Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 125(4): 619-28, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25416276

RESUMO

Genome sequencing studies of patient samples have implicated the involvement of various components of the epigenetic machinery in myeloid diseases, including the de novo DNA methyltransferase DNMT3A. We have recently shown that Dnmt3a is essential for hematopoietic stem cell differentiation. Here, we investigated the effect of loss of Dnmt3a on hematopoietic transformation by forcing the normally quiescent hematopoietic stem cells to divide in vivo. Mice transplanted with Dnmt3a-null bone marrow in the absence of wildtype support cells succumbed to bone marrow failure (median survival, 328 days) characteristic of myelodysplastic syndromes with symptoms including anemia, neutropenia, bone marrow hypercellularity, and splenomegaly with myeloid infiltration. Two out of 25 mice developed myeloid leukemia with >20%blasts in the blood and bone marrow. Four out of 25 primary mice succumbed to myeloproliferative disorders, some of which progressed to secondary leukemia after long latency. Exome sequencing identified cooperating c-Kit mutations found only in the leukemic samples. Ectopic introduction of c-Kit variants into a Dnmt3a-deficient background produced acute leukemia with a short latency (median survival, 67 days). Our data highlight crucial roles of Dnmt3a in normal and malignant hematopoiesis and suggest that a major role for this enzyme is to facilitate developmental progression of progenitor cells at multiple decision checkpoints.


Assuntos
Crise Blástica/mortalidade , Medula Óssea/enzimologia , Diferenciação Celular , Transformação Celular Neoplásica/metabolismo , DNA (Citosina-5-)-Metiltransferases , Células-Tronco Hematopoéticas/enzimologia , Leucemia Mieloide Aguda/enzimologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Animais , Crise Blástica/genética , Crise Blástica/patologia , Medula Óssea/patologia , Linhagem Celular , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , DNA Metiltransferase 3A , Hematopoese/genética , Células-Tronco Hematopoéticas/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , Mutação , Proteínas Proto-Oncogênicas c-kit/genética
2.
Blood Cancer Discov ; 3(3): 220-239, 2022 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-35394496

RESUMO

Clonal hematopoiesis (CH) refers to the age-related expansion of specific clones in the blood system, and manifests from somatic mutations acquired in hematopoietic stem cells (HSCs). Most CH variants occur in the gene DNMT3A, but while DNMT3A-mutant CH becomes almost ubiquitous in aging humans, a unifying molecular mechanism to illuminate how DNMT3A-mutant HSCs outcompete their counterparts is lacking. Here, we used interferon gamma (IFNγ) as a model to study the mechanisms by which Dnmt3a mutations increase HSC fitness under hematopoietic stress. We found Dnmt3a-mutant HSCs resist IFNγ-mediated depletion, and IFNγ-signaling is required for clonal expansion of Dnmt3a-mutant HSCs in vivo. Mechanistically, DNA hypomethylation-associated overexpression of Txnip in Dnmt3a-mutant HSCs leads to p53 stabilization and upregulation of p21. This preserves the functional potential of Dnmt3a-mutant HSCs through increased quiescence and resistance to IFNγ-induced apoptosis. These data identify a previously undescribed mechanism to explain increased fitness of DNMT3A-mutant clones under hematopoietic stress. SIGNIFICANCE: DNMT3A mutations are common variants in clonal hematopoiesis, and recurrent events in blood cancers. Yet the mechanisms by which these mutations provide hematopoietic stem cells a competitive advantage as a precursor to malignant transformation remain unclear. Here, we use inflammatory stress to uncover molecular mechanisms leading to this fitness advantage.See related commentary by De Dominici and DeGregori, p. 178. This article is highlighted in the In This Issue feature, p. 171.


Assuntos
DNA (Citosina-5-)-Metiltransferases , Hematopoese , Humanos , Proteínas de Transporte/genética , Hematopoiese Clonal , Células Clonais , DNA (Citosina-5-)-Metiltransferases/genética , Metilases de Modificação do DNA/genética , Hematopoese/genética , Células-Tronco Hematopoéticas
3.
Stem Cell Reports ; 14(4): 551-560, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32220332

RESUMO

The DNA methylation regulators DNMT3A and TET2 are recurrently mutated in hematological disorders. Despite possessing antagonistic biochemical activities, loss-of-function murine models show overlapping phenotypes in terms of increased hematopoietic stem cell (HSC) fitness. Here, we directly compared the effects of these mutations on hematopoietic progenitor function and disease initiation. In contrast to Dnmt3a-null HSCs, which possess limitless self-renewal in vivo, Tet2-null HSCs unexpectedly exhaust at the same rate as control HSCs in serial transplantation assays despite an initial increase in self-renewal. Moreover, loss of Tet2 more acutely sensitizes hematopoietic cells to the addition of a common co-operating mutation (Flt3ITD) than loss of Dnmt3a, which is associated with a more rapid expansion of committed progenitor cells. The effect of Tet2 mutation manifests more profound myeloid lineage skewing in committed hematopoietic progenitor cells rather than long-term HSCs. Molecular characterization revealed divergent transcriptomes and chromatin accessibility underlying these functional differences.


Assuntos
DNA (Citosina-5-)-Metiltransferases/genética , Proteínas de Ligação a DNA/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Mutação/genética , Proteínas Proto-Oncogênicas/genética , Adaptação Fisiológica , Animais , Diferenciação Celular , Autorrenovação Celular , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Proteínas Proto-Oncogênicas/metabolismo
4.
Leukemia ; 33(10): 2506-2521, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30936419

RESUMO

The histone demethylase KDM6B (JMJD3) is upregulated in blood disorders, suggesting that it may have important pathogenic functions. Here we examined the function of Kdm6b in hematopoietic stem cells (HSC) to evaluate its potential as a therapeutic target. Loss of Kdm6b lead to depletion of phenotypic and functional HSCs in adult mice, and Kdm6b is necessary for HSC self-renewal in response to inflammatory and proliferative stress. Loss of Kdm6b leads to a pro-differentiation poised state in HSCs due to the increased expression of the AP-1 transcription factor complex (Fos and Jun) and immediate early response (IER) genes. These gene expression changes occurred independently of chromatin modifications. Targeting AP-1 restored function of Kdm6b-deficient HSCs, suggesting that Kdm6b regulates this complex during HSC stress response. We also show Kdm6b supports developmental context-dependent leukemogenesis for T-cell acute lymphoblastic leukemia (T-ALL) and M5 acute myeloid leukemia (AML). Kdm6b is required for effective fetal-derived T-ALL and adult-derived AML, but not vice versa. These studies identify a crucial role for Kdm6b in regulating HSC self-renewal in different contexts, and highlight the potential of KDM6B as a therapeutic target in different hematopoietic malignancies.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Autorrenovação Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Diferenciação Celular/genética , Autorrenovação Celular/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/patologia , Fatores de Transcrição/genética , Regulação para Cima/genética
5.
Exp Hematol ; 57: 14-20, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28939416

RESUMO

Genome sequencing efforts have identified virtually all of the important mutations in adult myeloid malignancies. More recently, population studies have identified cancer-associated variants in the blood of otherwise healthy individuals as they age, a phenomenon termed clonal hematopoiesis of indeterminate potential (CHIP). This suggests that these mutations may occur in hematopoietic stem cells (HSCs) long before any clinical presentation but are not necessarily harbingers of transformation because only a fraction of individuals with CHIP develop hematopoietic pathologies. Delineation between CHIP variants that predispose for disease versus those that are more benign could be used as a prognostic factor to identify individuals at greater risk for transformation. To achieve this, the biological impact of CHIP variants on HSC function must be validated. One variant that has been identified recurrently in CHIP is a gain-of-function missense mutation in the imprinted gene GNAS (Guanine Nucleotide Binding Protein, Alpha Stimulating). In this study, we examined the effect of the GNASR201C variant on HSC function. Ectopic expression of GNASR201C supported transplantable HSC activity and improved lymphoid output in secondary recipients. Because declining lymphoid output is a hallmark of aging, GNASR201C mutations may sustain lymphoid-biased HSCs over time and maintain them in a developmental state favorable for transformation.


Assuntos
Cromograninas/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Mutação com Ganho de Função , Hematopoese/genética , Mutação de Sentido Incorreto , Animais , Transplante de Medula Óssea , Contagem de Células , Senescência Celular , Cromograninas/fisiologia , DNA Complementar/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/fisiologia , Redes Reguladoras de Genes , Genes Reporter , Vetores Genéticos/genética , Hematopoese/fisiologia , Células-Tronco Hematopoéticas , Humanos , Lentivirus/genética , Linfopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Transdução Genética , Quimeras de Transplante
6.
Cancer Cell ; 34(5): 741-756.e8, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-30423295

RESUMO

How specific genetic lesions contribute to transformation of non-malignant myeloproliferative neoplasms (MPNs) and myelodysplastic syndromes (MDSs) to secondary acute myeloid leukemia (sAML) are poorly understood. JARID2 is lost by chromosomal deletions in a proportion of MPN/MDS cases that progress to sAML. In this study, genetic mouse models and patient-derived xenografts demonstrated that JARID2 acts as a tumor suppressor in chronic myeloid disorders. Genetic deletion of Jarid2 either reduced overall survival of animals with MPNs or drove transformation to sAML, depending on the timing and context of co-operating mutations. Mechanistically, JARID2 recruits PRC2 to epigenetically repress self-renewal pathways in hematopoietic progenitor cells. These studies establish JARID2 as a bona fide hematopoietic tumor suppressor and highlight potential therapeutic targets.


Assuntos
Autorrenovação Celular/genética , Transformação Celular Neoplásica/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicas/genética , Transtornos Mieloproliferativos/genética , Complexo Repressor Polycomb 2/genética , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Autorrenovação Celular/fisiologia , Transformação Celular Neoplásica/patologia , Feminino , Deleção de Genes , Técnicas de Silenciamento de Genes , Genes Supressores de Tumor , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndromes Mielodisplásicas/patologia , Transtornos Mieloproliferativos/patologia , Proteína Proto-Oncogênica N-Myc/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Proteína 1 Parceira de Translocação de RUNX1/metabolismo , Transplante Heterólogo
7.
BMC Proc ; 8(Suppl 1 Genetic Analysis Workshop 18Vanessa Olmo): S10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25519353

RESUMO

Sequence kernel association test (SKAT) has become one of the most commonly used nonburden tests for analyzing rare variants. Performance of burden tests depends on the weighting of rare and common variants when collapsing them in a genomic region. Using the systolic and diastolic blood pressure phenotypes of 142 unrelated individuals in the Genetic Analysis Workshop 18 data, we investigated whether performance of SKAT also depends on the weighting scheme. We analyzed the entire sequencing data for all 200 replications using 3 weighting schemes: equal weighting, Madsen-Browning weighting, and SKAT default linear weighting. We considered two options: all single-nucleotide polymorphisms (SNPs) and only low-frequency SNPs. A SKAT default weighting scheme (which heavily downweights common variants) performed better for the genes in which causal SNPs are mostly rare. This SKAT default weighting scheme behaved similarly to other weighting schemes after eliminating all common SNPs. In contrast, the equal weighting scheme performed the best for MAP4 and FLT3, both of which included a common variant with a large effect. However, SKAT with all 3 weighting schemes performed poorly. Overall power across all causal genes was about 0.05, which was almost identical to the type I error rate. This poor performance is partly due to a small sample size because of the need to analyze only unrelated individuals. Because a half of causal SNPs were not found in the annotation file based on the 1000 Genomes Project, we suspect that performance was also affected by our use of incomplete annotation information.

8.
Exp Hematol ; 42(4): 317-327.e2, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24373928

RESUMO

Recent findings from several groups have identified distinct classes of hematopoietic stem cells (HSCs) in the bone marrow, each with inherent functional biases in terms of their differentiation, self-renewal, proliferation, and lifespan. It has previously been demonstrated that myeloid- and lymphoid-biased HSCs can be prospectively enriched based on their degree of Hoechst dye efflux. In the present study, we used differential Hoechst efflux to enrich lineage-biased HSC subtypes and analyzed their functional potentials. Despite similar outputs in vitro, bone marrow transplantation assays revealed contrasting lineage differentiation in vivo. To stratify the molecular differences underlying these contrasting functional potentials at the clonal level, single-cell gene expression analysis was performed using the Fluidigm BioMark system and revealed dynamic expression of genes including Meis1, CEBP/α, Sfpi1, and Dnmt3a. Finally, single-cell gene expression analysis was used to unravel the opposing proliferative responses of lineage-biased HSCs to the growth factor TGF-ß1, revealing a potential role for the cell cycle inhibitor Cdkn1c as molecular mediator. This work lends further credence to the concept of HSC heterogeneity, and it presents unprecedented molecular resolution of the HSC response to trophic factors using single-cell gene expression analysis.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , Regulação da Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Aloenxertos , Animais , Transplante de Medula Óssea , Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Inibidor de Quinase Dependente de Ciclina p57/biossíntese , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/biossíntese , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Transativadores/biossíntese , Fator de Crescimento Transformador beta1/biossíntese
9.
Cell Stem Cell ; 15(3): 350-364, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25130491

RESUMO

Epigenetic regulation of hematopoietic stem cells (HSCs) ensures lifelong production of blood and bone marrow. Recently, we reported that loss of de novo DNA methyltransferase Dnmt3a results in HSC expansion and impaired differentiation. Here, we report conditional inactivation of Dnmt3b in HSCs either alone or combined with Dnmt3a deletion. Combined loss of Dnmt3a and Dnmt3b was synergistic, resulting in enhanced HSC self-renewal and a more severe block in differentiation than in Dnmt3a-null cells, whereas loss of Dnmt3b resulted in a mild phenotype. Although the predominant Dnmt3b isoform in adult HSCs is catalytically inactive, its residual activity in Dnmt3a-null HSCs can drive some differentiation and generates paradoxical hypermethylation of CpG islands. Dnmt3a/Dnmt3b-null HSCs displayed activated ß-catenin signaling, partly accounting for the differentiation block. These data demonstrate distinct roles for Dnmt3b in HSC differentiation and provide insights into complementary de novo methylation patterns governing regulation of HSC fate decisions.


Assuntos
DNA (Citosina-5-)-Metiltransferases/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Animais , Apoptose , Diferenciação Celular/genética , Proliferação de Células , Ilhas de CpG/genética , Metilação de DNA/genética , DNA Metiltransferase 3A , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Isoenzimas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/metabolismo , Neoplasias/patologia , beta Catenina/metabolismo , DNA Metiltransferase 3B
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA