Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 135(11): 845-856, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-31932841

RESUMEN

Mutations in the epigenetic regulators DNMT3A and IDH1/2 co-occur in patients with acute myeloid leukemia and lymphoma. In this study, these 2 epigenetic mutations cooperated to induce leukemia. Leukemia-initiating cells from Dnmt3a-/- mice that express an IDH2 neomorphic mutant have a megakaryocyte-erythroid progenitor-like immunophenotype, activate a stem-cell-like gene signature, and repress differentiated progenitor genes. We observed an epigenomic dysregulation with the gain of repressive H3K9 trimethylation and loss of H3K9 acetylation in diseased mouse bone marrow hematopoietic stem and progenitor cells (HSPCs). HDAC inhibitors rapidly reversed the H3K9 methylation/acetylation imbalance in diseased mouse HSPCs while reducing the leukemia burden. In addition, using targeted metabolomic profiling for the first time in mouse leukemia models, we also showed that prostaglandin E2 is overproduced in double-mutant HSPCs, rendering them sensitive to prostaglandin synthesis inhibition. These data revealed that Dnmt3a and Idh2 mutations are synergistic events in leukemogenesis and that HSPCs carrying both mutations are sensitive to induced differentiation by the inhibition of both prostaglandin synthesis and HDAC, which may reveal new therapeutic opportunities for patients carrying IDH1/2 mutations.


Asunto(s)
Transformación Celular Neoplásica/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Neoplasias Hematológicas/genética , Hematopoyesis/genética , Isocitrato Deshidrogenasa/genética , Mutación , Animales , Línea Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Secuenciación de Inmunoprecipitación de Cromatina , Metilación de ADN , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Estudios de Asociación Genética/métodos , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Humanos , Metaboloma , Metabolómica/métodos , Ratones , Ratones Noqueados
2.
Haematologica ; 107(4): 887-898, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34092059

RESUMEN

Tatton-Brown-Rahman syndrome (TBRS) is an overgrowth disorder caused by germline heterozygous mutations in the DNA methyltransferase DNMT3A. DNMT3A is a critical regulator of hematopoietic stem cell (HSC) differentiation and somatic DNMT3A mutations are frequent in hematologic malignancies and clonal hematopoiesis. Yet, the impact of constitutive DNMT3A mutation on hematopoiesis in TBRS is undefined. In order to establish how constitutive mutation of DNMT3A impacts blood development in TBRS we gathered clinical data and analyzed blood parameters in 18 individuals with TBRS. We also determined the distribution of major peripheral blood cell lineages by flow cytometric analyses. Our analyses revealed non-anemic macrocytosis, a relative decrease in lymphocytes and increase in neutrophils in TBRS individuals compared to unaffected controls. We were able to recapitulate these hematologic phenotypes in multiple murine models of TBRS and identified rare hematological and non-hematological malignancies associated with constitutive Dnmt3a mutation. We further show that loss of DNMT3A in TBRS is associated with an altered DNA methylation landscape in hematopoietic cells affecting regions critical to stem cell function and tumorigenesis. Overall, our data identify key hematopoietic effects driven by DNMT3A mutation with clinical implications for individuals with TBRS and DNMT3A-associated clonal hematopoiesis or malignancies.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , Discapacidad Intelectual , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Células Germinativas/patología , Hematopoyesis/genética , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Ratones
3.
bioRxiv ; 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-37693622

RESUMEN

The DNA damage response is critical for maintaining genome integrity and is commonly disrupted in the development of cancer. PPM1D (protein phosphatase, Mg2+/Mn2+ dependent 1D) is a master negative regulator of the response; gain-of-function mutations and amplifications of PPM1D are found across several human cancers making it a relevant pharmacologic target. Here, we used CRISPR/Cas9 screening to identify synthetic-lethal dependencies of PPM1D, uncovering superoxide dismutase-1 (SOD1) as a potential target for PPM1D-mutant cells. We revealed a dysregulated redox landscape characterized by elevated levels of reactive oxygen species and a compromised response to oxidative stress in PPM1D-mutant cells. Altogether, our results demonstrate the protective role of SOD1 against oxidative stress in PPM1D-mutant leukemia cells and highlight a new potential therapeutic strategy against PPM1D-mutant cancers.

4.
Elife ; 122024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38896450

RESUMEN

The DNA damage response is critical for maintaining genome integrity and is commonly disrupted in the development of cancer. PPM1D (protein phosphatase Mg2+/Mn2+-dependent 1D) is a master negative regulator of the response; gain-of-function mutations and amplifications of PPM1D are found across several human cancers making it a relevant pharmacological target. Here, we used CRISPR/Cas9 screening to identify synthetic-lethal dependencies of PPM1D, uncovering superoxide dismutase-1 (SOD1) as a potential target for PPM1D-mutant cells. We revealed a dysregulated redox landscape characterized by elevated levels of reactive oxygen species and a compromised response to oxidative stress in PPM1D-mutant cells. Altogether, our results demonstrate a role for SOD1 in the survival of PPM1D-mutant leukemia cells and highlight a new potential therapeutic strategy against PPM1D-mutant cancers.


Asunto(s)
Proteína Fosfatasa 2C , Superóxido Dismutasa-1 , Proteína Fosfatasa 2C/metabolismo , Proteína Fosfatasa 2C/genética , Humanos , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Línea Celular Tumoral , Leucemia/genética , Sistemas CRISPR-Cas , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Mutaciones Letales Sintéticas , Mutación
5.
Mol Metab ; 86: 101969, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908793

RESUMEN

OBJECTIVES: Cachexia is a metabolic disorder and comorbidity with cancer and heart failure. The syndrome impacts more than thirty million people worldwide, accounting for 20% of all cancer deaths. In acute myeloid leukemia, somatic mutations of the metabolic enzyme isocitrate dehydrogenase 1 and 2 cause the production of the oncometabolite D2-hydroxyglutarate (D2-HG). Increased production of D2-HG is associated with heart and skeletal muscle atrophy, but the mechanistic links between metabolic and proteomic remodeling remain poorly understood. Therefore, we assessed how oncometabolic stress by D2-HG activates autophagy and drives skeletal muscle loss. METHODS: We quantified genomic, metabolomic, and proteomic changes in cultured skeletal muscle cells and mouse models of IDH-mutant leukemia using RNA sequencing, mass spectrometry, and computational modeling. RESULTS: D2-HG impairs NADH redox homeostasis in myotubes. Increased NAD+ levels drive activation of nuclear deacetylase Sirt1, which causes deacetylation and activation of LC3, a key regulator of autophagy. Using LC3 mutants, we confirm that deacetylation of LC3 by Sirt1 shifts its distribution from the nucleus into the cytosol, where it can undergo lipidation at pre-autophagic membranes. Sirt1 silencing or p300 overexpression attenuated autophagy activation in myotubes. In vivo, we identified increased muscle atrophy and reduced grip strength in response to D2-HG in male vs. female mice. In male mice, glycolytic intermediates accumulated, and protein expression of oxidative phosphorylation machinery was reduced. In contrast, female animals upregulated the same proteins, attenuating the phenotype in vivo. Network modeling and machine learning algorithms allowed us to identify candidate proteins essential for regulating oncometabolic adaptation in mouse skeletal muscle. CONCLUSIONS: Our multi-omics approach exposes new metabolic vulnerabilities in response to D2-HG in skeletal muscle and provides a conceptual framework for identifying therapeutic targets in cachexia.


Asunto(s)
Autofagia , Glutaratos , Músculo Esquelético , Transducción de Señal , Animales , Ratones , Músculo Esquelético/metabolismo , Masculino , Glutaratos/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Isocitrato Deshidrogenasa/genética , Caquexia/metabolismo , Femenino , Sirtuina 1/metabolismo , Sirtuina 1/genética , Ratones Endogámicos C57BL
6.
iScience ; 27(3): 109122, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38414863

RESUMEN

During aging, blood cell production becomes dominated by a limited number of variant hematopoietic stem cell (HSC) clones. Differentiated progeny of variant HSCs are thought to mediate the detrimental effects of such clonal hematopoiesis on organismal health, but the mechanisms are poorly understood. While somatic mutations in DNA methyltransferase 3A (DNMT3A) frequently drive clonal dominance, the aging milieu also likely contributes. Here, we examined in mice the interaction between high-fat diet (HFD) and reduced DNMT3A in hematopoietic cells; strikingly, this combination led to weight gain. HFD amplified pro-inflammatory pathways and upregulated inflammation-associated genes in mutant cells along a pro-myeloid trajectory. Aberrant DNA methylation during myeloid differentiation and in response to HFD led to pro-inflammatory activation and maintenance of stemness genes. These findings suggest that reduced DNMT3A in hematopoietic cells contributes to weight gain, inflammation, and metabolic dysfunction, highlighting a role for DNMT3A loss in the development of metabolic disorders.

8.
Nat Aging ; 3(11): 1387-1400, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37884767

RESUMEN

DNA methylation deregulation at partially methylated domains (PMDs) represents an epigenetic signature of aging and cancer, yet the underlying molecular basis and resulting biological consequences remain unresolved. We report herein a mechanistic link between disrupted DNA methylation at PMDs and the spatial relocalization of H3K9me3-marked heterochromatin in aged hematopoietic stem and progenitor cells (HSPCs) or those with impaired DNA methylation. We uncover that TET2 modulates the spatial redistribution of H3K9me3-marked heterochromatin to mediate the upregulation of endogenous retroviruses (ERVs) and interferon-stimulated genes (ISGs), hence contributing to functional decline of aged HSPCs. TET2-deficient HSPCs retain perinuclear distribution of heterochromatin and exhibit age-related clonal expansion. Reverse transcriptase inhibitors suppress ERVs and ISGs expression, thereby restoring age-related defects in aged HSPCs. Collectively, our findings deepen the understanding of the functional interplay between DNA methylation and histone modifications, which is vital for maintaining heterochromatin function and safeguarding genome stability in stem cells.


Asunto(s)
Células Madre Hematopoyéticas , Heterocromatina , Heterocromatina/genética , Células Madre Hematopoyéticas/metabolismo , Metilación de ADN/genética
9.
Nat Cell Biol ; 25(4): 528-539, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37024683

RESUMEN

Upon stimulation by extrinsic stimuli, stem cells initiate a programme that enables differentiation or self-renewal. Disruption of the stem state exit has catastrophic consequences for embryogenesis and can lead to cancer. While some elements of this stem state switch are known, major regulatory mechanisms remain unclear. Here we show that this switch involves a global increase in splicing efficiency coordinated by DNA methyltransferase 3α (DNMT3A), an enzyme typically involved in DNA methylation. Proper activation of murine and human embryonic and haematopoietic stem cells depends on messenger RNA processing, influenced by DNMT3A in response to stimuli. DNMT3A coordinates splicing through recruitment of the core spliceosome protein SF3B1 to RNA polymerase and mRNA. Importantly, the DNA methylation function of DNMT3A is not required and loss of DNMT3A leads to impaired splicing during stem cell turnover. Finally, we identify the spliceosome as a potential therapeutic target in DNMT3A-mutated leukaemias. Together, our results reveal a modality through which DNMT3A and the spliceosome govern exit from the stem state towards differentiation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Animales , Humanos , Ratones , Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Células Madre Hematopoyéticas/metabolismo
10.
Nat Genet ; 54(5): 625-636, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35534561

RESUMEN

DNA methyltransferase 3a (DNMT3A) plays a crucial role during mammalian development. Two isoforms of DNMT3A are differentially expressed from stem cells to somatic tissues, but their individual functions remain largely uncharacterized. Here we report that the long isoform DNMT3A1, but not the short DNMT3A2, is essential for mouse postnatal development. DNMT3A1 binds to and regulates bivalent neurodevelopmental genes in the brain. Strikingly, Dnmt3a1 knockout perinatal lethality could be partially rescued by DNMT3A1 restoration in the nervous system. We further show that the intrinsically disordered N terminus of DNMT3A1 is required for normal development and DNA methylation at DNMT3A1-enriched regions. Mechanistically, a ubiquitin-interacting motif embedded in a putative α-helix within the N terminus binds to mono-ubiquitinated histone H2AK119, probably mediating recruitment of DNMT3A1 to Polycomb-regulated regions. These data demonstrate an isoform-specific role for DNMT3A1 in mouse postnatal development and reveal the N terminus as a necessary regulatory domain for DNMT3A1 chromatin occupancy and functions in the nervous system.


Asunto(s)
Metilasas de Modificación del ADN , Histonas , Animales , Metilación de ADN , Metilasas de Modificación del ADN/metabolismo , Histonas/metabolismo , Ratones , Isoformas de Proteínas
11.
Elife ; 112022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-35635747

RESUMEN

DNA Methyltransferase 3 A (DNMT3A) is an important facilitator of differentiation of both embryonic and hematopoietic stem cells. Heterozygous germline mutations in DNMT3A lead to Tatton-Brown-Rahman Syndrome (TBRS), characterized by obesity and excessive height. While DNMT3A is known to impact feeding behavior via the hypothalamus, here we investigated a role in adipocyte progenitors utilizing heterozygous knockout mice that recapitulate cardinal TBRS phenotypes. These mice become morbidly obese due to adipocyte enlargement and tissue expansion. Adipose tissue in these mice exhibited defects in preadipocyte maturation and precocious activation of inflammatory gene networks, including interleukin-6 signaling. Adipocyte progenitor cell lines lacking DNMT3A exhibited aberrant differentiation. Furthermore, mice in which Dnmt3a was specifically ablated in adipocyte progenitors showed enlarged fat depots and increased progenitor numbers, partly recapitulating the TBRS obesity phenotypes. Loss of DNMT3A led to constitutive DNA hypomethylation, such that the DNA methylation landscape of young adipocyte progenitors resemble that of older wild-type mice. Together, our results demonstrate that DNMT3A coordinates both the central and local control of energy storage required to maintain normal weight and prevent inflammatory obesity.


Asunto(s)
Discapacidad Intelectual , Errores Innatos del Metabolismo , Obesidad Mórbida , Adipogénesis , Animales , ADN , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Discapacidad Intelectual/genética , Ratones
12.
J Urol ; 185(4): 1262-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21334025

RESUMEN

PURPOSE: Posterior rhabdosphincter reconstruction following radical prostatectomy was designed to improve early urinary continence. We executed a randomized clinical trial to test this conjecture in men undergoing robotic radical prostatectomy. MATERIALS AND METHODS: We conducted a phase II randomized clinical trial intended to detect a 25% difference in 3-month continence outcomes defined by a patient response of 0 or 1 to question 5 of the Expanded Prostate Cancer Index Composite questionnaire urinary domain, comparing standard running vesicourethral anastomosis (controls) to posterior rhabdosphincter reconstruction followed by standard running vesicourethral anastomosis (posterior rhabdosphincter reconstruction treated). Patients had clinically localized prostate cancer and were blinded. Surgeons were notified of computer randomization after prostate excision. Further continence outcomes were assessed by analysis of Expanded Prostate Cancer Index Composite questionnaire questions 1 and 12, International Prostate Symptom Score and 24-hour pad weights. Statistical significance was defined as p <0.05 RESULTS: A total of 94 patients were randomized, 47 to each arm. Preoperative clinical and functional variables were equivalent between study arms. There were no complications associated with either anastomotic technique. Of the 87 evaluable patients 62 (71.3%) met our 3-month continence definition. The null hypothesis was not rejected as 33 (81%) controls and 29 (63%) posterior rhabdosphincter reconstruction treated patients were continent at 3 months (chi-square p = 0.07, Fisher exact p = 0.1). Likewise there was no significant difference between arms in 24-hour pad weights (p = 0.14), International Prostate Symptom Score (p = 0.4), absence of daily leaks (p = 0.4) or perception of urinary function (p = 0.4). CONCLUSIONS: In this randomized clinical trial posterior rhabdosphincter reconstruction offered no advantage for return of early continence after robotic assisted radical prostatectomy.


Asunto(s)
Prostatectomía/métodos , Neoplasias de la Próstata/cirugía , Robótica , Incontinencia Urinaria/prevención & control , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Prostatectomía/efectos adversos , Método Simple Ciego , Resultado del Tratamiento , Incontinencia Urinaria/etiología
13.
Blood Adv ; 5(19): 3876-3890, 2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34492683

RESUMEN

IKAROS family zinc finger 1 (IKZF1) alterations represent a diverse group of genetic lesions that are associated with an increased risk of relapse in B-cell acute lymphoblastic leukemia. Due to the heterogeneity of concomitant lesions, it remains unclear how IKZF1 abnormalities directly affect cell function and therapy resistance, and whether their consideration as a prognostic indicator is valuable in improving outcome. CRISPR/Cas9 strategies were used to engineer multiple panels of isogeneic lymphoid leukemia cell lines with a spectrum of IKZF1 lesions to measure changes in chemosensitivity, gene expression, cell cycle, and in vivo engraftment that can be linked to loss of IKAROS protein. IKZF1 knockout and heterozygous null cells displayed relative resistance to a number of common therapies for B-cell acute lymphoblastic leukemia, including dexamethasone, asparaginase, and daunorubicin. Transcription profiling revealed a stem/myeloid cell-like phenotype and JAK/STAT upregulation after IKAROS loss. A CRISPR homology-directed repair strategy was also used to knock-in the dominant-negative IK6 isoform into the endogenous locus, and a similar drug resistance profile, with the exception of retained dexamethasone sensitivity, was observed. Interestingly, IKZF1 knockout and IK6 knock-in cells both have significantly increased sensitivity to cytarabine, likely owing to marked downregulation of SAMHD1 after IKZF1 knockout. Both types of IKZF1 lesions decreased the survival time of xenograft mice, with higher numbers of circulating blasts and increased organ infiltration. Given these findings, exact specification of IKZF1 status in patients may be a beneficial addition to risk stratification and could inform therapy.


Asunto(s)
Linfoma de Burkitt , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Humanos , Factor de Transcripción Ikaros/genética , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pronóstico , Recurrencia
14.
Leukemia ; 35(7): 1949-1963, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33159179

RESUMEN

Recurrent loss-of-function mutations of BCL6 co-repressor (BCOR) gene are found in about 4% of AML patients with normal karyotype and are associated with DNMT3a mutations and poor prognosis. Therefore, new anti-leukemia treatments and mouse models are needed for this combinatorial AML genotype. For this purpose, we first generated a Bcor-/- knockout mouse model characterized by impaired erythroid development (macrocytosis and anemia) and enhanced thrombopoiesis, which are both features of myelodysplasia/myeloproliferative neoplasms. We then created and characterized double Bcor-/-/Dnmt3a-/- knockout mice. Interestingly, these animals developed a fully penetrant acute erythroid leukemia (AEL) characterized by leukocytosis secondary to the expansion of blasts expressing c-Kit+ and the erythroid marker Ter119, macrocytic anemia and progressive reduction of the thrombocytosis associated with loss of Bcor alone. Transcriptomic analysis of double knockout bone marrow progenitors revealed that aberrant erythroid skewing was induced by epigenetic changes affecting specific transcriptional factors (GATA1-2) and cell-cycle regulators (Mdm2, Tp53). These findings prompted us to investigate the efficacy of demethylating agents in AEL, with significant impact on progressive leukemic burden and mice overall survival. Information gained from our model expands the knowledge on the biology of AEL and may help designing new rational treatments for patients suffering from this high-risk leukemia.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Leucemia Eritroblástica Aguda/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Anemia Macrocítica/genética , Anemia Macrocítica/patología , Animales , Médula Ósea/patología , Ciclo Celular/genética , ADN Metiltransferasa 3A , Modelos Animales de Enfermedad , Células Eritroides/patología , Leucemia Eritroblástica Aguda/patología , Ratones , Ratones Noqueados , Transcriptoma/genética
15.
Child Abuse Negl ; 110(Pt 3): 104539, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32534698

RESUMEN

BACKGROUND: Since 1995, Colorado has had a Title IV-E child welfare Stipend Program, most recently involving four universities in partnership with the Colorado Department of Human Services. OBJECTIVE: A ten-year cohort study was conducted to understand program graduates' experiences with organizational commitment, the impact of stipends on child welfare professional identity and desire to remain in the child welfare field. PARTICIPANTS AND METHODS: The mixed methods evaluation involved 245 stipend graduates from 2006 to 2016 from Bachelor of Social Work (BSW) and Master of Social Work (MSW) programs and included an online survey and focus groups. RESULTS: Results indicate stipend graduates remained in the workforce beyond payback periods, felt prepared for the job, appreciated the skills and networks gained from their programs, and expressed gratitude for the financial assistance. Graduates identified workforce factors including peer support, supervisor support, and mobility in the agency as key retention sources for remaining in child welfare.


Asunto(s)
Protección a la Infancia/psicología , Empleo/psicología , Satisfacción en el Trabajo , Lealtad del Personal , Estudiantes/psicología , Adolescente , Adulto , Estudios de Cohortes , Colorado , Femenino , Humanos , Masculino , Persona de Mediana Edad , Servicio Social/educación , Apoyo a la Formación Profesional , Universidades , Adulto Joven
16.
Nat Commun ; 11(1): 740, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029739

RESUMEN

Primary and acquired drug resistance imposes a major threat to achieving optimized clinical outcomes during cancer treatment. Aberrant changes in epigenetic modifications are closely involved in drug resistance of tumor cells. Using BET inhibitor (BETi) resistant leukemia cells as a model system, we demonstrated herein that genome-wide enhancer remodeling played a pivotal role in driving therapeutic resistance via compensational re-expression of pro-survival genes. Capitalizing on the CRISPR interference technology, we identified the second intron of IncRNA, PVT1, as a unique bona fide gained enhancer that restored MYC transcription independent of BRD4 recruitment in leukemia. A combined BETi and CDK7 inhibitor treatment abolished MYC transcription by impeding RNAPII loading without affecting PVT1-mediated chromatin looping at the MYC locus in BETi-resistant leukemia cells. Together, our findings have established the feasibility of targeting enhancer plasticity to overcome drug resistance associated with epigenetic therapies.


Asunto(s)
Leucemia Experimental/tratamiento farmacológico , Leucemia Experimental/genética , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Elementos de Facilitación Genéticos , Femenino , Genes myc/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/administración & dosificación , Humanos , Células Jurkat , Células K562 , Leucemia Experimental/metabolismo , Ratones , Modelos Genéticos , Fenilendiaminas/administración & dosificación , Pirimidinas/administración & dosificación , ARN Polimerasa II/metabolismo , ARN Largo no Codificante/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
17.
Cell Stem Cell ; 27(2): 326-335.e4, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32673568

RESUMEN

DNA methyltransferase 3A (DNMT3A) is the most commonly mutated gene in clonal hematopoiesis (CH). Somatic DNMT3A mutations arise in hematopoietic stem cells (HSCs) many years before malignancies develop, but difficulties in comparing their impact before malignancy with wild-type cells have limited the understanding of their contributions to transformation. To circumvent this limitation, we derived normal and DNMT3A mutant lymphoblastoid cell lines from a germline mosaic individual in whom these cells co-existed for nearly 6 decades. Mutant cells dominated the blood system, but not other tissues. Deep sequencing revealed similar mutational burdens and signatures in normal and mutant clones, while epigenetic profiling uncovered the focal erosion of DNA methylation at oncogenic regulatory regions in mutant clones. These regions overlapped with those sensitive to DNMT3A loss after DNMT3A ablation in HSCs and in leukemia samples. These results suggest that DNMT3A maintains a conserved DNA methylation pattern, the erosion of which provides a distinct competitive advantage to hematopoietic cells.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , Hematopoyesis , Células Clonales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Epigénesis Genética , Hematopoyesis/genética , Mutación/genética
18.
Cancer Lett ; 467: 1-8, 2019 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-31563562

RESUMEN

TET2 is among the most frequently mutated genes in hematological malignancies, as well as in healthy individuals with clonal hematopoiesis. Inflammatory stress is known to promote the expansion of Tet2-deficient hematopoietic stem cells, as well as the initiation of pre-leukemic conditions. Infection is one of the most frequent complications in hematological malignancies and antibiotics are commonly used to suppress infection-induced inflammation, but their application in TET2 mutation-associated cancers remained underexplored. In this study, we discovered that Tet2 depletion led to aberrant expansion of myeloid cells, which was correlated with elevated serum levels of pro-inflammatory cytokines at the pre-malignant stage. Antibiotics treatment suppressed the growth of Tet2-deficient myeloid and lymphoid tumor cells in vivo. Transcriptomic profiling further revealed significant changes in the expression of genes involved in the TNF-α signaling and other immunomodulatory pathways in antibiotics-treated tumor cells. Pharmacological inhibition of TNF-α signaling partially attenuated Tet2-deficient tumor cell growth in vivo. Therefore, our findings establish the feasibility of targeting pro-inflammatory pathways to curtail TET2 inactivation-associated hematological malignancies.


Asunto(s)
Antibacterianos/uso terapéutico , Proteínas de Unión al ADN/genética , Neoplasias Hematológicas/tratamiento farmacológico , Mutación con Pérdida de Función , Proteínas Proto-Oncogénicas/genética , Animales , Antibacterianos/farmacología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Citocinas/sangre , Dioxigenasas , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Ratones , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
19.
Cell Rep ; 23(1): 1-10, 2018 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-29617651

RESUMEN

Somatic mutations in DNMT3A are recurrent events across a range of blood cancers. Dnmt3a loss of function in hematopoietic stem cells (HSCs) skews divisions toward self-renewal at the expense of differentiation. Moreover, DNMT3A mutations can be detected in the blood of aging individuals, indicating that mutant cells outcompete normal HSCs over time. It is important to understand how these mutations provide a competitive advantage to HSCs. Here we show that Dnmt3a-null HSCs can regenerate over at least 12 transplant generations in mice, far exceeding the lifespan of normal HSCs. Molecular characterization reveals that this in vivo immortalization is associated with gradual and focal losses of DNA methylation at key regulatory regions associated with self-renewal genes, producing a highly stereotypical HSC phenotype in which epigenetic features are further buttressed. These findings lend insight into the preponderance of DNMT3A mutations in clonal hematopoiesis and the persistence of mutant clones after chemotherapy.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Eliminación de Gen , Hematopoyesis , Células Madre Hematopoyéticas/citología , Animales , Linaje de la Célula , Metilación de ADN , ADN Metiltransferasa 3A , Epigénesis Genética , Femenino , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL
20.
Cancer Cell ; 34(3): 499-512.e9, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30205049

RESUMEN

NPM1 is the most frequently mutated gene in cytogenetically normal acute myeloid leukemia (AML). In AML cells, NPM1 mutations result in abnormal cytoplasmic localization of the mutant protein (NPM1c); however, it is unknown whether NPM1c is required to maintain the leukemic state. Here, we show that loss of NPM1c from the cytoplasm, either through nuclear relocalization or targeted degradation, results in immediate downregulation of homeobox (HOX) genes followed by differentiation. Finally, we show that XPO1 inhibition relocalizes NPM1c to the nucleus, promotes differentiation of AML cells, and prolongs survival of Npm1-mutated leukemic mice. We describe an exquisite dependency of NPM1-mutant AML cells on NPM1c, providing the rationale for the use of nuclear export inhibitors in AML with mutated NPM1.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Proteínas Nucleares/genética , Anciano , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Carioferinas/metabolismo , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/patología , Ratones , Mutación , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteolisis , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/metabolismo , Triazoles/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA