Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Nature ; 543(7644): 205-210, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28241143

RESUMEN

With age, haematopoietic stem cells lose their ability to regenerate the blood system, and promote disease development. Autophagy is associated with health and longevity, and is critical for protecting haematopoietic stem cells from metabolic stress. Here we show that loss of autophagy in haematopoietic stem cells causes accumulation of mitochondria and an activated metabolic state, which drives accelerated myeloid differentiation mainly through epigenetic deregulations, and impairs haematopoietic stem-cell self-renewal activity and regenerative potential. Strikingly, most haematopoietic stem cells in aged mice share these altered metabolic and functional features. However, approximately one-third of aged haematopoietic stem cells exhibit high autophagy levels and maintain a low metabolic state with robust long-term regeneration potential similar to healthy young haematopoietic stem cells. Our results demonstrate that autophagy actively suppresses haematopoietic stem-cell metabolism by clearing active, healthy mitochondria to maintain quiescence and stemness, and becomes increasingly necessary with age to preserve the regenerative capacity of old haematopoietic stem cells.


Asunto(s)
Autofagia , Autorrenovación de las Células , Senescencia Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Animales , Autofagia/genética , Autorrenovación de las Células/genética , Senescencia Celular/genética , Epigénesis Genética , Femenino , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratones , Mitocondrias/metabolismo , Células Mieloides/citología , Células Mieloides/metabolismo
2.
Curr Opin Hematol ; 28(1): 57-63, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33186152

RESUMEN

PURPOSE OF REVIEW: Our understanding of the effects of aging on human hematopoiesis has advanced significantly in recent years, yet the full ramifications of these findings are not fully understood. This review summarizes these findings and discusses their implication as they relate to malignant hematopoiesis. RECENT FINDINGS: With human aging there is an impaired immune response, loss of hematopoietic stem cell (HSC) function, increase in clonal hematopoiesis, and higher frequency of myeloid malignancies. Although murine models have implicated abnormalities in DNA damage repair, autophagy, metabolism, and epigenetics, studies in primary human specimens are more limited. The development of age-related clonal hematopoiesis and the risk associated with this is one of the major findings in the field of recent years. This is accompanied by changes in bone marrow stem and progenitor composition, changes in the epigenetic program of stem cells and an inflammatory milieu in the bone marrow. The precise consequences of these changes for the development of age-related malignancies are still unclear. SUMMARY: Advances in the field have begun to reveal the mechanisms driving human HSC loss of function with age. It will be critical to delineate between normal and malignant aging in order to better prevent age-associated myeloid malignancies.


Asunto(s)
Envejecimiento , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucemia/patología , Animales , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunidad , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo
3.
Haematologica ; 105(4): 905-913, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31171641

RESUMEN

Healthy bone marrow progenitors yield a co-ordinated balance of hematopoietic lineages. This balance shifts with aging toward enhanced granulopoiesis with diminished erythropoiesis and lymphopoiesis, changes which likely contribute to the development of bone marrow disorders in the elderly. In this study, RUNX3 was identified as a hematopoietic stem and progenitor cell factor whose levels decline with aging in humans and mice. This decline is exaggerated in hematopoietic stem and progenitor cells from subjects diagnosed with unexplained anemia of the elderly. Hematopoietic stem cells from elderly unexplained anemia patients had diminished erythroid but unaffected granulocytic colony forming potential. Knockdown studies revealed human hematopoietic stem and progenitor cells to be strongly influenced by RUNX3 levels, with modest deficiencies abrogating erythroid differentiation at multiple steps while retaining capacity for granulopoiesis. Transcriptome profiling indicated control by RUNX3 of key erythroid transcription factors, including KLF1 and GATA1 These findings thus implicate RUNX3 as a participant in hematopoietic stem and progenitor cell aging, and a key determinant of erythroid-myeloid lineage balance.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Anciano , Envejecimiento , Animales , Diferenciación Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Eritropoyesis , Humanos , Ratones
4.
Cancer Discov ; 12(2): 450-467, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34531253

RESUMEN

An enhanced requirement for nutrients is a hallmark property of cancer cells. Here, we optimized an in vivo genetic screening strategy in acute myeloid leukemia (AML), which led to the identification of the myo-inositol transporter SLC5A3 as a dependency in this disease. We demonstrate that SLC5A3 is essential to support a myo-inositol auxotrophy in AML. The commonality among SLC5A3-dependent AML lines is the transcriptional silencing of ISYNA1, which encodes the rate-limiting enzyme for myo-inositol biosynthesis, inositol-3-phosphate synthase 1. We use gain- and loss-of-function experiments to reveal a synthetic lethal genetic interaction between ISYNA1 and SLC5A3 in AML, which function redundantly to sustain intracellular myo-inositol. Transcriptional silencing and DNA hypermethylation of ISYNA1 occur in a recurrent manner in human AML patient samples, in association with IDH1/IDH2 and CEBPA mutations. Our findings reveal myo-inositol as a nutrient dependency in AML caused by the aberrant silencing of a biosynthetic enzyme. SIGNIFICANCE: We show how epigenetic silencing can provoke a nutrient dependency in AML by exploiting a synthetic lethality relationship between biosynthesis and transport of myo-inositol. Blocking the function of this solute carrier may have therapeutic potential in an epigenetically defined subset of AML.This article is highlighted in the In This Issue feature, p. 275.


Asunto(s)
Proteínas de Choque Térmico/genética , Inositol/biosíntesis , Leucemia Mieloide Aguda/tratamiento farmacológico , Simportadores/genética , Animales , Biología Evolutiva , Humanos , Ratones
5.
Blood Cancer Discov ; 3(2): 116-135, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35015684

RESUMEN

Polycomb repressive epigenetic complexes are recurrently dysregulated in cancer. Unlike polycomb repressive complex 2 (PRC2), the role of PRC1 in oncogenesis and therapy resistance is not well-defined. Here, we demonstrate that highly recurrent mutations of the PRC1 subunits BCOR and BCORL1 in leukemia disrupt assembly of a noncanonical PRC1.1 complex, thereby selectively unlinking the RING-PCGF enzymatic core from the chromatin-targeting auxiliary subcomplex. As a result, BCOR-mutated PRC1.1 is localized to chromatin but lacks repressive activity, leading to epigenetic reprogramming and transcriptional activation at target loci. We define a set of functional targets that drive aberrant oncogenic signaling programs in PRC1.1-mutated cells and primary patient samples. Activation of these PRC1.1 targets in BCOR-mutated cells confers acquired resistance to treatment while sensitizing to targeted kinase inhibition. Our study thus reveals a novel epigenetic mechanism that explains PRC1.1 tumor-suppressive activity and identifies a therapeutic strategy in PRC1.1-mutated cancer. SIGNIFICANCE: We demonstrate that BCOR and BCORL1 mutations in leukemia unlink PRC1.1 repressive function from target genes, resulting in epigenetic reprogramming and activation of aberrant cell signaling programs that mediate treatment resistance. Our study provides mechanistic insights into the pathogenesis of PRC1.1-mutated leukemia that inform novel therapeutic approaches. This article is highlighted in the In This Issue feature, p. 85.


Asunto(s)
Carcinogénesis , Leucemia , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Cromatina , Epigénesis Genética/genética , Humanos , Leucemia/genética , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Transducción de Señal/genética
6.
Cancer Discov ; 11(9): 2300-2315, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33893150

RESUMEN

Hundreds of genes become aberrantly silenced in acute myeloid leukemia (AML), with most of these epigenetic changes being of unknown functional consequence. Here, we demonstrate how gene silencing can lead to an acquired dependency on the DNA repair machinery in AML. We make this observation by profiling the essentiality of the ubiquitination machinery in cancer cell lines using domain-focused CRISPR screening, which revealed Fanconi anemia (FA) proteins UBE2T and FANCL as unique dependencies in AML. We demonstrate that these dependencies are due to a synthetic lethal interaction between FA proteins and aldehyde dehydrogenase 2 (ALDH2), which function in parallel pathways to counteract the genotoxicity of endogenous aldehydes. We show DNA hypermethylation and silencing of ALDH2 occur in a recurrent manner in human AML, which is sufficient to confer FA pathway dependency. Our study suggests that targeting of the ubiquitination reaction catalyzed by FA proteins can eliminate ALDH2-deficient AML. SIGNIFICANCE: Aberrant gene silencing is an epigenetic hallmark of human cancer, but the functional consequences of this process are largely unknown. In this study, we show how an epigenetic alteration leads to an actionable dependency on a DNA repair pathway through the disabling of genetic redundancy.This article is highlighted in the In This Issue feature, p. 2113.


Asunto(s)
Aldehído Deshidrogenasa Mitocondrial/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Leucemia Mieloide Aguda/genética , Línea Celular Tumoral , Humanos , Ubiquitinación
7.
Front Immunol ; 11: 536442, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33224133

RESUMEN

Hematopoietic stem and progenitor cells (HSPC) experience a functional decline in response to chronic inflammation or aging. Haploinsufficiency of A20, or TNFAIP3, an innate immune regulator, is associated with a variety of autoimmune, inflammatory, and hematologic malignancies. Based on a prior analysis of epigenomic and transcriptomic changes during normal human aging, we find that the expression of A20 is significantly reduced in aged HSPC as compared to young HSPC. Here, we show that the partial reduction of A20 expression in young HSPC results in characteristic features of aging. Specifically, heterozygous deletion of A20 in hematopoietic cells resulted in expansion of the HSPC pool, reduced HSPC fitness, and myeloid-biased hematopoiesis. These findings suggest that altered expression of A20 in HSPC contributes to an aging-like phenotype, and that there may be a common underlying mechanism for diminished HSPC function between inflammatory states and aging.


Asunto(s)
Envejecimiento/inmunología , Hematopoyesis , Células Madre Hematopoyéticas/inmunología , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/inmunología , Envejecimiento/genética , Envejecimiento/patología , Animales , Eliminación de Gen , Células Madre Hematopoyéticas/patología , Heterocigoto , Ratones , Ratones Transgénicos , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética
8.
Genome Biol ; 21(1): 247, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32933554

RESUMEN

BACKGROUND: The three-dimensional genome organization is critical for gene regulation and can malfunction in diseases like cancer. As a key regulator of genome organization, CCCTC-binding factor (CTCF) has been characterized as a DNA-binding protein with important functions in maintaining the topological structure of chromatin and inducing DNA looping. Among the prolific binding sites in the genome, several events with altered CTCF occupancy have been reported as associated with effects in physiology or disease. However, hitherto there is no comprehensive survey of genome-wide CTCF binding patterns across different human cancers. RESULTS: To dissect functions of CTCF binding, we systematically analyze over 700 CTCF ChIP-seq profiles across human tissues and cancers and identify cancer-specific CTCF binding patterns in six cancer types. We show that cancer-specific lost and gained CTCF binding events are associated with altered chromatin interactions, partially with DNA methylation changes, and rarely with sequence mutations. While lost bindings primarily occur near gene promoters, most gained CTCF binding events exhibit enhancer activities and are induced by oncogenic transcription factors. We validate these findings in T cell acute lymphoblastic leukemia cell lines and patient samples and show that oncogenic NOTCH1 induces specific CTCF binding and they cooperatively activate expression of target genes, indicating transcriptional condensation phenomena. CONCLUSIONS: Specific CTCF binding events occur in human cancers. Cancer-specific CTCF binding can be induced by other transcription factors to regulate oncogenic gene expression. Our results substantiate CTCF binding alteration as a functional epigenomic signature of cancer.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias/metabolismo , Metilación de ADN , Humanos , Oncogenes , Receptor Notch1/metabolismo
9.
Cancer Discov ; 9(8): 1080-1101, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31085557

RESUMEN

Aging is associated with functional decline of hematopoietic stem cells (HSC) as well as an increased risk of myeloid malignancies. We performed an integrative characterization of epigenomic and transcriptomic changes, including single-cell RNA sequencing, during normal human aging. Lineage-CD34+CD38- cells [HSC-enriched (HSCe)] undergo age-associated epigenetic reprogramming consisting of redistribution of DNA methylation and reductions in H3K27ac, H3K4me1, and H3K4me3. This reprogramming of aged HSCe globally targets developmental and cancer pathways that are comparably altered in acute myeloid leukemia (AML) of all ages, encompassing loss of 4,646 active enhancers, 3,091 bivalent promoters, and deregulation of several epigenetic modifiers and key hematopoietic transcription factors, such as KLF6, BCL6, and RUNX3. Notably, in vitro downregulation of KLF6 results in impaired differentiation, increased colony-forming potential, and changes in expression that recapitulate aging and leukemia signatures. Thus, age-associated epigenetic reprogramming may form a predisposing condition for the development of age-related AML. SIGNIFICANCE: AML, which is more frequent in the elderly, is characterized by epigenetic deregulation. We demonstrate that epigenetic reprogramming of human HSCs occurs with age, affecting cancer and developmental pathways. Downregulation of genes epigenetically altered with age leads to impairment in differentiation and partially recapitulates aging phenotypes.This article is highlighted in the In This Issue feature, p. 983.


Asunto(s)
Diferenciación Celular/genética , Reprogramación Celular/genética , Senescencia Celular/genética , Epigénesis Genética , Células Madre Hematopoyéticas/metabolismo , Leucemia/genética , Leucemia/metabolismo , Citosina/metabolismo , Metilación de ADN , Susceptibilidad a Enfermedades , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Histonas/metabolismo , Humanos , Factor 6 Similar a Kruppel/genética , Factor 6 Similar a Kruppel/metabolismo , Leucemia/patología , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
10.
Leukemia ; 33(10): 2481-2494, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30923319

RESUMEN

The unbalanced translocation dic(1;7)(q10;p10) in myelodysplastic syndromes (MDS) is originated by centromeric juxtaposition resulting into 1q trisomy and 7q monosomy. More than half of cases arise after chemo/radio-therapy. To date, given the absence of genes within the centromeric regions, no specific molecular events have been identified in this cytogenetic subgroup. We performed the first comprehensive genetic and epigenetic analysis of MDS with dic(1;7)(q10;p10) compared to normal controls and therapy-related myeloid neoplasms (t-MNs). RNA-seq showed a unique downregulated signature in dic(1;7) cases, affecting more than 80% of differentially expressed genes. As revealed by pathway and gene ontology analyses, downregulation of ATP-binding cassette (ABC) transporters and lipid-related genes and upregulation of p53 signaling were the most relevant biological features of dic(1;7). Epigenetic supervised analysis revealed hypermethylation at intronic enhancers in the dicentric subgroup, in which low expression levels of enhancer putative target genes accounted for around 35% of the downregulated signature. Enrichment of Krüppel-like transcription factor binding sites emerged at enhancers. Furthermore, a specific hypermethylated pattern on 1q was found to underlie the hypo-expression of more than 50% of 1q-deregulated genes, despite trisomy. In summary, dic(1;7) in MDS establishes a specific transcriptional program driven by a unique epigenomic signature.


Asunto(s)
Epigénesis Genética/genética , Síndromes Mielodisplásicos/genética , Translocación Genética/genética , Adulto , Anciano , Anciano de 80 o más Años , Sitios de Unión/genética , Trastornos de los Cromosomas/genética , Regulación hacia Abajo/genética , Epigenómica/métodos , Femenino , Humanos , Cariotipificación/métodos , Masculino , Persona de Mediana Edad , Monosomía/genética , Estudios Retrospectivos , Transcripción Genética/genética , Trisomía/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA