Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
2.
Blood ; 139(24): 3505-3518, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35316324

RESUMEN

Oncogenic alterations underlying B-cell acute lymphoblastic leukemia (B-ALL) in adults remain incompletely elucidated. To uncover novel oncogenic drivers, we performed RNA sequencing and whole-genome analyses in a large cohort of unresolved B-ALL. We identified a novel subtype characterized by a distinct gene expression signature and the unique association of 2 genomic microdeletions. The 17q21.31 microdeletion resulted in a UBTF::ATXN7L3 fusion transcript encoding a chimeric protein. The 13q12.2 deletion resulted in monoallelic ectopic expression of the homeobox transcription factor CDX2, located 138 kb in cis from the deletion. Using 4C-sequencing and CRISPR interference experiments, we elucidated the mechanism of CDX2 cis-deregulation, involving PAN3 enhancer hijacking. CDX2/UBTF ALL (n = 26) harbored a distinct pattern of additional alterations including 1q gain and CXCR4 activating mutations. Within adult patients with Ph- B-ALL enrolled in GRAALL trials, patients with CDX2/UBTF ALL (n = 17/723, 2.4%) were young (median age, 31 years) and dramatically enriched in females (male/female ratio, 0.2, P = .002). They commonly presented with a pro-B phenotype ALL and moderate blast cell infiltration. They had poor response to treatment including a higher risk of failure to first induction course (19% vs 3%, P = .017) and higher post-induction minimal residual disease (MRD) levels (MRD ≥ 10-4, 93% vs 46%, P < .001). This early resistance to treatment translated into a significantly higher cumulative incidence of relapse (75.0% vs 32.4%, P = .004) in univariate and multivariate analyses. In conclusion, we discovered a novel B-ALL entity defined by the unique combination of CDX2 cis-deregulation and UBTF::ATXN7L3 fusion, representing a high-risk disease in young adults.


Asunto(s)
Factor de Transcripción CDX2 , Proteínas del Complejo de Iniciación de Transcripción Pol1 , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Factores de Transcripción , Adulto , Factor de Transcripción CDX2/genética , Femenino , Genes Homeobox , Humanos , Masculino , Neoplasia Residual/genética , Proteínas de Fusión Oncogénica , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción/genética
4.
Sci Rep ; 11(1): 2801, 2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531590

RESUMEN

Juvenile myelomonocytic leukemia (JMML) treatment primarily relies on hematopoietic stem cell transplantation and results in long-term overall survival of 50-60%, demonstrating a need to develop novel treatments. Dysregulation of the non-coding RNA transcriptome has been demonstrated before in this rare and unique disorder of early childhood. In this study, we investigated the therapeutic potential of targeting overexpressed long non-coding RNAs (lncRNAs) in JMML. Total RNA sequencing of bone marrow and peripheral blood mononuclear cell preparations from 19 untreated JMML patients and three healthy children revealed 185 differentially expressed lncRNA genes (131 up- and 54 downregulated). LNA GapmeRs were designed for 10 overexpressed and validated lncRNAs. Molecular knockdown (≥ 70% compared to mock control) after 24 h of incubation was observed with two or more independent GapmeRs in 6 of them. For three lncRNAs (lnc-THADA-4, lnc-ACOT9-1 and NRIR) knockdown resulted in a significant decrease of cell viability after 72 h of incubation in primary cultures of JMML mononuclear cells, respectively. Importantly, the extent of cellular damage correlated with the expression level of the lncRNA of interest. In conclusion, we demonstrated in primary JMML cell cultures that knockdown of overexpressed lncRNAs such as lnc-THADA-4, lnc-ACOT9-1 and NRIR may be a feasible therapeutic strategy.


Asunto(s)
Antineoplásicos/farmacología , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Mielomonocítica Juvenil/genética , ARN Largo no Codificante/metabolismo , Adolescente , Antineoplásicos/uso terapéutico , Médula Ósea/patología , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Técnicas de Silenciamiento del Gen , Voluntarios Sanos , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/sangre , Leucemia Mielomonocítica Juvenil/tratamiento farmacológico , Leucemia Mielomonocítica Juvenil/patología , Leucocitos Mononucleares , Masculino , Cultivo Primario de Células , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , RNA-Seq , Células Tumorales Cultivadas
6.
Leukemia ; 34(6): 1658-1668, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31776464

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare aggressive myelodysplastic/myeloproliferative neoplasm of early childhood, initiated by RAS-activating mutations. Genomic analyses have recently described JMML mutational landscape; however, the nature of JMML-propagating cells (JMML-PCs) and the clonal architecture of the disease remained until now elusive. Combining genomic (exome, RNA-seq), Colony forming assay and xenograft studies, we detect the presence of JMML-PCs that faithfully reproduce JMML features including the complex/nonlinear organization of dominant/minor clones, both at diagnosis and relapse. Further integrated analysis also reveals that although the mutations are acquired in hematopoietic stem cells, JMML-PCs are not always restricted to this compartment, highlighting the heterogeneity of the disease during the initiation steps. We show that the hematopoietic stem/progenitor cell phenotype is globally maintained in JMML despite overexpression of CD90/THY-1 in a subset of patients. This study shed new lights into the ontogeny of JMML, and the identity of JMML-PCs, and provides robust models to monitor the disease and test novel therapeutic approaches.


Asunto(s)
Células Madre Hematopoyéticas/patología , Leucemia Mielomonocítica Juvenil/patología , Células Madre Neoplásicas/patología , Adolescente , Animales , Niño , Preescolar , Femenino , Xenoinjertos , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/genética , Masculino , Ratones , Mutación
7.
Haematologica ; 103(8): 1278-1287, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29724903

RESUMEN

Heterozygous germline GATA2 mutations strongly predispose to leukemia, immunodeficiency, and/or lymphoedema. We describe a series of 79 patients (53 families) diagnosed since 2011, made up of all patients in France and Belgium, with a follow up of 2249 patients/years. Median age at first clinical symptoms was 18.6 years (range, 0-61 years). Severe infectious diseases (mycobacteria, fungus, and human papilloma virus) and hematologic malignancies were the most common first manifestations. The probability of remaining symptom-free was 8% at 40 years old. Among the 53 probands, 24 had missense mutations including 4 recurrent alleles, 21 had nonsense or frameshift mutations, 4 had a whole-gene deletion, 2 had splice defects, and 2 patients had complex mutations. There were significantly more cases of leukemia in patients with missense mutations (n=14 of 34) than in patients with nonsense or frameshift mutations (n=2 of 28). We also identify new features of the disease: acute lymphoblastic leukemia, juvenile myelomonocytic leukemia, fatal progressive multifocal leukoencephalopathy related to the JC virus, and immune/inflammatory diseases. A revised International Prognostic Scoring System (IPSS) score allowed a distinction to be made between a stable disease and hematologic transformation. Chemotherapy is of limited efficacy, and has a high toxicity with severe infectious complications. As the mortality rate is high in our cohort (up to 35% at the age of 40), hematopoietic stem cell transplantation (HSCT) remains the best choice of treatment to avoid severe infectious and/or hematologic complications. The timing of HSCT remains difficult to determine, but the earlier it is performed, the better the outcome.


Asunto(s)
Deficiencia GATA2/epidemiología , Mutación de Línea Germinal , Adulto Joven , Adolescente , Adulto , Bélgica , Niño , Preescolar , Francia , Deficiencia GATA2/complicaciones , Deficiencia GATA2/genética , Deficiencia GATA2/terapia , Neoplasias Hematológicas/etiología , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Recién Nacido , Infecciones/etiología , Persona de Mediana Edad , Mortalidad , Pronóstico , Encuestas y Cuestionarios
8.
Oncotarget ; 7(27): 41599-41611, 2016 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-27191650

RESUMEN

T cell acute lymphoblastic leukemia (T-ALL) develops through accumulation of multiple genomic alterations within T-cell progenitors resulting in clonal heterogeneity among leukemic cells. Human T-ALL xeno-transplantation in immunodeficient mice is a gold standard approach to study leukemia biology and we recently uncovered that the leukemia development is more or less rapid depending on T-ALL sample. The resulting human leukemia may arise through genetic selection and we previously showed that human T-ALL development in immune-deficient mice is significantly enhanced upon CD7+/CD34+ leukemic cell transplantations. Here we investigated the genetic characteristics of CD7+/CD34+ and CD7+/CD34- cells from newly diagnosed human T-ALL and correlated it to the speed of leukemia development. We observed that CD7+/CD34+ or CD7+/CD34- T-ALL cells that promote leukemia within a short-time period are genetically similar, as well as xenograft-derived leukemia resulting from both cell fractions. In the case of delayed T-ALL growth CD7+/CD34+ or CD7+/CD34- cells were either genetically diverse, the resulting xenograft leukemia arising from different but branched subclones present in the original sample, or similar, indicating decreased fitness to mouse micro-environment. Altogether, our work provides new information relating the speed of leukemia development in xenografts to the genetic diversity of T-ALL cell compartments.


Asunto(s)
Variación Genética , Trasplante de Neoplasias , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Trasplante Heterólogo , Animales , Antígenos CD34/metabolismo , Línea Celular Tumoral , Niño , Progresión de la Enfermedad , Heterogeneidad Genética , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Ratones Transgénicos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Factores de Tiempo , Adulto Joven
9.
Eur J Med Genet ; 59(3): 173-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26855057

RESUMEN

Noonan syndrome is associated with a range of malignancies including acute lymphoblastic leukemia (ALL). However, little information is available regarding the frequency, natural history, characteristics and prognosis of ALL in Noonan syndrome or RASopathies in general. Cross-referencing data from a large prospective cohort of 1176 patients having a molecularly confirmed RASopathy with data from the French childhood cancer registry allowed us to identify ALL in 6 (0.5%) patients including 4/778 (0.5%) with a germline PTPN11 mutation and 2/94 (2.1%) with a germline SOS1 mutation. None of the patients of our series with CFC syndrome (with germline BRAF or MAP2K1/MAP2K2 mutation - n = 121) or Costello syndrome (with HRAS mutation - n = 35) had an ALL. A total of 19 Noonan-ALL were gathered by adding our patients to those of the International Berlin-Munster-Frankfurt (I-BFM) study group and previously reported patients. Strikingly, all Noonan-associated ALL were B-cell precursor ALL, and high hyperdiploidy with more than 50 chromosomes was found in the leukemia cells of 13/17 (76%) patients with available genetics data. Our data suggest that children with Noonan syndrome are at higher risk to develop ALL. Like what is observed for somatic PTPN11 mutations, NS is preferentially associated with the development of hyperdiploid ALL that will usually respond well to chemotherapy. However, Noonan syndrome patients seem to have a propensity to develop post therapy myelodysplasia that can eventually be fatal. Hence, one should be particularly cautious when treating these patients.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras/epidemiología , Leucemia-Linfoma Linfoblástico de Células Precursoras/etiología , Proteínas ras/genética , Adolescente , Niño , Preescolar , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Neoplasias Primarias Secundarias/etiología , Síndrome de Noonan/complicaciones , Síndrome de Noonan/genética , Síndrome de Noonan/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Prevalencia , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína SOS1/genética , Proteínas ras/metabolismo
10.
Eur J Hum Genet ; 24(8): 1124-31, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26757980

RESUMEN

Noonan syndrome is a heterogeneous autosomal dominant disorder caused by mutations in at least eight genes involved in the RAS/MAPK signaling pathway. Recently, RIT1 (Ras-like without CAAX 1) has been shown to be involved in the pathogenesis of some patients. We report a series of 44 patients from 30 pedigrees (including nine multiplex families) with mutations in RIT1. These patients display a typical Noonan gestalt and facial phenotype. Among the probands, 8.7% showed postnatal growth retardation, 90% had congenital heart defects, 36% had hypertrophic cardiomyopathy (a lower incidence compared with previous report), 50% displayed speech delay and 52% had learning difficulties, but only 22% required special education. None had major skin anomalies. One child died perinatally of juvenile myelomonocytic leukemia. Compared with the canonical Noonan phenotype linked to PTPN11 mutations, patients with RIT1 mutations appear to be less severely growth retarded and more frequently affected by cardiomyopathy. Based on our experience, we estimate that RIT1 could be the cause of 5% of Noonan syndrome patients. Because mutations found constitutionally in Noonan syndrome are also found in several tumors in adulthood, we evaluated the potential contribution of RIT1 to leukemogenesis in Noonan syndrome. We screened 192 pediatric cases of acute lymphoblastic leukemias (96 B-ALL and 96 T-ALL) and 110 cases of juvenile myelomonocytic leukemias (JMML), but detected no variation in these tumoral samples, suggesting that Noonan patients with germline RIT1 mutations are not at high risk to developing JMML or ALL, and that RIT1 has at most a marginal role in these sporadic malignancies.


Asunto(s)
Leucemia Mielomonocítica Juvenil/genética , Mutación , Síndrome de Noonan/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas ras/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Leucemia Mielomonocítica Juvenil/patología , Masculino , Síndrome de Noonan/patología , Linaje , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología
11.
Blood ; 127(9): 1163-72, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26712910

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare and aggressive stem cell disease of early childhood. RAS activation constitutes the core component of oncogenic signaling. In addition, leukemic blasts in one-fourth of JMML patients present with monosomy 7, and more than half of patients show elevated age-adjusted fetal hemoglobin (HbF) levels. Hematopoietic stem cell transplantation is the current standard of care and results in an event-free survival rate of 50% to 60%, indicating that novel molecular-driven therapeutic options are urgently needed. Using gene expression profiling in a series of 82 patient samples, we aimed at understanding the molecular biology behind JMML and identified a previously unrecognized molecular subgroup characterized by high LIN28B expression. LIN28B overexpression was significantly correlated with higher HbF levels, whereas patients with monosomy 7 seldom showed enhanced LIN28B expression. This finding gives a biological explanation of why patients with monosomy 7 are rarely diagnosed with high age-adjusted HbF levels. In addition, this new fetal-like JMML subgroup presented with reduced levels of most members of the let-7 microRNA family and showed characteristic overexpression of genes involved in fetal hematopoiesis and stem cell self-renewal. Lastly, high LIN28B expression was associated with poor clinical outcome in our JMML patient series but was not independent from other prognostic factors such as age and age-adjusted HbF levels. In conclusion, we identified elevated LIN28B expression as a hallmark of a novel fetal-like subgroup in JMML.


Asunto(s)
Feto/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Proteínas de Unión al ARN/genética , Biomarcadores de Tumor/metabolismo , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 7/genética , Supervivencia sin Enfermedad , Femenino , Hemoglobina Fetal/metabolismo , Regulación Leucémica de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Masculino , Análisis Multivariante , Pronóstico , Proteínas de Unión al ARN/metabolismo
12.
Cell Rep ; 13(3): 504-515, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26456833

RESUMEN

Somatic PTPN11 mutations cause juvenile myelomonocytic leukemia (JMML). Germline PTPN11 defects cause Noonan syndrome (NS), and specific inherited mutations cause NS/JMML. Here, we report that hematopoietic cells differentiated from human induced pluripotent stem cells (hiPSCs) harboring NS/JMML-causing PTPN11 mutations recapitulated JMML features. hiPSC-derived NS/JMML myeloid cells exhibited increased signaling through STAT5 and upregulation of miR-223 and miR-15a. Similarly, miR-223 and miR-15a were upregulated in 11/19 JMML bone marrow mononuclear cells harboring PTPN11 mutations, but not those without PTPN11 defects. Reducing miR-223's function in NS/JMML hiPSCs normalized myelogenesis. MicroRNA target gene expression levels were reduced in hiPSC-derived myeloid cells as well as in JMML cells with PTPN11 mutations. Thus, studying an inherited human cancer syndrome with hiPSCs illuminated early oncogenesis prior to the accumulation of secondary genomic alterations, enabling us to discover microRNA dysregulation, establishing a genotype-phenotype association for JMML and providing therapeutic targets.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Leucemia Mielomonocítica Juvenil/metabolismo , Células Mieloides/citología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patología , MicroARNs/genética , Mutación , Células Mieloides/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética , Regulación hacia Arriba
13.
Nat Genet ; 47(11): 1334-40, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26457648

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare and severe myelodysplastic and myeloproliferative neoplasm of early childhood initiated by germline or somatic RAS-activating mutations. Genetic profiling and whole-exome sequencing of a large JMML cohort (118 and 30 cases, respectively) uncovered additional genetic abnormalities in 56 cases (47%). Somatic events were rare (0.38 events/Mb/case) and restricted to sporadic (49/78; 63%) or neurofibromatosis type 1 (NF1)-associated (8/8; 100%) JMML cases. Multiple concomitant genetic hits targeting the RAS pathway were identified in 13 of 78 cases (17%), disproving the concept of mutually exclusive RAS pathway mutations and defining new pathways activated in JMML involving phosphoinositide 3-kinase (PI3K) and the mTORC2 complex through RAC2 mutation. Furthermore, this study highlights PRC2 loss (26/78; 33% of sporadic JMML cases) that switches the methylation/acetylation status of lysine 27 of histone H3 in JMML cases with altered RAS and PRC2 pathways. Finally, the association between JMML outcome and mutational profile suggests a dose-dependent effect for RAS pathway activation, distinguishing very aggressive JMML rapidly progressing to acute myeloid leukemia.


Asunto(s)
Redes Reguladoras de Genes/genética , Leucemia Mielomonocítica Juvenil/genética , Mutación , Complejo Represivo Polycomb 2/genética , Transducción de Señal/genética , Proteínas ras/genética , Acetilación , Enfermedad Aguda , Niño , Preescolar , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Femenino , Histonas/metabolismo , Humanos , Lactante , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mielomonocítica Juvenil/metabolismo , Masculino , Metilación , Microscopía Confocal , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Análisis de Secuencia de ADN/métodos , Análisis de Supervivencia , Transcriptoma , Proteínas ras/metabolismo
14.
Haematologica ; 100(10): 1311-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26137961

RESUMEN

DNA copy number analysis has been instrumental for the identification of genetic alterations in B-cell precursor acute lymphoblastic leukemia. Notably, some of these genetic defects have been associated with poor treatment outcome and might be relevant for future risk stratification. In this study, we characterized recurrent deletions of CD200 and BTLA genes, mediated by recombination-activating genes, and used breakpoint-specific polymerase chain reaction assay to screen a cohort of 1154 cases of B-cell precursor acute lymphoblastic leukemia uniformly treated according to the EORTC-CLG 58951 protocol. CD200/BTLA deletions were identified in 56 of the patients (4.8%) and were associated with an inferior 8-year event free survival in this treatment protocol [70.2% ± 1.2% for patients with deletions versus 83.5% ± 6.4% for non-deleted cases (hazard ratio 2.02; 95% confidence interval 1.23-3.32; P=0.005)]. Genetically, CD200/BTLA deletions were strongly associated with ETV6-RUNX1-positive leukemias (P<0.0001), but were also identified in patients who did not have any genetic abnormality that is currently used for risk stratification. Within the latter population of patients, the presence of CD200/BTLA deletions was associated with inferior event-free survival and overall survival. Moreover, the multivariate Cox model indicated that these deletions had independent prognostic impact on event-free survival when adjusting for conventional risk criteria. All together, these findings further underscore the rationale for copy number profiling as an important tool for risk stratification in human B-cell precursor acute lymphoblastic leukemia. This trial was registered at www.ClinicalTrials.gov as #NCT00003728.


Asunto(s)
Antígenos CD/genética , Eliminación de Gen , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Receptores Inmunológicos/genética , Adolescente , Alelos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Niño , Preescolar , Puntos de Rotura del Cromosoma , Ensayos Clínicos como Asunto , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Femenino , Frecuencia de los Genes , Humanos , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/mortalidad , Pronóstico , Recurrencia
15.
J Med Genet ; 51(10): 689-97, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25097206

RESUMEN

BACKGROUND: Infants with Noonan syndrome (NS) are predisposed to developing juvenile myelomonocytic leukaemia (JMML) or JMML-like myeloproliferative disorders (MPD). Whereas sporadic JMML is known to be aggressive, JMML occurring in patients with NS is often considered as benign and transitory. However, little information is available regarding the occurrence and characteristics of JMML in NS. METHODS AND RESULTS: Within a large prospective cohort of 641 patients with a germline PTPN11 mutation, we identified MPD features in 36 (5.6%) patients, including 20 patients (3%) who fully met the consensus diagnostic criteria for JMML. Sixty percent of the latter (12/20) had severe neonatal manifestations, and 10/20 died in the first month of life. Almost all (11/12) patients with severe neonatal JMML were males. Two females who survived MPD/JMML subsequently developed another malignancy during childhood. Although the risk of developing MPD/JMML could not be fully predicted by the underlying PTPN11 mutation, some germline PTPN11 mutations were preferentially associated with myeloproliferation: 10/48 patients with NS (20.8%) with a mutation in codon Asp61 developed MPD/JMML in infancy. Patients with a p.Thr73Ile mutation also had more chances of developing MPD/JMML but with a milder clinical course. SNP array and whole exome sequencing in paired tumoral and constitutional samples identified no second acquired somatic mutation to explain the occurrence of myeloproliferation. CONCLUSIONS: JMML represents the first cause of death in PTPN11-associated NS. Few patients have been reported so far, suggesting that JMML may sometimes be overlooked due to early death, comorbidities or lack of confirmatory tests.


Asunto(s)
Leucemia Mielomonocítica Juvenil/complicaciones , Leucemia Mielomonocítica Juvenil/genética , Síndrome de Noonan/complicaciones , Síndrome de Noonan/genética , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Leucemia Mielomonocítica Juvenil/mortalidad , Leucemia Mielomonocítica Juvenil/fisiopatología , Masculino , Mutación , Síndrome de Noonan/mortalidad , Síndrome de Noonan/fisiopatología , Estudios Prospectivos , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética
16.
Hum Mol Genet ; 23(16): 4315-27, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24705357

RESUMEN

RASopathies, a family of disorders characterized by cardiac defects, defective growth, facial dysmorphism, variable cognitive deficits and predisposition to certain malignancies, are caused by constitutional dysregulation of RAS signalling predominantly through the RAF/MEK/ERK (MAPK) cascade. We report on two germline mutations (p.Gly39dup and p.Val55Met) in RRAS, a gene encoding a small monomeric GTPase controlling cell adhesion, spreading and migration, underlying a rare (2 subjects among 504 individuals analysed) and variable phenotype with features partially overlapping Noonan syndrome, the most common RASopathy. We also identified somatic RRAS mutations (p.Gly39dup and p.Gln87Leu) in 2 of 110 cases of non-syndromic juvenile myelomonocytic leukaemia, a childhood myeloproliferative/myelodysplastic disease caused by upregulated RAS signalling, defining an atypical form of this haematological disorder rapidly progressing to acute myeloid leukaemia. Two of the three identified mutations affected known oncogenic hotspots of RAS genes and conferred variably enhanced RRAS function and stimulus-dependent MAPK activation. Expression of an RRAS mutant homolog in Caenorhabditis elegans enhanced RAS signalling and engendered protruding vulva, a phenotype previously linked to the RASopathy-causing SHOC2(S2G) mutant. Overall, these findings provide evidence of a functional link between RRAS and MAPK signalling and reveal an unpredicted role of enhanced RRAS function in human disease.


Asunto(s)
Carcinogénesis/genética , Mutación/fisiología , Fenotipo , Proteínas ras/genética , Animales , Caenorhabditis elegans , Estudios de Cohortes , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Síndrome de Noonan/genética , Proteína Oncogénica v-akt/metabolismo , Transducción de Señal/genética , Proteínas ras/química , Proteínas ras/metabolismo
17.
Haematologica ; 98(4): 597-601, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23065506

RESUMEN

Deletion of the Ikaros (IKZF1) gene is an oncogenic lesion frequently associated with BCR-ABL1-positive acute lymphoblastic leukemias. It is also found in a fraction of BCR-ABL1-negative B-cell precursor acute lymphoblastic leukemias, and early studies showed it was associated with a higher risk of relapse. Therefore, screening tools are needed for evaluation in treatment protocols and possible inclusion in risk stratification. Besides monosomy 7 and large 7p abnormalities encompassing IKZF1, most IKZF1 alterations are short, intragenic deletions. Based on cohorts of patients, we mapped the microdeletion breakpoints and developed a breakpoint-specific fluorescent multiplex polymerase chain reaction that allows detection of recurrent intragenic deletions. This sensitive test could also detect IKZF1 subclonal deletions, whose prognostic significance should be evaluated. Moreover, we show that consensus breakpoint sequences can be used as clonal markers to monitor minimal residual disease. This paper could be useful for translational studies and in clinical management of BCP-ALL.


Asunto(s)
Factor de Transcripción Ikaros/genética , Reacción en Cadena de la Polimerasa Multiplex/métodos , Neoplasia Residual/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Eliminación de Secuencia , Adolescente , Niño , Preescolar , Puntos de Rotura del Cromosoma , Mapeo Cromosómico , Cromosomas Humanos Par 7/genética , Estudios de Cohortes , Hibridación Genómica Comparativa , ADN Intergénico/genética , Femenino , Pruebas Genéticas/métodos , Humanos , Lactante , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Pronóstico , Reproducibilidad de los Resultados , Factores de Riesgo , Sensibilidad y Especificidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...