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

Banco de datos
Tipo del documento
Publication year range
1.
J Mol Cell Cardiol ; 116: 57-68, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29408197

RESUMEN

Sphingosine-1-Phosphate (S1P) is a potent signaling lipid. The effects of S1P are mediated by the five S1P receptors (S1PR). In the endothelium S1PR1 is the predominant receptor and thus S1PR1 abundance limits S1P signaling. Recently, lncRNAs were identified as a novel class of molecules regulating gene expression. Interestingly, the lncRNA NONHSAT004848 (LISPR1, Long intergenic noncoding RNA antisense to S1PR1), is closely positioned to the S1P1 receptors gene and in part shares its promoter region. We hypothesize that LISPR1 controls endothelial S1PR1 expression and thus S1P-induced signaling in endothelial cells. In vitro transcription and translation as well as coding potential assessment showed that LISPR1 is indeed noncoding. LISPR1 was localized in both cytoplasm and nucleus and harbored a PolyA tail at the 3'end. In human umbilical vein endothelial cells, as well as human lung tissue, qRT-PCR and RNA-Seq revealed high expression of LISPR1. S1PR1 and LISPR1 were downregulated in human pulmonary diseases such as COPD. LISPR1 but also S1PR1 were induced by inflammation, shear stress and statins. Knockdown of LISPR1 attenuated endothelial S1P-induced migration and spheroid outgrowth of endothelial cells. LISPR1 knockdown decreased S1PR1 expression, which was paralleled by an increase of the binding of the transcriptional repressor ZNF354C to the S1PR1 promoter and a reduction of the recruitment of RNA Polymerase II to the S1PR1 5'end. This resulted in attenuated S1PR1 expression and attenuated S1P downstream signaling. Collectively, the disease relevant lncRNA LISPR1 acts as a novel regulatory unit important for S1PR1 expression and endothelial cell function.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Lisofosfolípidos/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Esfingosina/análogos & derivados , ADN/metabolismo , Regulación hacia Abajo/genética , Regulación de la Expresión Génica , Humanos , Pulmón/metabolismo , Neovascularización Fisiológica , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Proteínas Represoras/metabolismo , Esfingosina/metabolismo , Transcripción Genética
2.
Arterioscler Thromb Vasc Biol ; 35(7): 1645-52, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26023081

RESUMEN

OBJECTIVE: Altering endothelial biology through epigenetic modifiers is an attractive novel concept, which is, however, just in its beginnings. We therefore set out to identify chromatin modifiers important for endothelial gene expression and contributing to angiogenesis. APPROACH AND RESULTS: To identify chromatin modifying enzymes in endothelial cells, histone demethylases were screened by microarray and polymerase chain reaction. The histone 3 lysine 4 demethylase JARID1B was identified as a highly expressed enzyme at the mRNA and protein levels. Knockdown of JARID1B by shRNA in human umbilical vein endothelial cells attenuated cell migration, angiogenic sprouting, and tube formation. Similarly, pharmacological inhibition and overexpression of a catalytic inactive JARID1B mutant reduced the angiogenic capacity of human umbilical vein endothelial cells. To identify the in vivo relevance of JARID1B in the vascular system, Jarid1b knockout mice were studied. As global knockout results in increased mortality and developmental defects, tamoxifen-inducible and endothelial-specific knockout mice were generated. Acute knockout of Jarid1b attenuated retinal angiogenesis and endothelial sprout outgrowth from aortic segments. To identify the underlying mechanism, a microarray experiment was performed, which led to the identification of the antiangiogenic transcription factor HOXA5 to be suppressed by JARID1B. Importantly, downregulation or inhibition of JARID1B, but not of JARID1A and JARID1C, induced HOXA5 expression in human umbilical vein endothelial cells. Consistently, chromatin immunoprecipitation revealed that JARID1B occupies and reduces the histone 3 lysine 4 methylation levels at the HOXA5 promoter, demonstrating a direct function of JARID1B in endothelial HOXA5 gene regulation. CONCLUSIONS: JARID1B, by suppressing HOXA5, maintains the endothelial angiogenic capacity in a demethylase-dependent manner.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Epigénesis Genética , Proteínas de Homeodominio/genética , Histona Demetilasas con Dominio de Jumonji/fisiología , Neovascularización Fisiológica/genética , Proteínas Nucleares/fisiología , Fosfoproteínas/genética , Animales , Células Cultivadas , Células Endoteliales/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Ratones Noqueados , Fosfoproteínas/fisiología , Factores de Transcripción , Transcripción Genética , Venas Umbilicales
3.
Basic Res Cardiol ; 109(6): 439, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25204797

RESUMEN

Endothelial cells are important elements in the vascular response to danger-associated molecules signaling through toll-like receptors (TLRs). Flotillin-1 and -2 are markers of membrane rafts but their true endothelial function is unknown. We hypothesized that flotillins are required for TLR signaling in human umbilical vein endothelial cells (HUVECs). Knockdown of flotillin-1 by shRNA decreased the TLR3-mediated poly-I:C-induced but not the TLR4-mediated LPS-induced inflammatory activation of HUVEC. As TLR3 but not TLR4 signals through the endosomal compartment, flotillin-1 might be involved in the transport of poly-I:C to its receptor. Consistently, uptake of poly-I:C was attenuated by flotillin-1 knockdown and probably involved the scavenger receptor SCARA4 as revealed by knockdown of this receptor. To determine the underlying mechanism, SILAC proteomics was performed. Down-regulation of flotillin-1 led to a reduction of the structural caveolae proteins caveolin-1, cavin-1 and -2, suggesting a role of flotillin-1 in caveolae formation. Flotillin-1 and caveolin-1 colocalized within the cell, and knockdown of flotillin-1 decreased caveolin-1 expression in an endoplasmic reticulum stress-dependent manner. Importantly, downregulation of caveolin-1 also attenuated TLR3-induced signaling. To demonstrate the importance of this finding, cell adhesion was studied. Flotillin-1 shRNA attenuated the poly-I:C-mediated induction of the adhesion molecules VCAM-1 and ICAM-1. As a consequence, the poly-I:C-induced adhesion of peripheral blood mononuclear cells onto HUVECs was significantly attenuated by flotillin-1 shRNA. Collectively, these data suggest that interaction between flotillin-1 and caveolin-1 may facilitate the transport of TLR3-ligands to its intracellular receptor and enables inflammatory TLR3 signaling.


Asunto(s)
Células Endoteliales/fisiología , Proteínas de la Membrana/fisiología , Transducción de Señal/fisiología , Receptor Toll-Like 3/fisiología , Humanos
4.
FEBS Lett ; 593(5): 487-498, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30758047

RESUMEN

Histone3-lysine9 (H3K9) residues not only control gene expression, but also contribute to RNA splicing. Here, the H3K9 histone demethylase PHF8 was investigated in endothelial cells for its involvement in alternative splicing. An angiogenic sprouting assay shows the importance of PHF8 for endothelial cells. Immunoprecipitation reveals that PHF8 interacts with U1 spliceosomal proteins, such as SRPK1 and snRNP70. We identify the histocompatibility antigen HLA-G as a target of PHF8. The inclusion of HLA-G intron 4, with concomitant RNA Polymerase II accumulation at this intron is controlled by PHF8 and H3K9. Soluble HLA-G is generated after PHF8 knockdown, which leads to reduced T-cell proliferation. Collectively, PHF8 knockdown generates the immunosuppressive alternative splice product soluble HLA-G, which is secreted by endothelial cells to elicit a potential inhibitory effect on inflammation.


Asunto(s)
Empalme Alternativo , Antígenos HLA-G/genética , Histona Demetilasas/metabolismo , Factores de Transcripción/metabolismo , Proliferación Celular , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Técnicas de Silenciamiento del Gen , Histona Demetilasas/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Intrones , Unión Proteica , ARN Polimerasa II/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Linfocitos T/citología , Factores de Transcripción/genética
5.
Nat Commun ; 9(1): 2292, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895827

RESUMEN

Oxidized phospholipids (oxPAPC) induce endothelial dysfunction and atherosclerosis. Here we show that oxPAPC induce a gene network regulating serine-glycine metabolism with the mitochondrial methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) as a causal regulator using integrative network modeling and Bayesian network analysis in human aortic endothelial cells. The cluster is activated in human plaque material and by atherogenic lipoproteins isolated from plasma of patients with coronary artery disease (CAD). Single nucleotide polymorphisms (SNPs) within the MTHFD2-controlled cluster associate with CAD. The MTHFD2-controlled cluster redirects metabolism to glycine synthesis to replenish purine nucleotides. Since endothelial cells secrete purines in response to oxPAPC, the MTHFD2-controlled response maintains endothelial ATP. Accordingly, MTHFD2-dependent glycine synthesis is a prerequisite for angiogenesis. Thus, we propose that endothelial cells undergo MTHFD2-mediated reprogramming toward serine-glycine and mitochondrial one-carbon metabolism to compensate for the loss of ATP in response to oxPAPC during atherosclerosis.


Asunto(s)
Aminoácidos/metabolismo , Aminohidrolasas/metabolismo , Aterosclerosis/metabolismo , Células Endoteliales/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Enzimas Multifuncionales/metabolismo , Fosfolípidos/química , Animales , Aorta/citología , Teorema de Bayes , Enfermedades Cardiovasculares/metabolismo , Regulación de la Expresión Génica , Técnicas Genéticas , Glicina/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Familia de Multigenes , Neovascularización Patológica , Nucleótidos/química , Oxígeno/química , Probabilidad , Purinas/química , ARN Interferente Pequeño/metabolismo , Pez Cebra
6.
PLoS One ; 11(1): e0146645, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26751588

RESUMEN

Epigenetic marks critically control gene expression and thus the cellular activity state. The functions of many epigenetic modifiers in the vascular system have not yet been studied. We screened for histone modifiers in endothelial cells and observed a fairly high expression of the histone plant homeodomain finger protein 8 (PHF8). Given its high expression, we hypothesize that this histone demethylase is important for endothelial cell function. Overexpression of PHF8 catalyzed the removal of methyl-groups from histone 3 lysine 9 (H3K9) and H4K20, whereas knockdown of the enzyme increased H3K9 methylation. Knockdown of PHF8 by RNAi also attenuated endothelial proliferation and survival. As a functional readout endothelial migration and tube formation was studied. PHF8 siRNA attenuated the capacity for migration and developing of capillary-like structures. Given the impact of PHF8 on cell cycle genes, endothelial E2F transcription factors were screened, which led to the identification of the gene repressor E2F4 to be controlled by PHF8. Importantly, PHF8 maintains E2F4 but not E2F1 expression in endothelial cells. Consistently, chromatin immunoprecipitation revealed that PHF8 reduces the H3K9me2 level at the E2F4 transcriptional start site, demonstrating a direct function of PHF8 in endothelial E2F4 gene regulation. Conclusion: PHF8 by controlling E2F4 expression maintains endothelial function.


Asunto(s)
Movimiento Celular , Factor de Transcripción E2F4/metabolismo , Células Endoteliales/citología , Histona Demetilasas/metabolismo , Factores de Transcripción/metabolismo , Apoptosis , Catálisis , Línea Celular , Proliferación Celular , Supervivencia Celular , Metilación de ADN , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Histonas/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Microcirculación , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Sitio de Iniciación de la Transcripción
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda