Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 125(12): 1957-67, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25538045

RESUMEN

Sp1 and Sp3 belong to the specificity proteins (Sp)/Krüppel-like transcription factor family. They are closely related, ubiquitously expressed, and recognize G-rich DNA motifs. They are thought to regulate generic processes such as cell-cycle and growth control, metabolic pathways, and apoptosis. Ablation of Sp1 or Sp3 in mice is lethal, and combined haploinsufficiency results in hematopoietic defects during the fetal stages. Here, we show that in adult mice, conditional pan-hematopoietic (Mx1-Cre) ablation of either Sp1 or Sp3 has minimal impact on hematopoiesis, whereas the simultaneous loss of Sp1 and Sp3 results in severe macrothrombocytopenia. This occurs in a cell-autonomous manner as shown by megakaryocyte-specific (Pf4-Cre) double-knockout mice. We employed flow cytometry, cell culture, and electron microscopy and show that although megakaryocyte numbers are normal in bone marrow and spleen, they display a less compact demarcation membrane system and a striking inability to form proplatelets. Through megakaryocyte transcriptomics and platelet proteomics, we identified several cytoskeleton-related proteins and downstream effector kinases, including Mylk, that were downregulated upon Sp1/Sp3 depletion, providing an explanation for the observed defects in megakaryopoiesis. Supporting this notion, selective Mylk inhibition by ML7 affected proplatelet formation and stabilization and resulted in defective ITAM receptor-mediated platelet aggregation.


Asunto(s)
Plaquetas/citología , Megacariocitos/citología , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp3/genética , Animales , Azepinas/química , Plaquetas/metabolismo , Médula Ósea/metabolismo , Citometría de Flujo , Lectinas Tipo C/metabolismo , Ratones , Ratones Noqueados , Naftalenos/química , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteoma , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Bazo/metabolismo , Trombocitopenia/metabolismo , Factores de Transcripción/metabolismo
2.
Curr Opin Immunol ; 34: 68-74, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25726751

RESUMEN

Following infection, naïve CD4 T cells can differentiate into various functionally distinct effector and memory subsets, including T follicular helper (TFH) cells that orchestrate germinal center (GC) reactions necessary for high-affinity, pathogen-specific antibody responses. The origins and function of this cell type have been extensively examined in response to subunit immunization with model antigens. More recently, we are beginning to also appreciate the extent to which microbial infections shape the generation, function and maintenance of TFH cells. Here, we review recent advances and highlight additional knowledge gaps in our understanding of how microbial infections influence priming, differentiation, localization and activity of TFH cells following acute and chronic infections.


Asunto(s)
Infecciones/inmunología , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Diferenciación Celular , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , Memoria Inmunológica
3.
Cell Host Microbe ; 17(5): 628-41, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25891357

RESUMEN

The differentiation and protective capacity of Plasmodium-specific T cells are regulated by both positive and negative signals during malaria, but the molecular and cellular details remain poorly defined. Here we show that malaria patients and Plasmodium-infected rodents exhibit atypical expression of the co-stimulatory receptor OX40 on CD4 T cells and that therapeutic enhancement of OX40 signaling enhances helper CD4 T cell activity, humoral immunity, and parasite clearance in rodents. However, these beneficial effects of OX40 signaling are abrogated following coordinate blockade of PD-1 co-inhibitory pathways, which are also upregulated during malaria and associated with elevated parasitemia. Co-administration of biologics blocking PD-1 and promoting OX40 signaling induces excessive interferon-gamma that directly limits helper T cell-mediated support of humoral immunity and decreases parasite control. Our results show that targeting OX40 can enhance Plasmodium control and that crosstalk between co-inhibitory and co-stimulatory pathways in pathogen-specific CD4 T cells can impact pathogen clearance.


Asunto(s)
Diferenciación Celular , Inmunidad Humoral , Malaria/inmunología , Plasmodium/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptores OX40/metabolismo , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Regulación de la Expresión Génica , Humanos , Ratones
4.
Mol Cell Biol ; 33(19): 3879-92, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23897431

RESUMEN

Rad23a and Rad23b proteins are linked to nucleotide excision DNA repair (NER) via association with the DNA damage recognition protein xeroderma pigmentosum group C (XPC) are and known to be implicated in protein turnover by the 26S proteasome. Rad23b-null mice are NER proficient, likely due to the redundant function of the Rad23b paralogue, Rad23a. However, Rad23b-null midgestation embryos are anemic, and most embryos die before birth. Using an unbiased proteomics approach, we found that the majority of Rad23b-interacting partners are associated with the ubiquitin-proteasome system (UPS). We tested the requirement for Rad23b-dependent UPS activity in cellular proliferation and more specifically in the process of erythropoiesis. In cultured fibroblasts derived from embryos lacking Rad23b, proliferation rates were reduced. In fetal livers of Rad23b-null embryos, we observed reduced proliferation, accumulation of early erythroid progenitors, and a block during erythroid maturation. In primary wild-type (WT) erythroid cells, knockdown of Rad23b or chemical inhibition of the proteasome reduced survival and differentiation capability. Finally, the defects linked to Rad23b loss specifically affected fetal definitive erythropoiesis and stress erythropoiesis in adult mice. Together, these data indicate a previously unappreciated requirement for Rad23b and the UPS in regulation of proliferation in different cell types.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/genética , Eritropoyesis/genética , Complejo de la Endopetidasa Proteasomal/genética , Animales , Western Blotting , Diferenciación Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Citometría de Flujo , Hígado/citología , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA