Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26660446

RESUMEN

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Asunto(s)
Anemia Aplásica/genética , Enfermedades de la Médula Ósea/genética , Médula Ósea/patología , Factor de Transcripción GATA2/genética , Hemoglobinuria Paroxística/genética , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas c-kit/deficiencia , Eliminación de Secuencia , Dedos de Zinc/genética , Anemia Aplásica/diagnóstico , Anemia Aplásica/metabolismo , Anemia Aplásica/mortalidad , Animales , Biomarcadores , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/mortalidad , Trastornos de Fallo de la Médula Ósea , Huesos/patología , Inmunoprecipitación de Cromatina , Descalcificación Patológica/genética , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/química , Factor de Transcripción GATA2/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Genes Reporteros , Genotipo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/mortalidad , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunofenotipificación , Ratones , Ratones Noqueados , Pronóstico , Células de Población Lateral
2.
Stem Cells ; 32(11): 3023-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25078851

RESUMEN

Interferon gamma (IFNγ) promotes cell division of hematopoietic stem cells (HSCs) without affecting the total HSC number. We postulated that IFNγ stimulates differentiation of HSCs as part of the innate immune response. Here, we report that type II interferon signaling is required, both at baseline and during an animal model of LCMV infection, to maintain normal myeloid development. By separately evaluating myeloid-biased and lymphoid-biased HSC subtypes, we found that myeloid-biased HSCs express higher levels of IFNγ receptor and are specifically activated to divide after recombinant IFNγ exposure in vivo. While both HSC subtypes show increased expression of the transcription factor C/EBPß after infection, only the myeloid-biased HSCs are transiently depleted from the marrow during the type II interferon-mediated immune response to Mycobacterium avium infection, as measured both functionally and phenotypically. These findings indicate that IFNγ selectively permits differentiation of myeloid-biased HSCs during an innate immune response to infection. This represents the first report of a context and a mechanism for discriminate utilization of the alternate HSC subtypes. Terminal differentiation, at the expense of self-renewal, may compromise HSC populations during states of chronic inflammation.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Interferón gamma/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones Endogámicos C57BL , Células Mieloides/citología , Transducción de Señal/genética
3.
BMC Cancer ; 13: 371, 2013 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-23915344

RESUMEN

BACKGROUND: Uveal melanoma is a highly aggressive cancer with a strong propensity for metastasis, yet little is known about the biological mechanisms underlying this metastatic potential. We recently showed that most metastasizing uveal melanomas, which exhibit a class 2 gene expression profile, contain inactivating mutations in the tumor suppressor BAP1. The aim of this study was to investigate the role of BAP1 in uveal melanoma progression. METHODS: Uveal melanoma cells were studied following RNAi-mediated depletion of BAP1 using proliferation, BrdU incorporation, flow cytometry, migration, invasion, differentiation and clonogenic assays, as well as in vivo tumorigenicity experiments in NOD-SCID-Gamma mice. RESULTS: Depletion of BAP1 in uveal melanoma cells resulted in a loss of differentiation and gain of stem-like properties, including expression of stem cell markers, increased capacity for self-replication, and enhanced ability to grow in stem cell conditions. BAP1 depletion did not result in increased proliferation, migration, invasion or tumorigenicity. CONCLUSIONS: BAP1 appears to function in the uveal melanocyte lineage primarily as a regulator of differentiation, with cells deficient for BAP1 exhibiting stem-like qualities. It will be important to elucidate how this effect of BAP1 loss promotes metastasis and how to reverse this effect therapeutically.


Asunto(s)
Melanocitos/metabolismo , Melanocitos/patología , Melanoma/genética , Melanoma/patología , Proteínas Supresoras de Tumor/deficiencia , Ubiquitina Tiolesterasa/deficiencia , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/patología , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Xenoinjertos , Factor C1 de la Célula Huésped/metabolismo , Humanos , Neoplasias Hepáticas/secundario , Masculino , Melanoma/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Fenotipo , Unión Proteica , Interferencia de ARN , Reproducibilidad de los Resultados , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Neoplasias de la Úvea/metabolismo
4.
Cell Stem Cell ; 28(8): 1428-1442.e6, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-33743191

RESUMEN

Age-related clonal hematopoiesis (CH) is a risk factor for malignancy, cardiovascular disease, and all-cause mortality. Somatic mutations in DNMT3A are drivers of CH, but decades may elapse between the acquisition of a mutation and CH, suggesting that environmental factors contribute to clonal expansion. We tested whether infection provides selective pressure favoring the expansion of Dnmt3a mutant hematopoietic stem cells (HSCs) in mouse chimeras. We created Dnmt3a-mosaic mice by transplanting Dnmt3a-/- and WT HSCs into WT mice and observed the substantial expansion of Dnmt3a-/- HSCs during chronic mycobacterial infection. Injection of recombinant IFNγ alone was sufficient to phenocopy CH by Dnmt3a-/- HSCs upon infection. Transcriptional and epigenetic profiling and functional studies indicate reduced differentiation associated with widespread methylation alterations, and reduced secondary stress-induced apoptosis accounts for Dnmt3a-/- clonal expansion during infection. DNMT3A mutant human HSCs similarly exhibit defective IFNγ-induced differentiation. We thus demonstrate that IFNγ signaling induced during chronic infection can drive DNMT3A-loss-of-function CH.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas , Hematopoyesis , Animales , Hematopoyesis Clonal , ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Hematopoyéticas , Ratones , Mutación
5.
Exp Hematol ; 91: 39-45.e2, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32961298

RESUMEN

Mutations in the gene encoding DNA methyltransferase 3A (DNMT3A) comprise the majority of mutations found in clonal hematopoiesis (CH), an age-related condition that was recently found to affect outcomes in patients undergoing hematopoietic stem cell transplant (HSCT). Recent studies have indicated that patients with CH have worse prognoses after HSCT, suggesting stress imposed by HSCT preconditioning agents may impact hematopoietic stem cell (HSC) dynamics in transplant recipients. In this study, we used a competitive transplantation mouse model to investigate how treatment with the common preconditioning agents 5-fluorouracil (5-FU) and busulfan (BU) affect the prevalence of Dnmt3a-/- HSCs and progenitor cells in competition with wild-type cells. We found that, though sufficient to deplete peripheral blood counts, 5-FU preconditioning did not significantly alter the frequency of Dnmt3a-null hematopoietic stem and progenitor cells (HSPCs) in mosaic mice. In contrast, mice treated with BU had a sevenfold decline in total bone marrow cells and an increase in Dnmt3a-null HSPCs that was detectable in peripheral blood. Indeed, even though all mosaic mice had a starting engraftment of ∼10%-40%, 85%-100% of HSPCs were Dnmt3a-null in four of seven mice after BU treatment, indicating these cells expand dramatically during recovery. Overall, these results suggest that individual preconditioning regimens have different effects on the expansion of Dnmt3a-mutant cells in patients with pre-existing CH. Thus, the presence of CH-associated mutants should be evaluated prior to selecting preconditioning regimens for HSCT.


Asunto(s)
Busulfano/farmacología , ADN (Citosina-5-)-Metiltransferasas/deficiencia , Hematopoyesis/genética , Factores de Crecimiento de Célula Hematopoyética/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Médula Ósea/patología , División Celular/efectos de los fármacos , Linaje de la Célula , Células Clonales , ADN Metiltransferasa 3A , Fluorouracilo/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Ratones , Quimera por Radiación
6.
Cell Rep ; 33(12): 108530, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33357430

RESUMEN

During chronic infection, the inflammatory cytokine interferon gamma (IFNγ) damages hematopoietic stem cells (HSCs) by disrupting quiescence and promoting excessive terminal differentiation. However, the mechanism by which IFNγ hinders HSC quiescence remains undefined. Using intravital 3-dimensional microscopy, we find that IFNγ disrupts the normally close interaction between HSCs and CXCL12-abundant reticular (CAR) cells in the HSC niche. IFNγ stimulation increases expression of the cell surface protein BST2, which we find is required for IFNγ-dependent HSC relocalization and activation. IFNγ stimulation of HSCs increases their E-selectin binding by BST2 and homing to the bone marrow, which depends on E-selectin binding. Upon chronic infection, HSCs from mice lacking BST2 are more quiescent and more resistant to depletion than HSCs from wild-type mice. Overall, this study defines a critical mechanism by which IFNγ promotes niche relocalization and activation in response to inflammatory stimulation and identifies BST2 as a key regulator of HSC quiescence. VIDEO ABSTRACT.


Asunto(s)
Antígenos CD/inmunología , Células Madre Hematopoyéticas/inmunología , Interferón gamma/inmunología , Glicoproteínas de Membrana/inmunología , Animales , Quimiocina CXCL12/inmunología , Selectina E/inmunología , Proteínas Ligadas a GPI/inmunología , Humanos , Interferón gamma/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
7.
Methods Mol Biol ; 1686: 91-103, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29030815

RESUMEN

Cellular quiescence is a key component of hematopoietic stem cell (HSC) homeostasis; therefore, a reliable method to measure HSC cell division is critical in many studies. However, measuring the proliferation rate of largely quiescent and rare populations of cells can be challenging. Bromo-deoxyuridine (BrdU) incorporation into replicating DNA is a commonly used and highly reproducible method to detect cell division history. Here, we describe a protocol for BrdU incorporation analysis in hematopoietic stem and progenitor cells that can provide a sensitive measure of cell division even in rare cell populations. In combination with flow cytometry, this method can be generalized to analyze other cell populations and other tissues as identified by cell surface markers.


Asunto(s)
Bromodesoxiuridina/metabolismo , Proliferación Celular , Citometría de Flujo/métodos , Células Madre Hematopoyéticas/citología , Ciclo Celular , División Celular , Células Madre Hematopoyéticas/metabolismo , Homeostasis , Humanos
8.
Cell Rep ; 17(10): 2584-2595, 2016 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-27926863

RESUMEN

Chronic infections affect a third of the world's population and can cause bone marrow suppression, a severe condition that increases mortality from infection. To uncover the basis for infection-associated bone marrow suppression, we conducted repeated infection of WT mice with Mycobacterium avium. After 4-6 months, mice became pancytopenic. Their hematopoietic stem and progenitor cells (HSPCs) were severely depleted and displayed interferon gamma (IFN-γ) signaling-dependent defects in self-renewal. There was no evidence of increased HSPC mobilization or apoptosis. However, consistent with known effects of IFN-γ, transcriptome analysis pointed toward increased myeloid differentiation of HSPCs and revealed the transcription factor Batf2 as a potential mediator of IFN-γ-induced HSPC differentiation. Gain- and loss-of-function studies uncovered a role for Batf2 in myeloid differentiation in both murine and human systems. We thus demonstrate that chronic infection can deplete HSPCs and identify BATF2 as a mediator of infection-induced HSPC terminal differentiation.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Infecciones/genética , Animales , Células de la Médula Ósea/metabolismo , Proliferación Celular/genética , Autorrenovación de las Células/genética , Humanos , Infecciones/microbiología , Infecciones/patología , Interferón gamma/genética , Ratones , Mycobacterium avium/patogenicidad , Transducción de Señal
9.
Exp Hematol ; 43(10): 912-918.e2, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26072330

RESUMEN

Interferon α (IFNα) is used clinically to restore polyclonal hematopoiesis in patients with the myeloproliferative neoplasms polycythemia vera and essential thrombocythemia and to improve chemosensitivity in chronic myeloid leukemia patients. However, the mechanisms by which IFNα affects disease-initiating hematopoietic stem and progenitor cells (HSPCs) remain poorly understood. Although IFNα has been found to transiently impair quiescence of murine hematopoietic stem cells, its effects on human HSPCs have not been studied in vivo. Here, we compared bone marrow serially obtained from patients with myeloproliferative neoplasms before and during pegylated IFNα treatment against marrow serially obtained from patients on hydroxyurea. The percentage of HSPCs actively undergoing cell cycle was increased after pegylated IFNα treatment in a majority of patients compared with hydroxyurea-treated controls, suggesting that IFNα promotes cell division. Furthermore, transcriptional profiling revealed that cell cycle-associated genes were induced, whereas genes involved in HSPC quiescence were repressed during IFNα treatment. Compared with hydroxyurea-treated controls, pegylated IFNα-treated patients had similar numbers of HSPCs, but increased numbers of hematopoietic progenitors as determined by colony formation assay, indicating an increase in myeloid proliferation/differentiation. These effects occurred regardless of JAK2 mutational status. Together, these data provide the first in vivo evidence that pegylated IFNα promotes cell division and differentiation of human HSPCs.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Hidroxiurea/administración & dosificación , Interferón-alfa/administración & dosificación , Leucemia Mielógena Crónica BCR-ABL Positiva , Policitemia Vera , Trombocitemia Esencial , Anciano , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Femenino , Células Madre Hematopoyéticas/patología , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Ratones , Persona de Mediana Edad , Policitemia Vera/tratamiento farmacológico , Policitemia Vera/genética , Policitemia Vera/metabolismo , Policitemia Vera/patología , Trombocitemia Esencial/tratamiento farmacológico , Trombocitemia Esencial/genética , Trombocitemia Esencial/metabolismo , Trombocitemia Esencial/patología , Factores de Tiempo
10.
Clin Cancer Res ; 18(2): 408-16, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22038994

RESUMEN

PURPOSE: Metastasis is responsible for the death of most cancer patients, yet few therapeutic agents are available which specifically target the molecular events that lead to metastasis. We recently showed that inactivating mutations in the tumor suppressor gene BAP1 are closely associated with loss of melanocytic differentiation in uveal melanoma (UM) and metastasis. The purpose of this study was to identify therapeutic agents that reverse the phenotypic effects of BAP1 loss in UM. EXPERIMENTAL DESIGN: In silico screens were done to identify therapeutic compounds predicted to differentiate UM cells using Gene Set Enrichment Analysis and Connectivity Map databases. Valproic acid (VPA), trichostatin A, LBH-589, and suberoylanilide hydroxamic acid were evaluated for their effects on UM cells using morphologic evaluation, MTS viability assays, bromodeoxyuridine incorporation, flow cytometry, clonogenic assays, gene expression profiling, histone acetylation and ubiquitination assays, and a murine xenograft tumorigenicity model. RESULTS: Histone deacetylase (HDAC) inhibitors induced morphologic differentiation, cell-cycle exit, and a shift to a differentiated, melanocytic gene expression profile in cultured UM cells. VPA inhibited the growth of UM tumors in vivo. CONCLUSIONS: These findings suggest that HDAC inhibitors may have therapeutic potential for inducing differentiation and prolonged dormancy of micrometastatic disease in UM.


Asunto(s)
Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias de la Úvea/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Quimioradioterapia Adyuvante , Simulación por Computador , Técnicas de Silenciamiento del Gen , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Indoles , Melanoma/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Biológicos , Micrometástasis de Neoplasia/prevención & control , Panobinostat , Carga Tumoral/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Neoplasias de la Úvea/patología , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Vorinostat , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Science ; 330(6009): 1410-3, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21051595

RESUMEN

Metastasis is a defining feature of malignant tumors and is the most common cause of cancer-related death, yet the genetics of metastasis are poorly understood. We used exome capture coupled with massively parallel sequencing to search for metastasis-related mutations in highly metastatic uveal melanomas of the eye. Inactivating somatic mutations were identified in the gene encoding BRCA1-associated protein 1 (BAP1) on chromosome 3p21.1 in 26 of 31 (84%) metastasizing tumors, including 15 mutations causing premature protein termination and 5 affecting its ubiquitin carboxyl-terminal hydrolase domain. One tumor harbored a frameshift mutation that was germline in origin, thus representing a susceptibility allele. These findings implicate loss of BAP1 in uveal melanoma metastasis and suggest that the BAP1 pathway may be a valuable therapeutic target.


Asunto(s)
Melanoma/genética , Melanoma/secundario , Mutación , Metástasis de la Neoplasia , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Neoplasias de la Úvea/genética , Línea Celular Tumoral , Deleción Cromosómica , Cromosomas Humanos Par 3/genética , Mutación del Sistema de Lectura , Mutación de Línea Germinal , Humanos , Mutación Missense , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Análisis de Secuencia de ADN , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/química , Ubiquitina Tiolesterasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA