , Sirolimus/administración & dosificación , Resultado del Tratamiento , Adulto Joven
2.
Br J Dermatol ; 180(6): 1438-1448, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30417923

RESUMEN

BACKGROUND: Data on dermatological manifestations of Noonan syndrome (NS) remain heterogeneous and are based on limited dermatological expertise. OBJECTIVES: To describe the dermatological manifestations of NS, compare them with the literature findings, and test for dermatological phenotype-genotype correlations with or without the presence of PTPN11 mutations. METHODS: We performed a large 4-year, prospective, multicentric, collaborative dermatological and genetic study. RESULTS: Overall, 129 patients with NS were enrolled, including 65 patients with PTPN11-NS, 34 patients with PTPN11-NS with multiple lentigines (NSML), and 30 patients with NS who had a mutation other than PTPN11. Easy bruising was the most frequent dermatological finding in PTPN11-NS, present in 53·8% of patients. Multiple lentigines and café-au-lait macules (n ≥ 3) were present in 94% and 80% of cases of NSML linked to specific mutations of PTPN11, respectively. Atypical forms of NSML could be associated with NS with RAF1 or NRAS mutations. In univariate analysis, patients without a PTPN11 mutation showed (i) a significantly higher frequency of keratinization disorders (P = 0·001), including keratosis pilaris (P = 0·005), ulerythema ophryogenes (P = 0·0001) and palmar and/or plantar hyperkeratosis (P = 0·06, trend association), and (ii) a significantly higher frequency of scarce scalp hair (P = 0·035) and scarce or absent eyelashes (P = 0·06, trend association) than those with PTPN11 mutations. CONCLUSIONS: The cutaneous phenotype of NS with a PTPN11 mutation is generally mild and nonspecific, whereas the absence of a PTPN11 mutation is associated with a high frequency of keratinization disorders and hair abnormalities.


Asunto(s)
Estudios de Asociación Genética , Síndrome de Noonan/complicaciones , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Enfermedades de la Piel/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación , Síndrome de Noonan/genética , Fenotipo , Estudios Prospectivos , Adulto Joven
4.
J Med Genet ; 47(10): 686-91, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20543203

RESUMEN

BACKGROUND: CBL missense mutations have recently been associated with juvenile myelomonocytic leukaemia (JMML), an aggressive myeloproliferative and myelodysplastic neoplasm of early childhood characterised by excessive macrophage/monocyte proliferation. CBL, an E3 ubiquitin ligase and a multi-adaptor protein, controls proliferative signalling networks by downregulating the growth factor receptor signalling cascades in various cell types. METHODS AND RESULTS: CBL mutations were screened in 65 patients with JMML. A homozygous mutation of CBL was found in leukaemic cells of 4/65 (6%) patients. In all cases, copy neutral loss of heterozygosity of the 11q23 chromosomal region, encompassing the CBL locus, was demonstrated. Three of these four patients displayed additional features suggestive of an underlying developmental condition. A heterozygous germline CBL p.Y371H substitution was found in each of them and was inherited from the father in one patient. The germline mutation represents the first hit, with somatic loss of heterozygosity being the second hit positively selected in JMML cells. The three patients display a variable combination of dysmorphic features, hyperpigmented skin lesions and microcephaly that enable a 'CBL syndrome' to be tentatively delineated. Learning difficulties and postnatal growth retardation may be part of the phenotype. CONCLUSION: A report of germline mutations of CBL in three patients with JMML is presented here, confirming the existence of an unreported inheritable condition associated with a predisposition to JMML.


Asunto(s)
Mutación de Línea Germinal , Trastornos del Crecimiento , Leucemia Mielomonocítica Juvenil/genética , Microcefalia , Proteínas Proto-Oncogénicas c-cbl/genética , Niño , Preescolar , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Femenino , Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/complicaciones , Trastornos del Crecimiento/genética , Humanos , Leucemia Mielomonocítica Juvenil/complicaciones , Masculino , Microcefalia/complicaciones , Microcefalia/genética , Síndrome
5.
Arch Pediatr ; 28(5): 411-416, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34034929

RESUMEN

AIM: To evaluate the prognostic significance of initial central nervous system (CNS) involvement of children with acute lymphoblastic leukemia (ALL) enrolled in the EORTC 58951 trial. PATIENTS AND METHODS: From 1998 to 2008, 1930 ALL patients were included in the randomized EORTC 58951 trial. Overall treatment intensity was adjusted according to known prognostic factors including the level of minimal residual disease after induction treatment. CNS-directed therapy comprised four to 11 courses of i.v. methotrexate (5g/m2), and 10 to 19 intrathecal chemotherapy injections, depending on risk group and CNS status. Cranial irradiation was omitted for all patients. RESULTS: The overall 8-year event-free survival (EFS) and overall survival (OS) rates were 81.3% and 88.1%, respectively. In the CNS-1, TPL+, CNS-2, and CNS-3 groups, the 8-year EFS rates were 82.1%, 77.1%, 78.3%, and 57.4%, respectively. Multivariable analysis indicated that initial CNS-3 status, but not CNS-2 or TLP+, was an independent adverse predictor of outcome. The 8-year incidence of isolated CNS relapse was 1.7% and of isolated or combined CNS relapse it was 3.7%. NCI high-risk group, male sex, CNS-2 and CNS-3 status were independent predictors for a higher incidence of any CNS relapse. CONCLUSIONS: CNS-3 status remains associated with poor prognosis and requires intensification of both systemic and CNS-directed therapy. This trial was registered at https://clinicaltrials.gov/under/NCT00003728.


Asunto(s)
Sistema Nervioso Central/anomalías , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Valor Predictivo de las Pruebas , Adolescente , Biomarcadores de Tumor/análisis , Sistema Nervioso Central/fisiopatología , Niño , Preescolar , Irradiación Craneana/tendencias , Femenino , Humanos , Lactante , Masculino , Pediatría/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Pronóstico , Resultado del Tratamiento
8.
Diabetes Metab ; 35(3): 233-5, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19342262

RESUMEN

Activating mutations in genes KCNJ11 and ABCC8, which form the ATP-sensitive K+channel (K(ATP) channel), have been shown to cause transient or permanent neonatal diabetes. We describe here a rather different phenotype: two cases of adult diabetic patients-considered and treated as insulin-dependent diabetic patients since adolescence-who, in fact, turned out to be heterozygous for an ABCC8 mutation and able to successfully discontinue insulin while taking sulphonylurea treatment.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Autoanticuerpos/sangre , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Mutación , Canales de Potasio de Rectificación Interna/genética , Receptores de Droga/genética , Adolescente , Adulto , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Recién Nacido , Enfermedades del Recién Nacido/genética , Masculino , Persona de Mediana Edad , Receptores de Sulfonilureas
9.
Leukemia ; 33(8): 1910-1922, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30858550

RESUMEN

Minimal residual disease (MRD) is a powerful prognostic factor in acute lymphoblastic leukemia (ALL) and is used for patient stratification and treatment decisions, but its precise role in Philadelphia chromosome positive ALL is less clear. This uncertainty results largely from methodological differences relating to the use of real-time quantitative PCR (qRT-PCR) to measure BCR-ABL1 transcript levels for MRD analysis. We here describe the first results by the EURO-MRD consortium on standardization of qRT-PCR for the e1a2 BCR-ABL1 transcript in Ph + ALL, designed to overcome the lack of standardisation of laboratory procedures and data interpretation. Standardised use of EAC primer/probe sets and of centrally prepared plasmid standards had the greatest impact on reducing interlaboratory variability. In QC1 the proportion of analyses with BCR-ABL1/ABL1 ratios within half a log difference were 40/67 (60%) and 52/67 (78%) at 10-3 and 36/67 (53%) and 53/67 (79%) at 10-4BCR-ABL1/ABL1. Standardized RNA extraction, cDNA synthesis and cycler platforms did not improve results further, whereas stringent application of technical criteria for assay quality and uniform criteria for data interpretation and reporting were essential. We provide detailed laboratory recommendations for the standardized MRD analysis in routine diagnostic settings and in multicenter clinical trials for Ph + ALL.


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Cromosoma Filadelfia , Guías de Práctica Clínica como Asunto , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Consenso , Humanos , Neoplasia Residual , ARN Mensajero/análisis
11.
Leukemia ; 21(7): 1431-5, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17460701

RESUMEN

Data on secondary acute lymphoblastic leukaemia (sALL) following ALL treatment are very rare. However, the incidence might be underestimated as sALLs without a significant lineage shift might automatically be diagnosed as relapses. Examination of immunoglobulin and T-cell receptor gene rearrangements brought a new tool that can help in discrimination between relapse and sALL. We focused on the recurrences of childhood ALL to discover the real frequency of the sALL after ALL treatment. We compared clonal markers in matched presentation and recurrence samples of 366 patients treated according to the Berlin-Frankfurt-Munster (BFM)-based protocols. We found two cases of sALL and another three, where the recurrence is suspicious of being sALL rather than relapse. Our proposal for the 'secondary ALL after ALL' diagnostic criteria is as follows: (A) No clonal relationship between diagnosis and recurrence; (B) significant immunophenotypic shift--significant cytogenetic shift--gain/loss of a fusion gene. For the sALL (A) plus at least one (B) criterion should be fulfilled. With these criteria, the estimated frequency of the sALL after ALL is according to our data 0.5-1.5% of ALL recurrences on BFM-based protocols. Finally, we propose a treatment strategy for the patients with secondary disease.


Asunto(s)
Técnicas de Diagnóstico Molecular/métodos , Neoplasias Primarias Secundarias/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Antineoplásicos/efectos adversos , Preescolar , Diagnóstico Diferencial , Femenino , Reordenamiento Génico de Linfocito T , Genes de Inmunoglobulinas , Humanos , Inmunofenotipificación , Incidencia , Masculino , Neoplasias Primarias Secundarias/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/inducido químicamente , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Recurrencia
12.
Leukemia ; 21(4): 604-11, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17287850

RESUMEN

Most modern treatment protocols for acute lymphoblastic leukaemia (ALL) include the analysis of minimal residual disease (MRD). To ensure comparable MRD results between different MRD-polymerase chain reaction (PCR) laboratories, standardization and quality control are essential. The European Study Group on MRD detection in ALL (ESG-MRD-ALL), consisting of 30 MRD-PCR laboratories worldwide, has developed guidelines for the interpretation of real-time quantitative PCR-based MRD data. The application of these guidelines ensures identical interpretation of MRD data between different laboratories of the same MRD-based clinical protocol. Furthermore, the ESG-MRD-ALL guidelines will facilitate the comparison of MRD data obtained in different treatment protocols, including those with new drugs.


Asunto(s)
Reordenamiento Génico , Neoplasia Residual/genética , Reacción en Cadena de la Polimerasa/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Antígenos de Linfocitos T/genética , ADN de Neoplasias/genética , Genes de Inmunoglobulinas , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/microbiología
13.
Leukemia ; 21(1): 121-8, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17039236

RESUMEN

Recently, we and others described a new chromosomal rearrangement, that is, inv(7)(p15q34) and t(7;7)(p15;q34) involving the T-cell receptor beta (TCRbeta) (7q34) and the HOXA gene locus (7p15) in 5% of T-cell acute lymphoblastic leukemia (T-ALL) patients leading to transcriptional activation of especially HOXA10. To further address the clinical, immunophenotypical and molecular genetic findings of this chromosomal aberration, we studied 330 additional T-ALLs. This revealed TCRbeta-HOXA rearrangements in five additional patients, which brings the total to 14 cases in 424 patients (3.3%). Real-time quantitative PCR analysis for HOXA10 gene expression was performed in 170 T-ALL patients and detected HOXA10 overexpression in 25.2% of cases including all the cases with a TCRbeta-HOXA rearrangement (8.2%). In contrast, expression of the short HOXA10 transcript, HOXA10b, was almost exclusively found in the TCRbeta-HOXA rearranged cases, suggesting a specific role for the HOXA10b short transcript in TCRbeta-HOXA-mediated oncogenesis. Other molecular and/or cytogenetic aberrations frequently found in subtypes of T-ALL (SIL-TAL1, CALM-AF10, HOX11, HOX11L2) were not detected in the TCRbeta-HOXA rearranged cases except for deletion 9p21 and NOTCH1 activating mutations, which were present in 64 and 67%, respectively. In conclusion, this study defines TCRbeta-HOXA rearranged T-ALLs as a distinct cytogenetic subgroup by clinical, immunophenotypical and molecular genetic characteristics.


Asunto(s)
Proteínas de Homeodominio/genética , Leucemia-Linfoma de Células T del Adulto/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Adolescente , Adulto , Niño , Deleción Cromosómica , Inversión Cromosómica , Femenino , Reordenamiento Génico de Linfocito T , Proteínas Homeobox A10 , Humanos , Inmunofenotipificación , Leucemia-Linfoma de Células T del Adulto/patología , Leucemia-Linfoma de Células T del Adulto/fisiopatología , Masculino , Persona de Mediana Edad , Receptor Notch1/genética , Activación Transcripcional , Translocación Genética
14.
J Inherit Metab Dis ; 30(5): 827, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17703371

RESUMEN

The cardiofaciocutaneous (CFC) syndrome is characterized by congenital heart defect, developmental delay, peculiar facial appearance with bitemporal constriction, prominent forehead, downslanting palpebral fissures, curly sparse hair and abnormalities of the skin. CFC syndrome phenotypically overlaps with Noonan and Costello syndromes. Mutations of several genes (PTPN11, HRAS, KRAS, BRAF, MEK1 and MEK2), involved in the mitogen-activated protein kinase (MAPK) pathway, have been identified in CFC-Costello-Noonan patients. Coenzyme Q10 (CoQ10), a lipophilic molecule present in all cell membranes, functions as an electron carrier in the mitochondrial respiratory chain, where it transports electrons from complexes I and II to complex III. CoQ10 deficiency is a rare treatable mitochondrial disorder with various neurological (cerebellar ataxia, myopathy, epilepsy, mental retardation) and extraneurological (cardiomyopathy, nephropathy) signs that are responsive to CoQ10 supplementation. We report the case of a 4-year-old girl who presented a CFC syndrome, confirmed by the presence of a pathogenic R257Q BRAF gene mutation, together with a muscular CoQ10 deficiency. Her psychomotor development was severely impaired, hindered by muscular hypotonia and ataxia, both improving remarkably after CoQ10 treatment. This case suggests that there is a functional connection between the MAPK pathway and the mitochondria. This could be through the phosphorylation of a nuclear receptor essential for CoQ10 biosynthesis. Another hypothesis is that K-Ras, one of the proteins composing the MAPK pathway, might be recruited into the mitochondria to promote apoptosis. This case highlights that CoQ10 might contribute to the pathogenesis of CFC syndrome.


Asunto(s)
Anomalías Múltiples , Anomalías Craneofaciales/complicaciones , Cardiopatías Congénitas/complicaciones , Enfermedades Mitocondriales/complicaciones , Músculo Esquelético/enzimología , Anomalías Cutáneas/complicaciones , Ubiquinona/análogos & derivados , Anomalías Múltiples/enzimología , Preescolar , Coenzimas/deficiencia , Coenzimas/uso terapéutico , Anomalías Craneofaciales/enzimología , Femenino , Cardiopatías Congénitas/enzimología , Humanos , Sistema de Señalización de MAP Quinasas , Mitocondrias/enzimología , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/enzimología , Anomalías Cutáneas/enzimología , Síndrome , Resultado del Tratamiento , Ubiquinona/deficiencia , Ubiquinona/uso terapéutico
15.
J Med Genet ; 43(5): 401-5, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16443854

RESUMEN

BACKGROUND: Costello syndrome (CS) is a rare multiple congenital abnormality syndrome, associated with failure to thrive and developmental delay. One of the more distinctive features in childhood is the development of facial warts, often nasolabial and in other moist body surfaces. Individuals with CS have an increased risk of malignancy, suggested to be about 17%. Recently, mutations in the HRAS gene on chromosome 11p13.3 have been found to cause CS. METHODS: We report here the results of HRAS analysis in 43 individuals with a clinical diagnosis of CS. RESULTS: Mutations were found in 37 (86%) of patients. Analysis of parental DNA samples was possible in 16 cases for both parents and in three cases for one parent, and confirmed the mutations as de novo in all of these cases. Three novel mutations (G12C, G12E, and K117R) were found in five cases. CONCLUSIONS: These results confirm that CS is caused, in most cases, by heterozygous missense mutations in the proto-oncogene HRAS. Analysis of the major phenotypic features by mutation suggests a potential correlation between malignancy risk and genotype, which is highest for patients with an uncommon (G12A) substitution. These results confirm that mutation testing for HRAS is a reliable diagnostic test for CS.


Asunto(s)
Anomalías Múltiples/diagnóstico , Proteínas Proto-Oncogénicas p21(ras)/genética , Anomalías Múltiples/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Genotipo , Humanos , Lactante , Péptidos y Proteínas de Señalización Intracelular/genética , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Fenotipo , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Proto-Oncogenes Mas , Síndrome
16.
Arch Pediatr ; 14(11): 1356-65, 2007 Nov.
Artículo en Francés | MEDLINE | ID: mdl-17931842

RESUMEN

Transient (TNDM) and Permanent (PNDM) Neonatal Diabetes Mellitus are rare conditions occurring in about 1: 300,000 live births. In TNDM growth retarded infants develop diabetes in the first few weeks of life only to go into remission in a few months with possible relapse to a permanent diabetes state usually around adolescence or as adults. We believe that pancreatic dysfunction in this condition is maintained throughout life with relapse initiated at times of metabolic stress such as puberty or pregnancy. In PNDM, insulin secretory failure occurs in the late fetal or early postnatal period. A number of conditions are associated with PNDM, some of which have been elucidated at the molecular levels. Among those, the very recently elucidated mutations in KCNJ11 and ABCC8 gene, encoding the Kir6.2 and SUR1 subunit of the pancreatic K(ATP) channel involved in regulation of insulin secretion accounts for one third to a half of the PNDM cases. Patients with TNDM are more likely to have intrauterine growth retardation and less likely to develop ketoacidosis than patients with PNDM. In TNDM, patients are younger at the diagnosis of diabetes and have lower initial insulin requirements. Considerable overlap occurs between the two groups, so that TNDM cannot be distinguished from PNDM based on clinical features. Very early onset diabetes mellitus seems to be unrelated to autoimmunity in most instances. Recurrent diabetes is common in patients with "transient" neonatal diabetes mellitus and, consequently, prolonged follow-up is imperative. Molecular analysis of chromosome 6 anomalies, the KCNJ11 and ABCC8 genes encoding Kir6.2 and SUR1 provide a tool to identify transient from permanent neonatal diabetes mellitus in the neonatal period. This analysis also has potentially important therapeutic consequences leading to transfer some patients, those with mutations in KCNJ11 and ABCC8 from insulin therapy to sulfonylureas. Realizing how difficult it is to take care of a child of this age with diabetes mellitus should prompt clinicians to transfer these children to specialized centers. Insulin therapy and high caloric intake are the basis of the treatment. Insulin pump may offer an interesting therapeutic tool in this age group in experienced hands.


Asunto(s)
Diabetes Mellitus/etiología , Transportadoras de Casetes de Unión a ATP/genética , Cromosomas Humanos Par 6/genética , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/terapia , Retardo del Crecimiento Fetal , Asesoramiento Genético , Humanos , Hipoglucemiantes/uso terapéutico , Recién Nacido , Insulina/uso terapéutico , Sistemas de Infusión de Insulina , Mutación , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/genética , Pronóstico , Receptores de Droga/genética , Receptores de Sulfonilureas
17.
Arch Pediatr ; 24(5): 453-456, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28347637
18.
Leukemia ; 31(7): 1491-1501, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27899802

RESUMEN

Children with P2RY8-CRLF2-positive acute lymphoblastic leukemia have an increased relapse risk. Their mutational and transcriptional landscape, as well as the respective patterns at relapse remain largely elusive. We, therefore, performed an integrated analysis of whole-exome and RNA sequencing in 41 major clone fusion-positive cases including 19 matched diagnosis/relapse pairs. We detected a variety of frequently subclonal and highly instable JAK/STAT but also RTK/Ras pathway-activating mutations in 76% of cases at diagnosis and virtually all relapses. Unlike P2RY8-CRLF2 that was lost in 32% of relapses, all other genomic alterations affecting lymphoid development (58%) and cell cycle (39%) remained stable. Only IKZF1 alterations predominated in relapsing cases (P=0.001) and increased from initially 36 to 58% in matched cases. IKZF1's critical role is further corroborated by its specific transcriptional signature comprising stem cell features with signs of impaired lymphoid differentiation, enhanced focal adhesion, activated hypoxia pathway, deregulated cell cycle and increased drug resistance. Our findings support the notion that P2RY8-CRLF2 is dispensable for relapse development and instead highlight the prominent rank of IKZF1 for relapse development by mediating self-renewal and homing to the bone marrow niche. Consequently, reverting aberrant IKAROS signaling or its disparate programs emerges as an attractive potential treatment option in these leukemias.


Asunto(s)
Fusión Génica , Genómica , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Citocinas/genética , Receptores Purinérgicos P2Y/genética , Transcripción Genética , Adolescente , Niño , Preescolar , Dosificación de Gen , Genes Supresores de Tumor , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/fisiología , Lactante , Quinasas Janus/fisiología , Polimorfismo de Nucleótido Simple , Factores de Transcripción STAT/fisiología
19.
Diabetes ; 49(1): 108-13, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10615957

RESUMEN

Transient neonatal diabetes mellitus (TNDM) is estimated to occur in approximately 1 in 500,000 births and represents 50-60% of cases of neonatal diabetes. The pattern of inheritance of TNDM and its association with chromosome 6 uniparental disomy is consistent with the presence on chromosome 6 of an imprinted gene involved in pancreatic beta-cell development. Systematic screening for chromosome 6 abnormalities in nine families with 13 individuals affected by TNDM revealed paternal isodisomy of chromosome 6 in one child and paternally derived trisomy of the chromosomal region 6q in six children from three unrelated families. To delineate more accurately the region suspected of harboring the gene of interest, precise mapping of the duplicated area was performed in children exhibiting partial 6q trisomy by using microsatellite markers. The smallest region of duplication observed in our patients was flanked by markers D6S308 and D6S1010, which are separated by <1 cM. These findings confirm that TNDM may result from the overexpression of a gene located on chromosome 6q that is exclusively expressed from the paternal allele at least during some periods of life and further refine the localization of this gene.


Asunto(s)
Cromosomas Humanos Par 6/genética , Diabetes Mellitus/genética , Enfermedades del Recién Nacido/genética , Aneuploidia , Padre , Femenino , Pruebas Genéticas , Impresión Genómica , Humanos , Lactante , Recién Nacido , Masculino , Repeticiones de Microsatélite , Linaje , Trisomía
20.
Leukemia ; 10(9): 1486-91, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8751467

RESUMEN

We have carried out a screening for loss of heterozygosity (LOH) in 51 children with B-lineage acute lymphoblastic leukemia (ALL). Forty-six highly polymorphic microsatellite markers located in subtelomeric areas of every chromosome arm were analyzed in each patient. Allelic losses were encountered at 21 of the 46 loci tested (46%). The frequency of LOH at a given locus was usually < 10% and fractional allelic loss, calculated as the ratio of chromosomal arms displaying loss among all informative arms for each patient, ranged from 0.025 to 0.31 (mean, 0.063). This study provides further evidence that deletional events are a major type of genetic alteration found in childhood ALL. The diversity of the loci displaying LOH suggests that, as in solid tumors, numerous tumor suppressor genes are likely to participate in leukemogenesis. However, the overall low frequency of LOH, as well as the absence of microsatellite instability suggest that the genetic instability is lower in childhood ALL than in most of the solid tumors.


Asunto(s)
Linfoma de Burkitt/genética , Eliminación de Gen , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Alelos , Niño , Aberraciones Cromosómicas , ADN de Neoplasias/genética , ADN Satélite/genética , Diploidia , Marcadores Genéticos , Heterocigoto , Humanos , Cariotipificación , Telómero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA