Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Clin Exp Med ; 7(9): 2541-9, 2014.
Article in English | MEDLINE | ID: mdl-25356107

ABSTRACT

Endothelium dysfunction has been understood primarily in terms of abnormal vasomotor function, which plays an important role in the pathogenesis of diabetes and chronic diabetic complications. However, it has not been fully studied that the endothelium may regulate metabolism itself. The response gene to complement 32 (RGC-32) has be considered as an angiogenic inhibitor in the context of endothelial cells. We found that RGC-32 was induced by high fat diet in vivo and by glucose or insulin in endothelial cells, and then we set out to investigate the role of endothelial RGC-32 in metabolism. DNA array analysis and qPCR results showed that glutamine-fructose-6-phosphate aminotransferase [isomerizing] 1 (GFPT1), solute carrier family 2 (facilitated glucose transporter), member 12 (SLC2A12, GLUT12) and glucagon-like peptide 2 receptor (GLP2R) may be among possible glucose metabolism related downstream genes of RGC-32. Additionally, in the mice with endothelial specific over-expressed RGC-32, the disposal of carbohydrate was improved without changing insulin sensitivity when mice were faced with high fat diet challenges. Taken together, our findings suggest that RGC-32 in the endothelial cells regulates glucose metabolism related genes and subsequent helps to maintain the homeostasis of blood glucose.

2.
Int J Mol Sci ; 15(3): 3816-33, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24595242

ABSTRACT

Tumor necrosis factor α (TNF-α) influences endothelial cell viability by altering the regulatory molecules involved in induction or suppression of apoptosis. However, the underlying mechanisms are still not completely understood. In this study, we demonstrated that A20 (also known as TNFAIP3, tumor necrosis factor α-induced protein 3, and an anti-apoptotic protein) regulates the inhibitor of apoptosis protein-2 (cIAP-2) expression upon TNF-α induction in endothelial cells. Inhibition of A20 expression by its siRNA resulted in attenuating expression of TNF-α-induced cIAP-2, yet not cIAP-1 or XIAP. A20-induced cIAP-2 expression can be blocked by the inhibition of phosphatidyl inositol-3 kinase (PI3-K), but not nuclear factor (NF)-κB, while concomitantly increasing the number of endothelial apoptotic cells and caspase 3 activation. Moreover, TNF-α-mediated induction of apoptosis was enhanced by A20 inhibition, which could be rescued by cIAP-2. Taken together, these results identify A20 as a cytoprotective factor involved in cIAP-2 inhibitory pathway of TNF-α-induced apoptosis. This is consistent with the idea that endothelial cell viability is dependent on interactions between inducers and suppressors of apoptosis, susceptible to modulation by TNF-α.


Subject(s)
Apoptosis/drug effects , DNA-Binding Proteins/metabolism , Endothelial Cells/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein , Blotting, Western , Caspase 3/metabolism , Cattle , Cells, Cultured , Chromones/pharmacology , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Morpholines/pharmacology , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitin-Protein Ligases
3.
PLoS One ; 9(2): e88143, 2014.
Article in English | MEDLINE | ID: mdl-24520353

ABSTRACT

Related Transcriptional Enhancer Factor-1 (RTEF-1) has been suggested to induce angiogenesis through regulating target genes. Whether RTEF-1 has a direct role in angiogenesis and what specific genes are involved in RTEF-1 driven angiogenisis have not been elucidated. We found that over-expressing RTEF-1 in Human dermal microvascular endothelial cells-1 (HMEC-1) significantly increased endothelial cell aggregation, growth and migration while the processes were inhibited by siRNA of RTEF-1. In addition, we observed that Endothelial differentiation gene-1 (Edg-1) expression was up-regulated by RTEF-1 at the transcriptional level. RTEF-1 could bind to Edg-1 promoter and subsequently induce its activity. Edg-1 siRNA significantly blocked RTEF-1-driven increases in endothelial cell aggregation in a Matrigel assay and retarded RTEF-1-induced endothelial cell growth and migration. Pertussis Toxin (PTX), a Gi/Go protein sensitive inhibitor, was found to inhibit RTEF-1 driven endothelial cell aggregation and migration. Our data demonstrates that Edg-1 is a potential target gene of RTEF-1 and is involved in RTEF-1-induced angiogenesis in endothelial cells. Gi/Go protein coupled receptor pathway plays a role in RTEF-1 driven angiogenesis in endothelial cells.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Muscle Proteins/metabolism , Neovascularization, Physiologic/genetics , Receptors, Lysosphingolipid/genetics , Transcription Factors/metabolism , Animals , Cell Aggregation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Mice , Neovascularization, Physiologic/drug effects , Pertussis Toxin/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine-1-Phosphate Receptors , TEA Domain Transcription Factors
4.
Am J Physiol Heart Circ Physiol ; 306(2): H233-42, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24213609

ABSTRACT

The related transcriptional enhancer factor-1 (RTEF-1) increases gene transcription of hypoxia-inducible factor 1α (HIF-1α) and enhances angiogenesis in endothelium. Both hypoxia and inflammatory factor TNF-α regulate gene expression of HIF-1α, but how RTEF-1 and TNF-α coordinately regulate HIF-1α gene transcription is unclear. Here, we found that RTEF-1 interacts with p65 subunit of NF-κB, a primary mediator of TNF-α. RTEF-1 increased HIF-1α promoter activity, whereas expression of p65 subunit inhibited the stimulatory effect. By contrast, knockdown of p65 markedly enhanced RTEF-1 stimulation on the HIF-1α promoter activity (7-fold). A physical interaction between RTEF-1 and p65 was confirmed by coimmunoprecipitation experiments in cells and glutathione S-transferase (GST)-pull-down assays. A computational analysis of RTEF-1 crystal structures revealed that a conserved surface of RTEF-1 potentially interacts with p65 via four amino acid residues located at T347, Y349, R351, and Y352. We performed site-directed mutagenesis and GST-pull-down assays and demonstrated that Tyr352 (Y352) in RTEF-1 is a key site for the formation of RTEF-1 and p65-NF-κB complex. An alanine mutation at Y352 of RTEF-1 disrupted the interaction of RTEF-1 with p65. Moreover, expression of RTEF-1 decreased TNF-α-induced HIF-1α promoter activity, IL-1ß, and IL-6 mRNA levels in cells; however, the effect of RTEF-1 was largely lost when Y352 was mutated to alanine. These results indicate that RTEF-1 interacts with p65-NF-κB through Y352 and that they antagonize each other for HIF-1α transcriptional activation, suggesting a novel mechanism by which RTEF-1 regulates gene expression, linking hypoxia to inflammation.


Subject(s)
DNA-Binding Proteins/metabolism , Molecular Docking Simulation , Muscle Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Binding Sites , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Sequence Data , Muscle Proteins/chemistry , Muscle Proteins/genetics , Mutagenesis, Site-Directed , Mutation, Missense , Promoter Regions, Genetic , Protein Binding , TEA Domain Transcription Factors , Transcription Factor RelA/chemistry , Transcription Factor RelA/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic
5.
Circ Res ; 111(8): 991-1001, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22843786

ABSTRACT

RATIONALE: Related transcriptional enhancer factor-1 (RTEF-1) plays an important role in endothelial cell function by regulating angiogenesis; however, the mechanism underlying the role of RTEF-1 in the endothelium in vivo is not well defined. OBJECTIVE: We investigated the biological functions of RTEF-1 by disrupting the gene that encodes it in mice endothelium -specific RTEF-1-deficient transgenic mice (RTEF-1(-/-)). METHODS AND RESULTS: RTEF-1(-/-) mice showed significantly increased blood glucose levels and insulin resistance, accompanied by decreased levels of insulin-like growth factor binding protein-1 (IGFBP-1) mRNA in the endothelium and decreased serum IGFBP-1 levels. Additionally, the RTEF-1(-/-) phenotype was exacerbated when the mice were fed a high-fat diet, which correlated with decreased IGFBP-1 levels. In contrast, vascular endothelial cadherin/RTEF-1-overexpressing(1) transgenic mice (VE-Cad/RTEF1) demonstrated improved glucose clearance and insulin sensitivity in response to a high-fat diet. Furthermore, we demonstrated that RTEF-1 upregulates IGFBP-1 through selective binding and promotion of transcription from the insulin response element site. Insulin prevented RTEF-1 expression and significantly inhibited IGFBP-1 transcription in endothelial cells in a dose-dependent fashion. CONCLUSIONS: To the best of our knowledge, this is the first report demonstrating that RTEF-1 stimulates promoter activity through an insulin response element and also mediates the effects of insulin on gene expression. These results show that RTEF-1-stimulated IGFBP-1 expression may be central to the mechanism by which RTEF-1 attenuates blood glucose levels. These findings provide the basis for novel insights into the transcriptional regulation of IGFBP-1 and contribute to our understanding of the role of vascular endothelial cells in metabolism.


Subject(s)
Blood Glucose/metabolism , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Muscle Proteins/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose/genetics , DNA-Binding Proteins/genetics , Endothelial Cells/cytology , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , HEK293 Cells , Hearing/physiology , Homeostasis/physiology , Humans , Insulin Resistance/physiology , Mice , Mice, Knockout , Muscle Proteins/genetics , Obesity/genetics , Obesity/metabolism , Promoter Regions, Genetic/physiology , RNA, Small Interfering/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics
6.
Arterioscler Thromb Vasc Biol ; 32(8): 1951-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22652601

ABSTRACT

OBJECTIVE: Capillary network formation represents a specialized endothelial cell function and is a prerequisite to establish a continuous vessel lumen. Formation of endothelial cell connections that form the vascular structure is regulated, at least in part, at the transcriptional level. We report here that related transcription enhancer factor-1 (RTEF-1) plays an important role in vascular structure formation. METHODS AND RESULTS: Knockdown of RTEF-1 by small interfering RNA or blockage of RTEF-1 function by the transcription enhancer activators domain decreased endothelial connections in a Matrigel assay, whereas overexpression of RTEF-1 in endothelial cells resulted in a significant increase in cell connections and aggregation. In a model of oxygen-induced retinopathy, endothelial-specific RTEF-1 overexpressing mice had enhanced angiogenic sprouting and vascular structure remodeling, resulting in the formation of a denser and more highly interconnected superficial capillary plexus. Mechanistic studies revealed that RTEF-1 induced the expression of functional gap junction proteins including connexin 43, connexin 40, and connexin 37. Blocking connexin 43 function inhibited RTEF-1-induced endothelial cell connections and aggregation. CONCLUSIONS: These findings provide novel insights into the transcriptional control of endothelial function in the coordination of cell-cell connections.


Subject(s)
Cell Communication , Connexins/physiology , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Endothelial Cells/physiology , Muscle Proteins/metabolism , Muscle Proteins/physiology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Cell Aggregation , Cells, Cultured , Connexin 43/genetics , DNA-Binding Proteins/genetics , Humans , Mice , Muscle Proteins/genetics , Neovascularization, Physiologic , TEA Domain Transcription Factors , Transcription Factors/genetics
7.
Cardiovasc Res ; 90(2): 325-34, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21169295

ABSTRACT

AIMS: Related transcription enhancer factor-1 (RTEF-1) has previously been demonstrated to play an important role in both endothelial cells and cardiomyocytes. However, the function of RTEF-1 in the communication between these two adjacent cell types has not been elucidated. METHODS AND RESULTS: We have found that endothelium-specific RTEF-1 transgenic mice (VE-Cad/RTEF-1) developed significant cardiac hypertrophy after transverse aortic constriction surgery, as evidenced by an increased ratio of heart weight to tibia length, enlarged cardiomyocyte size, thickened left ventricular wall and elevated expression of hypertrophic gene markers, with up-regulation of vascular endothelial growth factor B (VEGF-B). Additionally, VEGF-B was increased in endothelial cells from VE-Cad/RTEF-1 mice, as well as in endothelial cells with forced RTEF-1 expression (HMEC-1/RTEF-1), and coincidentally decreased when RTEF-1 was deficient in HMEC-1. Using chromatin immunoprecipitation and luciferase assays, we found that RTEF-1 increased VEGF-B promoter activity through a direct interaction. Hypertrophy-associated genes and protein synthesis were up-regulated in cardiomyocytes that were incubated with conditioned medium from HMEC-1/RTEF-1 and the endothelial cells of VE-Cad/RTEF-1 mice. This effect could be abrogated by treating the myocytes with VEGF-B small interfering RNA and extracellular signal-regulated kinase 1/2 inhibitor. CONCLUSION: Our data demonstrated that increased RTEF-1 in endothelial cells upregulates VEGF-B, which is able to stimulate hypertrophic genes in cardiomyocytes. These results suggest that the RTEF-1-driven increase of VEGF-B plays an important role in communication between the endothelium and myocardium.


Subject(s)
Cardiomegaly , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelium, Vascular/physiology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor B/genetics , Vascular Endothelial Growth Factor B/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Blood Pressure/physiology , Cadherins/genetics , Cadherins/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , MAP Kinase Signaling System/physiology , Mice , Mice, Transgenic , Myoblasts/cytology , Myocardium/metabolism , Myocardium/pathology , Phosphorylation/physiology , Rats , Rats, Sprague-Dawley , TEA Domain Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...