Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Circ Res ; 130(3): 366-383, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-34986653

RESUMEN

BACKGROUND: The chromatin-remodeling enzyme BRG1 (brahma-related gene 1) regulates gene expression in a variety of rapidly differentiating cells during embryonic development. However, the critical genes that BRG1 regulates during lymphatic vascular development are unknown. METHODS: We used genetic and imaging techniques to define the role of BRG1 in murine embryonic lymphatic development, although this approach inadvertently expanded our study to multiple interacting cell types. RESULTS: We found that omental macrophages fine-tune an unexpected developmental process by which erythrocytes escaping from naturally discontinuous omental blood vessels are collected by nearby lymphatic vessels. Our data indicate that circulating fibrin(ogen) leaking from gaps in omental blood vessels can trigger inflammasome-mediated IL-1ß (interleukin-1ß) production and secretion from nearby macrophages. IL-1ß destabilizes adherens junctions in omental blood and lymphatic vessels, contributing to both extravasation of erythrocytes and their uptake by lymphatics. BRG1 regulates IL-1ß production in omental macrophages by transcriptionally suppressing the inflammasome trigger RIPK3 (receptor interacting protein kinase 3). CONCLUSIONS: Genetic deletion of Brg1 in embryonic macrophages leads to excessive IL-1ß production, erythrocyte leakage from blood vessels, and blood-filled lymphatics in the developing omentum. Altogether, these results highlight a novel context for epigenetically regulated crosstalk between macrophages, blood vessels, and lymphatics.


Asunto(s)
Vasos Sanguíneos/metabolismo , ADN Helicasas/metabolismo , Interleucina-1beta/metabolismo , Vasos Linfáticos/metabolismo , Proteínas Nucleares/metabolismo , Epiplón/metabolismo , Factores de Transcripción/metabolismo , Uniones Adherentes/metabolismo , Animales , Vasos Sanguíneos/embriología , ADN Helicasas/genética , Eritrocitos/metabolismo , Inflamasomas/metabolismo , Vasos Linfáticos/embriología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares/genética , Epiplón/irrigación sanguínea , Epiplón/embriología , Factores de Transcripción/genética
2.
PLoS One ; 16(6): e0253519, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34153072

RESUMEN

The receptor-interacting protein kinase 3 (RIPK3) is a multi-functional protein best known for facilitating cellular necroptosis and inflammation. Recent evidence from our lab indicates that RIPK3 expression must be tightly regulated in endothelial cells to promote angiogenesis, to maintain vascular integrity during embryogenesis, and to provide protection from postnatal atherosclerosis. RIPK3 activity and stability are regulated by post-translational modifications and caspase-dependent cleavage. However, less is known about the transcriptional regulation of Ripk3. Here we utilized an unbiased CRISPR-based technology called genomic locus proteomics (GLoPro) to screen transcription factors and coregulatory proteins associated with the Ripk3 locus in a murine endothelial cell line. We found that 41 nuclear proteins are specifically enriched at the Ripk3 locus, including the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway components NFκB1 and IKBKG. We further verified that NFκB1 and IKBKG directly bind the Ripk3 promoter and prevent TNFα-induced Ripk3 transcription in cultured human primary endothelial cells. Moreover, NFκB1 prevents RIPK3-mediated death of primary endothelial cells. These data provide new insights into NF-κB signaling and Ripk3 transcriptional regulation in endothelial cells.


Asunto(s)
Células Endoteliales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Subunidad p50 de NF-kappa B/genética , Proteómica/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Factores de Transcripción/genética
3.
Cell Death Differ ; 27(2): 618-631, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31235857

RESUMEN

Physiological hypoxia can trigger transcriptional events that influence many developmental processes during mammalian embryogenesis. One way that hypoxia affects transcription is by engaging chromatin-remodeling complexes. We now report that chromodomain helicase DNA binding protein 4 (CHD4), an enzyme belonging to the nucleosome remodeling and deacetylase (NuRD) chromatin-remodeling complex, is required for transcriptional repression of the receptor-interacting protein kinase 3 (Ripk3)-a critical executor of the necroptosis cell death program-in hypoxic murine embryonic endothelial cells. Genetic deletion of Chd4 in murine embryonic endothelial cells in vivo results in upregulation of Ripk3 transcripts and protein prior to vascular rupture and lethality at midgestation, and concomitant deletion of Ripk3 partially rescues these phenotypes. In addition, CHD4 binds to and prevents acetylation of the Ripk3 promoter in cultured endothelial cells grown under hypoxic conditions to prevent excessive Ripk3 transcription. These data demonstrate that excessive RIPK3 is detrimental to embryonic vascular integrity and indicate that CHD4 suppresses Ripk3 transcription when the embryonic environment is particularly hypoxic prior to the establishment of fetal-placental circulation at midgestation. Altogether, this research provides new insights into regulators of Ripk3 transcription and encourages future studies into the mechanism by which excessive RIPK3 damages embryonic blood vessels.


Asunto(s)
Cromatina/metabolismo , ADN Helicasas/metabolismo , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Hipoxia de la Célula , Células Cultivadas , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
4.
Methods Mol Biol ; 1846: 335-344, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30242771

RESUMEN

Chromatin immunoprecipitation (ChIP) is an antibody-based method used to identify protein-DNA interactions and sites of protein modifications to chromatin in living cells. ChIP is a powerful method for identifying genomic sites at which epigenetic changes occur in cell types of interest because many antibodies have been developed that recognize specific epigenetic modifications of histone tails. This chapter provides detailed ChIP and subsequent polymerase chain reaction (ChIP-PCR) protocols for use in cultured endothelial cells. These protocols will allow investigators to make consistent and quantitative discoveries about epigenetic changes that occur in endothelial cells at specific genomic sites under varying treatment conditions.


Asunto(s)
Células Endoteliales/metabolismo , Epigénesis Genética , Perfilación de la Expresión Génica , Cromatina , Inmunoprecipitación de Cromatina , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
5.
Arterioscler Thromb Vasc Biol ; 37(9): 1674-1682, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28729363

RESUMEN

OBJECTIVE: The chromatin remodeling enzyme BRG1 (brahma-related gene 1) transcriptionally regulates target genes important for early blood vessel development and primitive hematopoiesis. However, because Brg1 deletion in vascular progenitor cells results in lethal anemia by embryonic day 10.5 (E10.5), roles for BRG1 in embryonic vascular development after midgestation are unknown. In this study, we sought to determine whether endothelial cell BRG1 regulates genes important for vascular development or maintenance later in embryonic development. APPROACH AND RESULTS: Using mice with temporally inducible deletion of endothelial BRG1 (Brg1fl/fl;Cdh5(PAC)-CreERT2 ), we found that Brg1 excision between E9.5 and 11.5 results in capillary dilation and lethal hemorrhage by E14.5. This phenotype strongly resembles that seen when the SRF (serum response factor) transcription factor is deleted from embryonic endothelial cells. Although expression of Srf and several of its known endothelial cell target genes are downregulated in BRG1-depleted endothelial cells, we did not detect binding of BRG1 at these gene promoters, indicating that they are not direct BRG1 target genes. Instead, we found that BRG1 binds to the promoters of the SRF cofactors Mrtfa and Mrtfb (myocardin-related transcription factors A and B) in endothelial cells, and these genes are downregulated in Brg1-deficient endothelial cells. CONCLUSIONS: BRG1 promotes transcription of endothelial Mrtfa and Mrtfb, which elevates expression of SRF and SRF target genes that establish embryonic capillary integrity. These data highlight a new and temporally specific role for BRG1 in embryonic vasculature and provide novel information about epigenetic regulation of Mrtf expression and SRF signaling in developing blood vessels.


Asunto(s)
Capilares/metabolismo , ADN Helicasas/metabolismo , Células Endoteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neovascularización Fisiológica , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Antígenos CD/genética , Sitios de Unión , Cadherinas/genética , Capilares/embriología , Línea Celular , ADN Helicasas/deficiencia , ADN Helicasas/genética , Epigénesis Genética , Genotipo , Edad Gestacional , Integrasas/genética , Ratones Noqueados , Morfogénesis , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Fenotipo , Regiones Promotoras Genéticas , Interferencia de ARN , Factor de Respuesta Sérica/genética , Factor de Respuesta Sérica/metabolismo , Transducción de Señal , Transactivadores/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transfección
6.
mBio ; 6(3): e00462, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26106078

RESUMEN

UNLABELLED: Although it is established that oxygen availability regulates cellular metabolism and growth, little is known regarding how intracellular pathogens use host factors to grow at physiological oxygen levels. Therefore, large-scale human small interfering RNA screening was performed to identify host genes important for growth of the intracellular protozoan parasite Toxoplasma gondii at tissue oxygen tensions. Among the genes identified by this screen, we focused on the hexokinase 2 (HK2) gene because its expression is regulated by hypoxia-inducible transcription factor 1 (HIF-1), which is important for Toxoplasma growth. Toxoplasma increases host HK2 transcript and protein levels in a HIF-1-dependent manner. In addition, parasite growth at 3% oxygen is restored in HIF-1-deficient cells transfected with HK2 expression plasmids. Both HIF-1 activation and HK2 expression were accompanied by increases in host glycolytic flux, suggesting that enhanced HK2 expression in parasite-infected cells is functionally significant. Parasite dependence on host HK2 and HIF-1 expression is not restricted to transformed cell lines, as both are required for parasite growth in nontransformed C2C12 myoblasts and HK2 is upregulated in vivo following infection. While HK2 is normally associated with the cytoplasmic face of the outer mitochondrial membrane at physiological O2 levels, HK2 relocalizes to the host cytoplasm following infection, a process that is required for parasite growth at 3% oxygen. Taken together, our findings show that HIF-1-dependent expression and relocalization of HK2 represent a novel mechanism by which Toxoplasma establishes its replicative niche at tissue oxygen tensions. IMPORTANCE: Little is known regarding how the host cell contributes to the survival of the intracellular parasite Toxoplasma gondii at oxygen levels that mimic those found in tissues. Our previous work showed that Toxoplasma activates the expression of an oxygen-regulated transcription factor that is required for growth. Here, we report that Toxoplasma regulates the abundance and activity of a key host metabolic enzyme, hexokinase 2, by activating HIF-1 and by promoting dissociation of hexokinase 2 from the mitochondrial membrane. Collectively, our data reveal HIF-1/hexokinase 2 as a novel target for an intracellular pathogen that acts by reprograming the host cell's metabolism to create an environment conducive for parasite replication at physiological oxygen levels.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Hexoquinasa/metabolismo , Interacciones Huésped-Patógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Toxoplasma/fisiología , Animales , Pruebas Genéticas , Glucólisis , Células HeLa , Humanos , Análisis de Flujos Metabólicos , Ratones Endogámicos C57BL , Monocitos/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Toxoplasmosis Animal/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...