Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(11): e2218330120, 2023 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-36893259

RESUMEN

Heterozygous inactivating mutations of the KMT2D methyltransferase and the CREBBP acetyltransferase are among the most common genetic alterations in B cell lymphoma and co-occur in 40 to 60% of follicular lymphoma (FL) and 30% of EZB/C3 diffuse large B cell lymphoma (DLBCL) cases, suggesting they may be coselected. Here, we show that combined germinal center (GC)-specific haploinsufficiency of Crebbp and Kmt2d synergizes in vivo to promote the expansion of abnormally polarized GCs, a common preneoplastic event. These enzymes form a biochemical complex on select enhancers/superenhancers that are critical for the delivery of immune signals in the GC light zone and are only corrupted upon dual Crebbp/Kmt2d loss, both in mouse GC B cells and in human DLBCL. Moreover, CREBBP directly acetylates KMT2D in GC-derived B cells, and, consistently, its inactivation by FL/DLBCL-associated mutations abrogates its ability to catalyze KMT2D acetylation. Genetic and pharmacologic loss of CREBBP and the consequent decrease in KMT2D acetylation lead to reduced levels of H3K4me1, supporting a role for this posttranslational modification in modulating KMT2D activity. Our data identify a direct biochemical and functional interaction between CREBBP and KMT2D in the GC, with implications for their role as tumor suppressors in FL/DLBCL and for the development of precision medicine approaches targeting enhancer defects induced by their combined loss.


Asunto(s)
Linfoma Folicular , Linfoma de Células B Grandes Difuso , Animales , Humanos , Ratones , Acetilación , Linfocitos B/metabolismo , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Centro Germinal , Linfoma Folicular/genética , Linfoma Folicular/metabolismo , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/patología , Mutación , Procesamiento Proteico-Postraduccional
3.
Nature ; 607(7920): 808-815, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35794478

RESUMEN

Diffuse large B cell lymphoma (DLBCL) is the most common B cell non-Hodgkin lymphoma and remains incurable in around 40% of patients. Efforts to sequence the coding genome identified several genes and pathways that are altered in this disease, including potential therapeutic targets1-5. However, the non-coding genome of DLBCL remains largely unexplored. Here we show that active super-enhancers are highly and specifically hypermutated in 92% of samples from individuals with DLBCL, display signatures of activation-induced cytidine deaminase activity, and are linked to genes that encode B cell developmental regulators and oncogenes. As evidence of oncogenic relevance, we show that the hypermutated super-enhancers linked to the BCL6, BCL2 and CXCR4 proto-oncogenes prevent the binding and transcriptional downregulation of the corresponding target gene by transcriptional repressors, including BLIMP1 (targeting BCL6) and the steroid receptor NR3C1 (targeting BCL2 and CXCR4). Genetic correction of selected mutations restored repressor DNA binding, downregulated target gene expression and led to the counter-selection of cells containing corrected alleles, indicating an oncogenic dependency on the super-enhancer mutations. This pervasive super-enhancer mutational mechanism reveals a major set of genetic lesions deregulating gene expression, which expands the involvement of known oncogenes in DLBCL pathogenesis and identifies new deregulated gene targets of therapeutic relevance.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Linfoma de Células B Grandes Difuso , Mutación , Oncogenes , Regulación hacia Abajo , Elementos de Facilitación Genéticos/genética , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Oncogenes/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-6/genética , Receptores CXCR4/genética , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/metabolismo
4.
Immunity ; 51(3): 535-547.e9, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31519498

RESUMEN

Inactivating mutations of the CREBBP and EP300 acetyltransferases are among the most common genetic alterations in diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL). Here, we examined the relationship between these two enzymes in germinal center (GC) B cells, the normal counterpart of FL and DLBCL, and in lymphomagenesis by using conditional GC-directed deletion mouse models targeting Crebbp or Ep300. We found that CREBBP and EP300 modulate common as well as distinct transcriptional programs implicated in separate anatomic and functional GC compartments. Consistently, deletion of Ep300 but not Crebbp impaired the fitness of GC B cells in vivo. Combined loss of Crebbp and Ep300 completely abrogated GC formation, suggesting that these proteins partially compensate for each other through common transcriptional targets. This synthetic lethal interaction was retained in CREBBP-mutant DLBCL cells and could be pharmacologically targeted with selective small molecule inhibitors of CREBBP and EP300 function. These data provide proof-of-principle for the clinical development of EP300-specific inhibitors in FL and DLBCL.


Asunto(s)
Linfocitos B/fisiología , Proteína de Unión a CREB/genética , Proteína p300 Asociada a E1A/genética , Epigénesis Genética/genética , Centro Germinal/fisiología , Linfoma Folicular/etiología , Linfoma de Células B Grandes Difuso/genética , Acetiltransferasas/genética , Animales , Línea Celular , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Eliminación de Secuencia/genética , Transcripción Genética/genética
5.
Blood ; 131(22): 2454-2465, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29650799

RESUMEN

Dissecting the pathogenesis of classical Hodgkin lymphoma (cHL), a common cancer in young adults, remains challenging because of the rarity of tumor cells in involved tissues (usually <5%). Here, we analyzed the coding genome of cHL by microdissecting tumor and normal cells from 34 patient biopsies for a total of ∼50 000 singly isolated lymphoma cells. We uncovered several recurrently mutated genes, namely, STAT6 (32% of cases), GNA13 (24%), XPO1 (18%), and ITPKB (16%), and document the functional role of mutant STAT6 in sustaining tumor cell viability. Mutations of STAT6 genetically and functionally cooperated with disruption of SOCS1, a JAK-STAT pathway inhibitor, to promote cHL growth. Overall, 87% of cases showed dysregulation of the JAK-STAT pathway by genetic alterations in multiple genes (also including STAT3, STAT5B, JAK1, JAK2, and PTPN1), attesting to the pivotal role of this pathway in cHL pathogenesis and highlighting its potential as a new therapeutic target in this disease.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/genética , Quinasas Janus/genética , Mutación , Factores de Transcripción STAT/genética , Línea Celular Tumoral , Análisis Mutacional de ADN , Enfermedad de Hodgkin/metabolismo , Enfermedad de Hodgkin/patología , Humanos , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
6.
Cancer Discov ; 7(3): 322-337, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28069569

RESUMEN

Inactivating mutations of the CREBBP acetyltransferase are highly frequent in diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL), the two most common germinal center (GC)-derived cancers. However, the role of CREBBP inactivation in lymphomagenesis remains unclear. Here, we show that CREBBP regulates enhancer/super-enhancer networks with central roles in GC/post-GC cell fate decisions, including genes involved in signal transduction by the B-cell receptor and CD40 receptor, transcriptional control of GC and plasma cell development, and antigen presentation. Consistently, Crebbp-deficient B cells exhibit enhanced response to mitogenic stimuli and perturbed plasma cell differentiation. Although GC-specific loss of Crebbp was insufficient to initiate malignant transformation, compound Crebbp-haploinsufficient/BCL2-transgenic mice, mimicking the genetics of FL and DLBCL, develop clonal lymphomas recapitulating the features of the human diseases. These findings establish CREBBP as a haploinsufficient tumor-suppressor gene in GC B cells and provide insights into the mechanisms by which its loss contributes to lymphomagenesis.Significance: Loss-of-function mutations of CREBBP are common and early lesions in FL and DLBCL, suggesting a prominent role in lymphoma initiation. Our studies identify the cellular program by which reduced CREBBP dosage facilitates malignant transformation, and have direct implications for targeted lymphoma therapy based on drugs affecting CREBBP-mediated chromatin acetylation. Cancer Discov; 7(3); 322-37. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 235.


Asunto(s)
Linfocitos B/patología , Proteína de Unión a CREB/genética , Genes Supresores de Tumor , Linfoma de Células B Grandes Difuso/genética , Animales , Linfocitos B/metabolismo , Proteína de Unión a CREB/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Centro Germinal/patología , Haploinsuficiencia , Humanos , Linfoma Folicular/genética , Linfoma Folicular/patología , Linfoma de Células B Grandes Difuso/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/patología , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo
7.
Nat Med ; 21(10): 1190-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26366712

RESUMEN

Mutations in the gene encoding the KMT2D (or MLL2) methyltransferase are highly recurrent and occur early during tumorigenesis in diffuse large B cell lymphoma (DLBCL) and follicular lymphoma (FL). However, the functional consequences of these mutations and their role in lymphomagenesis are unknown. Here we show that FL- and DLBCL-associated KMT2D mutations impair KMT2D enzymatic activity, leading to diminished global H3K4 methylation in germinal-center (GC) B cells and DLBCL cells. Conditional deletion of Kmt2d early during B cell development, but not after initiation of the GC reaction, results in an increase in GC B cells and enhances B cell proliferation in mice. Moreover, genetic ablation of Kmt2d in mice overexpressing Bcl2 increases the incidence of GC-derived lymphomas resembling human tumors. These findings suggest that KMT2D acts as a tumor suppressor gene whose early loss facilitates lymphomagenesis by remodeling the epigenetic landscape of the cancer precursor cells. Eradication of KMT2D-deficient cells may thus represent a rational therapeutic approach for targeting early tumorigenic events.


Asunto(s)
Proteínas de Unión al ADN/genética , Centro Germinal/citología , Linfoma de Células B Grandes Difuso/genética , Proteínas de Neoplasias/genética , Animales , Linfocitos B/patología , Proliferación Celular , Metilación de ADN , Epigénesis Genética , Silenciador del Gen , Humanos , Linfoma de Células B Grandes Difuso/etiología , Ratones , Mutación Missense , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA