Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 8(1): 2833, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29434282

RESUMEN

In adult mammals, hematopoietic stem cells (HSCs) reside in the bone marrow and are in part regulated by the bone marrow microenvironment, called the stem cell niche. We have previously identified the bone marrow morphogen osteopontin (OPN), which is abundantly present in the bone marrow extracellular matrix, as a negative regulator of the size of the HSC pool under physiological conditions. Here, we study the impact of OPN on HSC function during aging using an OPN-knockout mouse model. We show that during aging OPN deficiency is associated with an increase in lymphocytes and a decline in erythrocytes in peripheral blood. In a bone marrow transplantation setting, aged OPN-deficient stem cells show reduced reconstitution ability likely due to insufficient differentiation of HSCs into more mature cells. In serial bone marrow transplantation, aged OPN-/- bone marrow cells fail to adequately reconstitute red blood cells and platelets, resulting in severe anemia and thrombocytopenia as well as premature deaths of recipient mice. Thus, OPN has different effects on HSCs in aged and young animals and is particularly important to maintain stem cell function in aging mice.


Asunto(s)
Envejecimiento/genética , Anemia/genética , Células Madre Hematopoyéticas/citología , Osteopontina/genética , Trombocitopenia/genética , Envejecimiento/sangre , Envejecimiento/metabolismo , Anemia/sangre , Anemia/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Eritrocitos/metabolismo , Técnicas de Inactivación de Genes , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteopontina/metabolismo , Nicho de Células Madre , Trombocitopenia/sangre , Trombocitopenia/metabolismo
2.
Sci Rep ; 7: 41427, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-28128288

RESUMEN

Activating mutations leading to ligand-independent signaling of the stem cell factor receptor KIT are associated with several hematopoietic malignancies. One of the most common alterations is the D816V mutation. In this study, we characterized mice, which conditionally express the humanized KITD816V receptor in the adult hematopoietic system to determine the pathological consequences of unrestrained KIT signaling during blood cell development. We found that KITD816V mutant animals acquired a myeloproliferative neoplasm similar to polycythemia vera, marked by a massive increase in red blood cells and severe splenomegaly caused by excessive extramedullary erythropoiesis. Moreover, we found mobilization of stem cells from bone marrow to the spleen. Splenectomy prior to KITD816V induction prevented expansion of red blood cells, but rapidly lead to a state of aplastic anemia and bone marrow fibrosis, reminiscent of post polycythemic myeloid metaplasia, the spent phase of polycythemia vera. Our results show that the extramedullary hematopoietic niche microenvironment significantly influences disease outcome in KITD816V mutant mice, turning this model a valuable tool for studying the interplay between functionally abnormal hematopoietic cells and their microenvironment during development of polycythemia vera-like disease and myelofibrosis.


Asunto(s)
Neoplasias de la Médula Ósea/genética , Neoplasias de la Médula Ósea/patología , Transformación Celular Neoplásica/patología , Proteínas Proto-Oncogénicas c-kit/genética , Bazo/patología , Microambiente Tumoral , Animales , Células de la Médula Ósea/patología , Neoplasias de la Médula Ósea/sangre , Proliferación Celular , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Eritrocitos/patología , Fibrosis , Factor de Transcripción GATA2/metabolismo , Regulación Neoplásica de la Expresión Génica , Hematopoyesis , Hematopoyesis Extramedular , Células Madre Hematopoyéticas/metabolismo , Ratones Endogámicos C57BL , Fenotipo , Policitemia Vera/genética , Policitemia Vera/patología , Transducción de Señal , Bazo/cirugía , Esplenomegalia/patología
3.
Blood ; 129(4): 460-472, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27683414

RESUMEN

Epithelial-to-mesenchymal-transition (EMT) is critical for normal embryogenesis and effective postnatal wound healing, but is also associated with cancer metastasis. SNAIL, ZEB, and TWIST families of transcription factors are key modulators of the EMT process, but their precise roles in adult hematopoietic development and homeostasis remain unclear. Here we report that genetic inactivation of Zeb2 results in increased frequency of stem and progenitor subpopulations within the bone marrow (BM) and spleen and that these changes accompany differentiation defects in multiple hematopoietic cell lineages. We found no evidence that Zeb2 is critical for hematopoietic stem cell self-renewal capacity. However, knocking out Zeb2 in the BM promoted a phenotype with several features that resemble human myeloproliferative disorders, such as BM fibrosis, splenomegaly, and extramedullary hematopoiesis. Global gene expression and intracellular signal transduction analysis revealed perturbations in specific cytokine and cytokine receptor-related signaling pathways following Zeb2 loss, especially the JAK-STAT and extracellular signal-regulated kinase pathways. Moreover, we detected some previously unknown mutations within the human Zeb2 gene (ZFX1B locus) from patients with myeloid disease. Collectively, our results demonstrate that Zeb2 controls adult hematopoietic differentiation and lineage fidelity through widespread modulation of dominant signaling pathways that may contribute to blood disorders.


Asunto(s)
Citocinas/genética , Transición Epitelial-Mesenquimal/genética , Hematopoyesis Extramedular/genética , Proteínas de Homeodominio/genética , Mielofibrosis Primaria/genética , Proteínas Represoras/genética , Esplenomegalia/genética , Adulto , Animales , Secuencia de Bases , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular , Linaje de la Célula/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Humanos , Quinasas Janus/genética , Quinasas Janus/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mutación , Mielofibrosis Primaria/metabolismo , Mielofibrosis Primaria/patología , Proteínas Represoras/deficiencia , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Bazo/metabolismo , Bazo/patología , Esplenomegalia/metabolismo , Esplenomegalia/patología , Células Madre/metabolismo , Células Madre/patología , Transcripción Genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
4.
PLoS One ; 9(10): e109266, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25286245

RESUMEN

Specialized blood cells are generated through the entire life of an organism by differentiation of a small number of hematopoietic stem cells (HSC). There are strictly regulated mechanisms assuring a constant and controlled production of mature blood cells. Although such mechanisms are not completely understood, some factors regulating cell cycle and differentiation have been identified. We have previously shown that Caspase-3 is an important regulator of HSC homeostasis and cytokine responsiveness. p21cip1/waf1 is a known cell cycle regulator, however its role in stem cell homeostasis seems to be limited. Several reports indicate interactions between p21cip1/waf1 and Caspase-3 in a cell type dependent manner. Here we studied the impact of simultaneous depletion of both factors on HSC homeostasis. Depletion of both Caspase-3 and p21cip1/waf1 resulted in an even more pronounced increase in the frequency of hematopoietic stem and progenitor cells. In addition, simultaneous deletion of both genes revealed a further increase of cell proliferation compared to single knock-outs and WT control mice, while apoptosis or self-renewal ability were not affected in any of the genotypes. Upon transplantation, p21cip1/waf1-/- bone marrow did not reveal significant alterations in engraftment of lethally irradiated mice, while Caspase-3 deficient HSPC displayed a significant reduction of blood cell production. However, when both p21cip1/waf1 and Caspase-3 were eliminated this differentiation defect caused by Caspase-3 deficiency was abrogated.


Asunto(s)
Caspasa 3/deficiencia , Caspasa 3/genética , Diferenciación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Eliminación de Gen , Células Madre Hematopoyéticas/citología , Animales , Apoptosis/genética , Proliferación Celular/genética , Autorrenovación de las Células/genética , Femenino , Técnicas de Inactivación de Genes , Trasplante de Células Madre Hematopoyéticas , Ratones , Transducción de Señal/genética
5.
Blood ; 117(21): 5620-30, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21355089

RESUMEN

Zeb2 (Sip1/Zfhx1b) is a member of the zinc-finger E-box-binding (ZEB) family of transcriptional repressors previously demonstrated to regulate epithelial-to-mesenchymal transition (EMT) processes during embryogenesis and tumor progression. We found high Zeb2 mRNA expression levels in HSCs and hematopoietic progenitor cells (HPCs), and examined Zeb2 function in hematopoiesis through a conditional deletion approach using the Tie2-Cre and Vav-iCre recombination mouse lines. Detailed cellular analysis demonstrated that Zeb2 is dispensable for hematopoietic cluster and HSC formation in the aorta-gonadomesonephros region of the embryo, but is essential for normal HSC/HPC differentiation. In addition, Zeb2-deficient HSCs/HPCs fail to properly colonize the fetal liver and/or bone marrow and show enhanced adhesive properties associated with increased ß1 integrin and Cxcr4 expression. Moreover, deletion of Zeb2 resulted in embryonic (Tie2-Cre) and perinatal (Vav-icre) lethality due to severe cephalic hemorrhaging and decreased levels of angiopoietin-1 and, subsequently, improper pericyte coverage of the cephalic vasculature. These results reveal essential roles for Zeb2 in embryonic hematopoiesis and are suggestive of a role for Zeb2 in hematopoietic-related pathologies in the adult.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Transición Epitelial-Mesenquimal , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/fisiología , Proteínas Represoras/fisiología , Animales , Cadherinas/metabolismo , Movimiento Celular , Femenino , Citometría de Flujo , Genes Letales , Células Madre Hematopoyéticas/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc , Dedos de Zinc
6.
Neuron ; 66(5): 724-38, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20547130

RESUMEN

Precise apposition of presynaptic and postsynaptic domains is a fundamental property of all neuronal circuits. Experiments in vitro suggest that Neuroligins and Neurexins function as key regulatory proteins in this process. In a genetic screen, we recovered several mutant alleles of Drosophila neuroligin 1 (dnlg1) that cause a severe reduction in bouton numbers at neuromuscular junctions (NMJs). In accord with reduced synapse numbers, these NMJs show reduced synaptic transmission. Moreover, lack of postsynaptic DNlg1 leads to deficits in the accumulation of postsynaptic glutamate receptors, scaffold proteins, and subsynaptic membranes, while increased DNlg1 triggers ectopic postsynaptic differentiation via its cytoplasmic domain. DNlg1 forms discrete clusters adjacent to postsynaptic densities. Formation of these clusters depends on presynaptic Drosophila Neurexin (DNrx). However, DNrx binding is not an absolute requirement for DNlg1 function. Instead, other signaling components are likely involved in DNlg1 transsynaptic functions, with essential interactions organized by the DNlg1 extracellular domain but also by the cytoplasmic domain.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Diferenciación Celular/fisiología , Proteínas de Drosophila/fisiología , Unión Neuromuscular/fisiología , Receptores de Glutamato/fisiología , Sinapsis/fisiología , Alelos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Diferenciación Celular/genética , Drosophila , Proteínas de Drosophila/genética , Mutación , Unión Neuromuscular/genética , Unión Neuromuscular/ultraestructura , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Receptores de Glutamato/genética , Receptores de Glutamato/ultraestructura , Sinapsis/genética , Sinapsis/ultraestructura , Transmisión Sináptica/genética , Transmisión Sináptica/fisiología
7.
Ann N Y Acad Sci ; 1176: 219-27, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19796250

RESUMEN

A decade of research on human embryonic stem cells (ESC) has paved the way for the discovery of alternative approaches to generating pluripotent stem cells. Combinatorial overexpression of a limited number of proteins linked to pluripotency in ESC was recently found to reprogram differentiated somatic cells back to a pluripotent state, enabling the derivation of isogenic (patient-specific) pluripotent stem cell lines. Current research is focusing on improving reprogramming protocols (e.g., circumventing the use of retroviral technology and oncoproteins), and on methods for differentiation into transplantable tissues of interest. In mouse ESC, we have previously shown that the embryonic morphogens BMP4 and Wnt3a direct blood formation via activation of Cdx and Hox genes. Ectopic expression of Cdx4 and HoxB4 enables the generation of mouse ESC-derived hematopoietic stem cells (HSC) capable of multilineage reconstitution of lethally irradiated adult mice. Here, we explore hematopoietic development from human induced pluripotent stem (iPS) cells generated in our laboratory. Our data show robust differentiation of iPS cells to mesoderm and to blood lineages, as shown by generation of CD34(+)CD45(+) cells, hematopoietic colony activity, and gene expression data, and suggest conservation of blood patterning pathways between mouse and human hematopoietic development.


Asunto(s)
Separación Celular/métodos , Hematopoyesis , Células Madre Hematopoyéticas/fisiología , Células Madre Pluripotentes/fisiología , Animales , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Células Madre Pluripotentes/citología
8.
Blood ; 113(17): 4049-51, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19218548

RESUMEN

Members of the caudal (cdx) family of homeobox proteins are essential regulators of embryonic blood development in zebrafish. Previously, we reported that the murine homologues (Cdx1, Cdx2, and Cdx4) affect formation and differentiation of embryonic stem cell (ESC)-derived hematopoietic progenitor cells. Consistent with the notion that embryonic pathways can reactivate during adult oncogenesis, recent studies suggest involvement of CDX2 in human acute myeloid leukemia (AML). Here we study CDX2 in healthy and leukemic human lymphoid cells, and show that a majority of leukemic samples display various degrees of aberrant CDX2 expression. Analysis of a cohort of 37 childhood acute lymphoblastic leukemia (ALL) patients treated in our hospital reveals that high CDX2 expression levels at diagnosis correlate with persistence of minimal residual disease (MRD) during the course of treatment. Thus, CDX2 expression levels may serve as a marker for adverse prognosis in pediatric ALL.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Homeodominio/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Factor de Transcripción CDX2 , Estudios de Casos y Controles , Niño , Proteínas de Homeodominio/genética , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transcripción Genética/genética
9.
Cell Stem Cell ; 2(6): 584-94, 2008 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-18522851

RESUMEN

Limited responsiveness to inflammatory cytokines is a feature of adult hematopoietic stem cells and contributes to the relative quiescence and durability of the stem cell population in vivo. Here we report that the executioner Caspase, Caspase-3, unexpectedly participates in that process. Mice deficient in Caspase-3 had increased numbers of immunophenotypic long-term repopulating stem cells in association with multiple functional changes, most prominently cell cycling. Though these changes were cell autonomous, they reflected altered activation by exogenous signals. Caspase-3(-/-) cells exhibited cell type-specific changes in phosphorylated members of the Ras-Raf-MEK-ERK pathway in response to specific cytokines, while notably, members of other pathways, such as pSTAT3, pSTAT5, pAKT, pp38 MAPK, pSmad2, and pSmad3, were unaffected. Caspase-3 contributes to stem cell quiescence, dampening specific signaling events and thereby cell responsiveness to microenvironmental stimuli.


Asunto(s)
Caspasa 3/biosíntesis , Células Madre Hematopoyéticas/citología , Animales , Diferenciación Celular/genética , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/enzimología , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Ratones Noqueados , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal/genética , Quinasas raf/metabolismo , Proteínas ras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...