Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 137(4): 485-492, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33067622

RESUMEN

Erdheim-Chester disease (ECD) is a clonal hematopoietic disorder characterized by the accumulation of foamy histiocytes within organs (in particular, frequent retroperitoneal involvement) and a high frequency of BRAFV600E mutations. Although ECD is not commonly recognized to have overt peripheral blood (PB) or bone marrow (BM) disease, we recently identified that ECD patients have a high frequency of a concomitant myeloid malignancy. We thus conducted a systematic clinical and molecular analysis of the BM from 120 ECD patients. Surprisingly, 42.5% of ECD patients (51 of 120) had clonal hematopoiesis whereas 15.8% of patients (19 of 120) developed an overt hematologic malignancy (nearly all of which were a myeloid neoplasm). The most frequently mutated genes in BM were TET2, ASXL1, DNMT3A, and NRAS. ECD patients with clonal hematopoiesis were more likely to be older (P < .0001), have retroperitoneal involvement (P = .02), and harbor a BRAFV600E mutation (P = .049) than those without clonal hematopoiesis. The presence of the TET2 mutation was associated with a BRAFV600E mutation in tissue ECD lesions (P = .0006) and TET2-mutant ECD patients were more likely to have vascular involvement than TET2 wild-type ECD patients. Clonal hematopoiesis mutations in ECD were detected in cells derived from CD34+CD38- BM progenitors and PB monocytes but less frequently present in PB B and T lymphocytes. These data identify a heretofore unrecognized high frequency of clonal hematopoiesis in ECD patients, reaffirm the development of additional high risk of myeloid neoplasms in ECD, and provide evidence of a BM-based precursor cell of origin for many patients with ECD.


Asunto(s)
Hematopoyesis Clonal , Enfermedad de Erdheim-Chester/fisiopatología , Cariotipo Anormal , Adulto , Factores de Edad , Anciano , Médula Ósea/patología , Transformación Celular Neoplásica/genética , Hematopoyesis Clonal/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Progresión de la Enfermedad , Enfermedad de Erdheim-Chester/genética , Exones/genética , Femenino , Genes Relacionados con las Neoplasias , Humanos , Leucemia Mieloide/genética , Masculino , Persona de Mediana Edad , Mieloma Múltiple/genética , Mutación , Síndromes Mielodisplásicos/genética , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/patología , Especificidad de Órganos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética
2.
Blood ; 136(5): 585-595, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32457988

RESUMEN

Epigenetic changes during B-cell differentiation generate distinct DNA methylation signatures specific for B-cell subsets, including memory B cells (MBCs) and plasma cells (PCs). Waldenström macroglobulinemia (WM) is a B-cell malignancy uniquely comprising a mixture of lymphocytic and plasmacytic phenotypes. Here, we integrated genome-wide DNA methylation, transcriptome, mutation, and phenotypic features of tumor cells from 35 MYD88-mutated WM patients in relation to normal plasma and B-cell subsets. Patients naturally segregate into 2 groups according to DNA methylation patterns, related to normal MBC and PC profiles, and reminiscent of other memory and PC-derived malignancies. Concurrent analysis of DNA methylation changes in normal and WM development captured tumor-specific events, highlighting a selective reprogramming of enhancer regions in MBC-like WM and repressed and heterochromatic regions in PC-like WM. MBC-like WM hypomethylation was enriched in motifs belonging to PU.1, TCF3, and OCT2 transcription factors and involved elevated MYD88/TLR pathway activity. PC-like WM displayed marked global hypomethylation and selective overexpression of histone genes. Finally, WM subtypes exhibited differential genetic, phenotypic, and clinical features. MBC-like WM harbored significantly more clonal CXCR4 mutations (P = .015), deletion 13q (P = .006), splenomegaly (P = .02), and thrombocytopenia (P = .004), whereas PC-like WM harbored more deletion 6q (P = .012), gain 6p (P = .033), had increased frequencies of IGHV3 genes (P = .002), CD38 expression (P = 4.1e-5), and plasmacytic differentiation features (P = .008). Together, our findings illustrate a novel approach to subclassify WM patients using DNA methylation and reveal divergent molecular signatures among WM patients.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Metilación de ADN/genética , Células Plasmáticas/inmunología , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/inmunología , Humanos , Macroglobulinemia de Waldenström/clasificación
3.
Blood ; 119(14): 3211-8, 2012 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-22343920

RESUMEN

A cohort of MDS patients was examined for mutations affecting 4 splice genes (SF3B1, SRSF2, ZRSR2, and U2AF35) and evaluated in the context of clinical and molecular markers. Splice gene mutations were detected in 95 of 221 patients. These mutations were mutually exclusive and less likely to occur in patients with complex cytogenetics or TP53 mutations. SF3B1(mut) patients presented with lower hemoglobin levels, increased WBC and platelet counts, and were more likely to have DNMT3A mutations. SRSF2(mut) patients clustered in RAEB-1 and RAEB-2 subtypes and exhibited pronounced thrombocytopenias. ZRSR2(mut) patients clustered in International Prognostic Scoring System intermediate-1 and intermediate-2 risk groups, had higher percentages of bone marrow blasts, and more often displayed isolated neutropenias. SRSF2 and ZRSR2 mutations were more common in TET2(mut) patients. U2AF35(mut) patients had an increased prevalence of chromosome 20 deletions and ASXL1 mutations. Multivariate analysis revealed an inferior overall survival and a higher AML transformation rate for the genotype ZRSR2(mut)/TET2(wt) (overall survival: hazard ratio = 3.3; 95% CI, 1.4-7.7; P = .006; AML transformation: hazard ratio = 3.6; 95% CI, 2-4.2; P = .026). Our results demonstrate that splice gene mutations are among the most frequent molecular aberrations in myelodysplastic syndrome, define distinct clinical phenotypes, and show preferential associations with mutations targeting transcriptional regulation.


Asunto(s)
Mutación , Síndromes Mielodisplásicos/genética , Fenotipo , Empalme del ARN/genética , Adulto , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/genética , Femenino , Estudios de Asociación Genética , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/mortalidad , Proteínas Nucleares/genética , Fosfoproteínas/genética , Pronóstico , Factores de Empalme de ARN , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteínas/genética , Factores de Empalme Serina-Arginina , Factor de Empalme U2AF , Análisis de Supervivencia
4.
Haematologica ; 98(11): 1748-52, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23872306

RESUMEN

STAT3 protein phosphorylation is a frequent event in various hematologic malignancies and solid tumors. Acquired STAT3 mutations have been recently identified in 40% of patients with T-cell large granular lymphocytic leukemia, a rare T-cell disorder. In this study, we investigated the mutational status of STAT3 in a large series of patients with lymphoid and myeloid diseases. STAT3 mutations were identified in 1.6% (4 of 258) of patients with T-cell neoplasms, in 2.5% (2 of 79) of patients with diffuse large B-cell lymphoma but in no other B-cell lymphoma patients (0 of 104) or patients with myeloid malignancies (0 of 96). Functional in vitro assays indicated that the STAT3Y640F mutation leads to a constitutive phosphorylation of the protein. STA21, a STAT3 small molecule inhibitor, inhibited the proliferation of two distinct STAT3 mutated cell lines. Using a mouse bone marrow transplantation assay, we observed that STAT3Y640F expression leads to the development of myeloproliferative neoplasms with expansion of either myeloid cells or megakaryocytes. Together, these data indicate that the STAT3Y640F mutation leads to constitutive activation of STAT3, induces malignant hematopoiesis in vivo, and may represent a novel therapeutic target in some lymphoid malignancies.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Modelos Animales de Enfermedad , Neoplasias Hematológicas/genética , Mutación/genética , Trastornos Mieloproliferativos/genética , Factor de Transcripción STAT3/genética , Animales , Neoplasias Hematológicas/diagnóstico , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Trastornos Mieloproliferativos/diagnóstico
5.
N Engl J Med ; 360(22): 2289-301, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19474426

RESUMEN

BACKGROUND: The myelodysplastic syndromes and myeloproliferative disorders are associated with deregulated production of myeloid cells. The mechanisms underlying these disorders are not well defined. METHODS: We conducted a combination of molecular, cytogenetic, comparative-genomic-hybridization, and single-nucleotide-polymorphism analyses to identify a candidate tumor-suppressor gene common to patients with myelodysplastic syndromes, myeloproliferative disorders, and acute myeloid leukemia (AML). The coding sequence of this gene, TET2, was determined in 320 patients. We analyzed the consequences of deletions or mutations in TET2 with the use of in vitro clonal assays and transplantation of human tumor cells into mice. RESULTS: We initially identified deletions or mutations in TET2 in three patients with myelodysplastic syndromes, in three of five patients with myeloproliferative disorders, in two patients with primary AML, and in one patient with secondary AML. We selected the six patients with myelodysplastic syndromes or AML because they carried acquired rearrangements on chromosome 4q24; we selected the five patients with myeloproliferative disorders because they carried a dominant clone in hematopoietic progenitor cells that was positive for the V617F mutation in the Janus kinase 2 (JAK2) gene. TET2 defects were observed in 15 of 81 patients with myelodysplastic syndromes (19%), in 24 of 198 patients with myeloproliferative disorders (12%) (with or without the JAK2 V617F mutation), in 5 of 21 patients with secondary AML (24%), and in 2 of 9 patients with chronic myelomonocytic leukemia (22%). TET2 defects were present in hematopoietic stem cells and preceded the JAK2 V617F mutation in the five samples from patients with myeloproliferative disorders that we analyzed. CONCLUSIONS: Somatic mutations in TET2 occur in about 15% of patients with various myeloid cancers.


Asunto(s)
Proteínas de Unión al ADN/genética , Leucemia Mieloide Aguda/genética , Mutación , Síndromes Mielodisplásicos/genética , Trastornos Mieloproliferativos/genética , Proteínas Proto-Oncogénicas/genética , Secuencia de Aminoácidos , Animales , Antígenos CD34 , Cromosomas Humanos Par 4/genética , Hibridación Genómica Comparativa , Dioxigenasas , Reordenamiento Génico , Células Madre Hematopoyéticas/inmunología , Humanos , Janus Quinasa 2/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Eliminación de Secuencia
6.
Blood ; 114(15): 3285-91, 2009 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-19666869

RESUMEN

Oncogenic pathways underlying in the development of myelodysplastic syndromes (MDS) remain poorly characterized, but mutations of the ten-eleven translocation 2 (TET2) gene are frequently observed. In the present work, we evaluated the prognostic impact of TET2 mutations in MDS. Frameshift, nonsense, missense mutations, or defects in gene structure were identified in 22 (22.9%) of 96 patients (95% confidence interval [CI], 14.5-31.3 patients). Mutated and unmutated patients did not significantly differ in initial clinical or hematologic parameters. The 5-year OS was 76.9% (95% CI, 49.2%-91.3%) in mutated versus 18.3% (95% CI, 4.2%-41.1%) in unmutated patients (P = .005). The 3-year leukemia-free survival was 89.3% (95% CI, 63.1%-97.0%) in mutated versus 63.7% (95% CI, 48.2%-75.4%) in unmutated patients (P = .035). In univariate analysis (Cox proportional hazard model), the absence of TET2 mutation was associated with a 4.1-fold (95% CI, 1.4-12.0-fold) increased risk of death (P = .009). In multivariate analysis adjusted for age, International Prognostic Scoring System, and transfusion requirement, the presence of TET2 mutation remained an independent factor of favorable prognosis (hazard ratio, 5.2; 95% CI, 1.6-16.3; P = .005). These results indicate that TET2 mutations observed in approximately 20% of patients, irrespective of the World Health Organization or French-American-British subtype, represent a molecular marker for good prognosis in MDS.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/mortalidad , Proteínas Proto-Oncogénicas/genética , Anciano , Anciano de 80 o más Años , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Marcadores Genéticos , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas/metabolismo , Factores de Riesgo , Tasa de Supervivencia
7.
Blood ; 112(10): 4220-6, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18755984

RESUMEN

Oncogenic activation of tyrosine kinase signaling pathway is recurrent in human leukemia. To gain insight into the oncogenic process leading to acute megakaryoblastic leukemia (AMKL), we performed sequence analyses of a subset of oncogenes known to be activated in human myeloid and myeloproliferative disorders. In a series of human AMKL samples from both Down syndrome and non-Down syndrome patients, mutations were identified within KIT, FLT3, JAK2, JAK3, and MPL genes, with a higher frequency in DS than in non-DS patients. The novel mutations were analyzed using BaF3 cells, showing that JAK3 mutations were activating mutations. Finally, we report a novel constitutively active MPL mutant, MPLT487A, observed in a non-Down syndrome childhood AMKL that induces a myeloproliferative disease in mouse bone marrow transplantation assay.


Asunto(s)
Síndrome de Down/genética , Leucemia Megacarioblástica Aguda/genética , Mutación , Proteínas de Neoplasias/genética , Adulto , Anciano , Animales , Línea Celular Tumoral , Niño , Preescolar , Síndrome de Down/metabolismo , Femenino , Humanos , Lactante , Recién Nacido , Leucemia Megacarioblástica Aguda/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Trasplante de Neoplasias
8.
Blood Cancer J ; 10(3): 38, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32170099

RESUMEN

Aberrant NF-κB activation is a hallmark of most B-cell malignancies. Recurrent inactivating somatic mutations in the NFKBIE gene, which encodes IκBε, an inhibitor of NF-κB-inducible activity, are reported in several B-cell malignancies with highest frequencies in chronic lymphocytic leukemia and primary mediastinal B-cell lymphoma, and account for a fraction of NF-κB pathway activation. The impact of NFKBIE deficiency on B-cell development and function remains, however, largely unknown. Here, we show that Nfkbie-deficient mice exhibit an amplification of marginal zone B cells and an expansion of B1 B-cell subsets. In germinal center (GC)-dependent immune response, Nfkbie deficiency triggers expansion of GC B-cells through increasing cell proliferation in a B-cell autonomous manner. We also show that Nfkbie deficiency results in hyperproliferation of a B1 B-cell subset and leads to increased NF-κB activation in these cells upon Toll-like receptor stimulation. Nfkbie deficiency cooperates with mutant MYD88 signaling and enhances B-cell proliferation in vitro. In aged mice, Nfkbie absence drives the development of an oligoclonal indolent B-cell lymphoproliferative disorders, resembling monoclonal B-cell lymphocytosis. Collectively, these findings shed light on an essential role of IκBε in finely tuning B-cell development and function.


Asunto(s)
Proteínas I-kappa B/deficiencia , Leucemia Linfocítica Crónica de Células B/etiología , Proteínas Proto-Oncogénicas/deficiencia , Animales , Leucemia Linfocítica Crónica de Células B/genética , Ratones
10.
Cancer Discov ; 9(6): 796-811, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31018969

RESUMEN

The ETS-domain transcription factors divide into subfamilies based on protein similarities, DNA-binding sequences, and interaction with cofactors. They are regulated by extracellular clues and contribute to cellular processes, including proliferation and transformation. ETS genes are targeted through genomic rearrangements in oncogenesis. The PU.1/SPI1 gene is inactivated by point mutations in human myeloid malignancies. We identified a recurrent somatic mutation (Q226E) in PU.1/SPI1 in Waldenström macroglobulinemia, a B-cell lymphoproliferative disorder. It affects the DNA-binding affinity of the protein and allows the mutant protein to more frequently bind and activate promoter regions with respect to wild-type protein. Mutant SPI1 binding at promoters activates gene sets typically promoted by other ETS factors, resulting in enhanced proliferation and decreased terminal B-cell differentiation in model cell lines and primary samples. In summary, we describe oncogenic subversion of transcription factor function through subtle alteration of DNA binding leading to cellular proliferation and differentiation arrest. SIGNIFICANCE: The demonstration that a somatic point mutation tips the balance of genome-binding pattern provides a mechanistic paradigm for how missense mutations in transcription factor genes may be oncogenic in human tumors.This article is highlighted in the In This Issue feature, p. 681.


Asunto(s)
Regulación de la Expresión Génica , Mutación Missense , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Macroglobulinemia de Waldenström/genética , Macroglobulinemia de Waldenström/metabolismo , Animales , Azepinas/farmacología , Linfocitos B/citología , Linfocitos B/metabolismo , Secuencia de Bases , Sitios de Unión , Línea Celular , Proliferación Celular , Humanos , Lenalidomida/farmacología , Ratones , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Motivos de Nucleótidos , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Transactivadores/genética , Factores de Transcripción/metabolismo , Triazoles/farmacología , Macroglobulinemia de Waldenström/diagnóstico
11.
Haematologica ; 93(7): 1081-5, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18492690

RESUMEN

Homeobox containing transcription factors are frequently deregulated in human hematologic malignant diseases either indirectly through an abnormality of an upstream factor, or directly through rearrangement of the gene itself. Study of T-cell acute lymphoblastic leukemia identified the related non-clustered homeobox transcription factors, TLX1 and TLX3, as frequently ectopically expressed as a result of chromosomal translocations. We report the deregulation of a non-clustered homeobox gene in a new type of t(5;14)(q35;q11) translocation in a mature peripheral B-cell leukemia. This translocation results in the ectopic expression of the CSX1/NKX2-5 gene on chromosome 5q35 due to its juxtaposition to the TCR delta gene on chromosome 14q11. Expression of the CSX1/NKX2-5 protein conferred enhanced replating potential to transduced murine bone marrow cells. Our study establishes that deregulation of homeobox encoding genes is not restricted to acute leukemic proliferations, but is also observed in chronic malignant diseases.


Asunto(s)
Linfocitos B/metabolismo , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Factores de Transcripción/genética , Activación Transcripcional , Proliferación Celular , Enfermedad Crónica , Citogenética , Femenino , Proteína Homeótica Nkx-2.5 , Humanos , Persona de Mediana Edad , Modelos Biológicos , Mutación , Análisis de Secuencia de ADN , Translocación Genética
12.
Blood Adv ; 2(6): 703-714, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29581109

RESUMEN

The TET2 gene encodes an α-ketoglutarate-dependent dioxygenase able to oxidize 5-methylcytosine into 5-hydroxymethylcytosine, which is a step toward active DNA demethylation. TET2 is frequently mutated in myeloid malignancies but also in B- and T-cell malignancies. TET2 somatic mutations are also identified in healthy elderly individuals with clonal hematopoiesis. Tet2-deficient mouse models showed widespread hematological differentiation abnormalities, including myeloid, T-cell, and B-cell malignancies. We show here that, similar to what is observed with constitutive Tet2-deficient mice, B-cell-specific Tet2 knockout leads to abnormalities in the B1-cell subset and a development of B-cell malignancies after long latency. Aging Tet2-deficient mice accumulate clonal CD19+ B220low immunoglobulin M+ B-cell populations with transplantable ability showing similarities to human chronic lymphocytic leukemia, including CD5 expression and sensitivity to ibrutinib-mediated B-cell receptor (BCR) signaling inhibition. Exome sequencing of Tet2-/- malignant B cells reveals C-to-T and G-to-A mutations that lie within single-stranded DNA-specific activation-induced deaminase (AID)/APOBEC (apolipoprotein B messenger RNA editing enzyme, catalytic polypeptide-like) cytidine deaminases targeted motif, as confirmed by the lack of a B-cell tumor in compound Tet2-Aicda-deficient mice. Finally, we show that Tet2 deficiency accelerates and exacerbates T-cell leukemia/lymphoma 1A-induced leukemogenesis. Together, our data establish that Tet2 deficiency predisposes to mature B-cell malignancies, which development might be attributed in part to AID-mediated accumulating mutations and BCR-mediated signaling.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Leucemia de Células B/genética , Linfoma de Células B/genética , Proteínas Proto-Oncogénicas/deficiencia , Alelos , Animales , Linfocitos B , Biomarcadores , Supervivencia Celular , Dioxigenasas , Citometría de Flujo , Genotipo , Leucemia de Células B/metabolismo , Leucemia de Células B/patología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Mutación , Receptores de Antígenos de Linfocitos B/metabolismo
13.
Cancer Discov ; 8(12): 1632-1653, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30274972

RESUMEN

TET2 somatic mutations occur in ∼10% of diffuse large B-cell lymphomas (DLBCL) but are of unknown significance. Herein, we show that TET2 is required for the humoral immune response and is a DLBCL tumor suppressor. TET2 loss of function disrupts transit of B cells through germinal centers (GC), causing GC hyperplasia, impaired class switch recombination, blockade of plasma cell differentiation, and a preneoplastic phenotype. TET2 loss was linked to focal loss of enhancer hydroxymethylation and transcriptional repression of genes that mediate GC exit, such as PRDM1. Notably, these enhancers and genes are also repressed in CREBBP-mutant DLBCLs. Accordingly, TET2 mutation in patients yields a CREBBP-mutant gene-expression signature, CREBBP and TET2 mutations are generally mutually exclusive, and hydroxymethylation loss caused by TET2 deficiency impairs enhancer H3K27 acetylation. Hence, TET2 plays a critical role in the GC reaction, and its loss of function results in lymphomagenesis through failure to activate genes linked to GC exit signals. SIGNIFICANCE: We show that TET2 is required for exit of the GC, B-cell differentiation, and is a tumor suppressor for mature B cells. Loss of TET2 phenocopies CREBBP somatic mutation. These results advocate for sequencing TET2 in patients with lymphoma and for the testing of epigenetic therapies to treat these tumors.See related commentary by Shingleton and Dave, p. 1515.This article is highlighted in the In This Issue feature, p. 1494.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Centro Germinal/metabolismo , Linfoma de Células B Grandes Difuso/genética , Células Plasmáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Animales , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Epigénesis Genética/genética , Perfilación de la Expresión Génica/métodos , Centro Germinal/patología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hiperplasia , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Ratones Noqueados , Ratones Transgénicos , Mutación , Células Plasmáticas/patología , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
14.
Oncotarget ; 9(39): 25630-25646, 2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29876013

RESUMEN

Epigenetic alterations have been associated with both pathogenesis and progression of cancer. By screening of library compounds, we identified a novel hybrid epi-drug MC2884, a HAT/EZH2 inhibitor, able to induce bona fide cancer-selective cell death in both solid and hematological cancers in vitro, ex vivo and in vivo xenograft models. Anticancer action was due to an epigenome modulation by H3K27me3, H3K27ac, H3K9/14ac decrease, and to caspase-dependent apoptosis induction. MC2884 triggered mitochondrial pathway apoptosis by up-regulation of cleaved-BID, and strong down-regulation of BCL2. Even aggressive models of cancer, such as p53-/- or TET2-/- cells, responded to MC2884, suggesting MC2884 therapeutic potential also for the therapy of TP53 or TET2-deficient human cancers. MC2884 induced massive apoptosis in ex vivo human primary leukemia blasts with poor prognosis in vivo, by targeting BCL2 expression. MC2884-treatment reduced acetylation of the BCL2 promoter at higher level than combined p300 and EZH2 inhibition. This suggests a key role for BCL-2 reduction in potentiating responsiveness, also in combination therapy with BCL2 inhibitors. Finally, we identified both the mechanism of MC2884 action as well as a potential therapeutic scheme of its use. Altogether, this provides proof of concept for the use of epi-drugs coupled with epigenome analyses to 'personalize' precision medicine.

15.
DNA Repair (Amst) ; 43: 78-88, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27289557

RESUMEN

The family of Ten-Eleven Translocation (TET) proteins is implicated in the process of active DNA demethylation and thus in epigenetic regulation. TET 1, 2 and 3 proteins are oxygenases that can hydroxylate 5-methylcytosine (5-mC) into 5-hydroxymethylcytosine (5-hmC) and further oxidize 5-hmC into 5-formylcytosine (5-fC) and 5-carboxylcytosine (5-caC). The base excision repair (BER) pathway removes the resulting 5-fC and 5-caC bases paired with a guanine and replaces them with regular cytosine. The question arises whether active modification of 5-mC residues and their subsequent elimination could affect the genomic DNA stability. Here, we generated two inducible cell lines (Ba/F3-EPOR, and UT7) overexpressing wild-type or catalytically inactive human TET2 proteins. Wild-type TET2 induction resulted in an increased level of 5-hmC and a cell cycle defect in S phase associated with higher level of phosphorylated P53, chromosomal and centrosomal abnormalities. Furthermore, in a thymine-DNA glycosylase (Tdg) deficient context, the TET2-mediated increase of 5-hmC induces mutagenesis characterized by GC>AT transitions in CpG context suggesting a mutagenic potential of 5-hmC metabolites. Altogether, these data suggest that TET2 activity and the levels of 5-hmC and its derivatives should be tightly controlled to avoid genetic and chromosomal instabilities. Moreover, TET2-mediated active demethylation might be a very dangerous process if used to entirely demethylate the genome and might rather be used only at specific loci.


Asunto(s)
5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Mutagénesis , Proteínas Proto-Oncogénicas/genética , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Secuencia de Bases , Línea Celular , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Epigénesis Genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Hidroxilación , Células Progenitoras de Megacariocitos/citología , Células Progenitoras de Megacariocitos/metabolismo , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Fase S , Timina ADN Glicosilasa/deficiencia , Timina ADN Glicosilasa/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
16.
Oncogene ; 21(17): 2652-61, 2002 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-11965538

RESUMEN

The p16(INK4A-ARF) locus plays a crucial role in the control of cellular proliferation via both the Rb and P53 pathways. We previously demonstrated that this locus is altered in human skin carcinomas. In the present study we have studied the expression of the p16(INK4A-ARF) locus following UVB irradiation of normal human keratinocytes both at the mRNA (RT-PCR) and at the protein (Western blotting) levels. Our data confirmed that P16(INK4A) protein is induced by UVB at low (30 mJ cm(2)) and high (100 mJ cm(2)) doses and is observed after a single or repeated exposure implying that this response is involved in both the immediate and adaptative response to UVB. The apparent absence of induction p16(INK4A) mRNA suggested that P16(INK4A) protein is upregulated at the post-transcriptional level. Analysis by flow cytometry and BrdU staining indicated that the highest protein level of P16(INK4A) in the cells was associated with a G(2) cell cycle arrest. Comparative analysis of P16(INK4A) and P53 showed that they were differentially modulated in keratinocytes according to the UVB dose and regimen. Low, acute or repeated UVB exposures led to accumulation of both P16(INK4A) and p53, whereas at high UVB doses, P53 and P53-dependent genes were not induced or even downregulated and only a slight but reproducible stabilization of P16(INK4A) protein was observed. In our conditions, P14(ARF) did not seem to participate in the UV response in these cells as P14(ARF) protein did not vary. These results infer that P16(INK4A) plays a role in cell cycle regulation of keratinocytes submitted to UVB irradiation. They also reinforce our previous demonstration of the importance of inactivation of this gene in UV-induced skin carcinogenesis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación de la Expresión Génica/efectos de la radiación , Queratinocitos/efectos de la radiación , Proteínas Nucleares , Proteínas Proto-Oncogénicas c-bcl-2 , Western Blotting , Bromodesoxiuridina , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Ciclinas/metabolismo , Cartilla de ADN/química , Relación Dosis-Respuesta en la Radiación , Citometría de Flujo , Humanos , Immunoblotting , Queratinocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , ARN Mensajero/metabolismo , Dosis de Radiación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Rayos Ultravioleta , Regulación hacia Arriba , Proteína X Asociada a bcl-2
17.
Cancer Discov ; 4(9): 1088-101, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24920063

RESUMEN

UNLABELLED: Appropriate cancer care requires a thorough understanding of the natural history of the disease, including the cell of origin, the pattern of clonal evolution, and the functional consequences of the mutations. Using deep sequencing of flow-sorted cell populations from patients with chronic lymphocytic leukemia (CLL), we established the presence of acquired mutations in multipotent hematopoietic progenitors. Mutations affected known lymphoid oncogenes, including BRAF, NOTCH1, and SF3B1. NFKBIE and EGR2 mutations were observed at unexpectedly high frequencies, 10.7% and 8.3% of 168 advanced-stage patients, respectively. EGR2 mutations were associated with a shorter time to treatment and poor overall survival. Analyses of BRAF and EGR2 mutations suggest that they result in deregulation of B-cell receptor (BCR) intracellular signaling. Our data propose disruption of hematopoietic and early B-cell differentiation through the deregulation of pre-BCR signaling as a phenotypic outcome of CLL mutations and show that CLL develops from a pre-leukemic phase. SIGNIFICANCE: The origin and pathogenic mechanisms of CLL are not fully understood. The current work indicates that CLL develops from pre-leukemic multipotent hematopoietic progenitors carrying somatic mutations. It advocates for abnormalities in early B-cell differentiation as a phenotypic convergence of the diverse acquired mutations observed in CLL.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Mutación , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/patología , Fosfoproteínas/genética , Factores de Empalme de ARN , Receptores de Antígenos de Linfocitos B/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/genética , Transducción de Señal
18.
Cancer Cell ; 20(1): 25-38, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21723201

RESUMEN

Loss-of-function mutations affecting one or both copies of the Ten-Eleven-translocation (TET)2 gene have been described in various human myeloid malignancies. We report that inactivation of Tet2 in mouse perturbs both early and late steps of hematopoiesis including myeloid and lymphoid differentiation in a cell-autonomous manner, endows the cells with competitive advantage, and eventually leads to the development of malignancies. We subsequently observed TET2 mutations in human lymphoid disorders. TET2 mutations could be detected in immature progenitors endowed with myeloid colony-forming potential. Our results show that the mutations present in lymphoid tumor cells may occur at both early and later steps of lymphoid development and indicate that impairment of TET2 function or/and expression predisposes to the development of hematological malignancies.


Asunto(s)
Proteínas de Unión al ADN/genética , Silenciador del Gen , Hematopoyesis , Linfoma/patología , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas/genética , Animales , Antígenos CD34/metabolismo , Linaje de la Célula , Dioxigenasas , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Homeostasis , Humanos , Linfoma/metabolismo , Ratones , Modelos Animales , Mutación/genética , Células Mieloides/metabolismo , Células Mieloides/patología , Lesiones Precancerosas/metabolismo
19.
Blood ; 108(10): 3560-3, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16873674

RESUMEN

Subtle variation in the expression or function of a small group of transcription factors can drive leukemogenesis. The CEBPA protein is known to regulate the balance between cell proliferation and differentiation during early hematopoietic development and myeloid differentiation. In human myeloid leukemia, CEBPA is frequently inactivated by mutation and indirect and posttranslational mechanisms, in keeping with tumor suppressor properties. We report that CEBPA is activated by juxtaposition to the immunoglobulin gene enhancer upon its rearrangement with the immunoglobulin heavy-chain locus in precursor B-cell acute lymphoblastic leukemia harboring t(14;19)(q32;q13). Overexpression of apparently normal CEBPA RNA or protein was observed in 6 patients. These data indicate that CEBPA may exhibit oncogenic as well as tumor suppressor properties in human leukemogenesis.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Translocación Genética , Adulto , Anciano , Niño , Cromosomas Humanos Par 14 , Cromosomas Humanos Par 19 , Femenino , Reordenamiento Génico , Genes de Inmunoglobulinas , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras B/etiología , ARN Neoplásico/análisis
20.
Blood ; 108(13): 4198-201, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16926283

RESUMEN

The t(5;14)(q35;q32) chromosomal translocation is specifically observed in up to 20% of childhood T-cell acute lymphoblastic leukemia (T-ALL). It affects the BCL11B/CTIP2 locus on chromosome 14 and the RANBP17-TLX3/HOX11L2 region on chromosome 5. It leads to ectopic activation of TLX3/HOX11L2. To investigate the reasons of the association between t(5;14) and T-ALL, we isolated the translocation breakpoints in 8 t(5;14) patients. Sequence analyses did not involve recombinase activity in the genesis of the translocation. We used DNAse1 hypersensitive experiments to locate transcriptional regulatory elements downstream of BCL11B. By transient transfection experiments, 2 of the 6 regions demonstrated cis-activation properties in T cells and were also effective on the TLX3 promoter. Our data indicate that the basis of the specific association between t(5;14) and T-ALL lies on the juxtaposition of TLX3 to long-range cis-activating regions active during T-cell differentiation.


Asunto(s)
Cromosomas Humanos Par 14/genética , Cromosomas Humanos Par 5/genética , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Leucemia-Linfoma de Células T del Adulto/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Oncogénicas/genética , Proteínas Represoras/genética , Translocación Genética , Proteínas Supresoras de Tumor/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Homeodominio/biosíntesis , Humanos , Células Jurkat , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/patología , Proteínas Oncogénicas/biosíntesis , Proteínas de Fusión Oncogénica/biosíntesis , Regiones Promotoras Genéticas/genética , Proteínas Represoras/biosíntesis , Linfocitos T/metabolismo , Linfocitos T/patología , Transcripción Genética , Proteínas Supresoras de Tumor/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA