Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(7): e69979, 2013.
Article in English | MEDLINE | ID: mdl-23922881

ABSTRACT

BACKGROUND: The development of diabetic angiopathy is associated with profound vascular endothelial cells (VEC) dysfunction and apoptosis. Glycated low density lipoproteins (gly-LDL) continuously produced in the setting of diabetic patients play an important role in causing VEC dysfunction and apoptosis. However, the underlying molecular mechanism remains largely elusive. Protein L-isoaspartyl methyltransferase (PIMT) is a widely expressed protein repair enzyme by multiple cell types of arterial wall including VEC. Our previous proteomic studies showed that the expression of PIMT was significantly decreased in the aorta of diabetic rats as compared with control rats and treatment with grape seed procyanidin extracts significantly increased the PIMT expression in diabetic rats. We hypothesized that PIMT plays a critical role in gly-LDL induced VEC apoptosis; grape seed procyanidin B2 (GSPB2) protect against gly-LDL induced VEC apoptosis through PIMT regulation. METHODS AND RESULTS: HUVEC transfected negative control and PIMT siRNA were treated with or without GSPB2 (10 µmol/L) for 48 h. Moreover, HUVEC of PIMT overexpression were stimulated by gly-LDL (50 µg/ml) in the presence or absence of GSPB2 (10 µmol/L) for 48 h. Our results showed that gly-LDL downregulated PIMT expression and PIMT overexpression or GSPB2 significantly attenuated gly-LDL induced VEC apoptosis. PIMT siRNA increased VEC apoptosis with up-regulation of p53, cytochrome c release, caspase-9 and caspase-3 activation. Mechanistically, overexpression of PIMT or GSPB2 increased the phosphorylation of ERK1/2 and GSK3ß in the gly-LDL induced VEC. CONCLUSION: In summary, our study identified PIMT as a key player responsible for gly-LDL induced VEC apoptosis and GSPB2 protect against gly-LDL induced VEC apoptosis by PIMT up-regulation. Targeting PIMT including use of GSPB2 could be turned into clinical application in the fighting against diabetic vascular complications.


Subject(s)
Apoptosis/drug effects , Biflavonoids/pharmacology , Catechin/pharmacology , Grape Seed Extract/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/enzymology , Lipoproteins, LDL/pharmacology , Proanthocyanidins/pharmacology , Protective Agents/pharmacology , Protein D-Aspartate-L-Isoaspartate Methyltransferase/metabolism , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucose/pharmacology , Glycation End Products, Advanced , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Human Umbilical Vein Endothelial Cells/drug effects , Humans , In Situ Nick-End Labeling , Phosphorylation/drug effects , Plasmids/metabolism , RNA, Small Interfering/metabolism , Rats , Transduction, Genetic , Transfection , Tumor Suppressor Protein p53/metabolism
2.
Biochim Biophys Acta ; 1832(6): 805-16, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23474305

ABSTRACT

Diabetic nephropathy, as a severe microvascular complication of diabetic mellitus, has become the leading cause of end-stage renal diseases. However, no effective therapeutic strategy has been developed to prevent renal damage progression to end stage renal disease. Hence, the present study evaluated the protective effects of grape seed procyanidin B2 (GSPB2) and explored its molecular targets underlying diabetic nephropathy by a comprehensive quantitative proteomic analysis in db/db mice. Here, we found that oral administration of GSPB2 significantly attenuated the renal dysfunction and pathological changes in db/db mice. Proteome analysis by isobaric tags for relative and absolute quantification (iTRAQ) identified 53 down-regulated and 60 up-regulated proteins after treatment with GSPB2 in db/db mice. Western blot analysis confirmed that milk fat globule EGF-8 (MFG-E8) was significantly up-regulated in diabetic kidney. MFG-E8 silencing by transfection of MFG-E8 shRNA improved renal histological lesions by inhibiting phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2), Akt and glycogen synthase kinase-3beta (GSK-3ß) in kidneys of db/db mice. In contrast, over-expression of MFG-E8 by injection of recombinant MFG-E8 resulted in the opposite effects. GSPB2 treatment significantly decreased protein levels of MFG-E8, phospho-ERK1/2, phospho-Akt, and phospho-GSK-3ß in the kidneys of db/db mice. These findings yield insights into the pathogenesis of diabetic nephropathy, revealing MFG-E8 as a new therapeutic target and indicating GSPB2 as a prospective therapy by down-regulation of MFG-E8, along with ERK1/2, Akt and GSK-3ß signaling pathway.


Subject(s)
Antigens, Surface/biosynthesis , Biflavonoids/pharmacology , Catechin/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , MAP Kinase Signaling System/drug effects , Milk Proteins/biosynthesis , Proanthocyanidins/pharmacology , Up-Regulation/drug effects , Animals , Antigens, Surface/genetics , Biflavonoids/chemistry , Catechin/chemistry , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diabetic Nephropathies/prevention & control , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Grape Seed Extract/chemistry , Grape Seed Extract/pharmacokinetics , Kidney/metabolism , Kidney/pathology , MAP Kinase Signaling System/genetics , Male , Mice , Milk Proteins/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Proanthocyanidins/chemistry , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/genetics
3.
PLoS One ; 7(12): e52541, 2012.
Article in English | MEDLINE | ID: mdl-23285083

ABSTRACT

BACKGROUND: Atherosclerosis is one of the major complications of type 2 diabetic patients (T2DM), leading to morbidity and mortality. Grape seed procyanidin B2 (GSPB2) has demonstrated protective effect against atherosclerosis, which is believed to be, at least in part, a result of its antioxidative effects. The aim of this study is to identify the target protein of GSPB2 responsible for the protective effect against atherosclerosis in patients with DM. METHODS AND RESULTS: GSPB2 (30 mg/kg body weight/day) were administrated to db/db mice for 10 weeks. Proteomics of the aorta extracts by iTRAQ analysis was obtained from db/db mice. The results showed that expression of 557 proteins were either up- or down-regulated in the aorta of diabetic mice. Among those proteins, 139 proteins were normalized by GSPB2 to the levels comparable to those in control mice. Among the proteins regulated by GSPB2, the milk fat globule epidermal growth factor-8 (MFG-E8) was found to be increased in serum level in T2DM patients; the serum level of MFG-E8 was positively correlated with carotid-femoral pulse wave velocity (CF-PWV). Inhibition of MFG-E8 by RNA interference significantly suppressed whereas exogenous recombinant MFG-E8 administration exacerbated atherogenesis the db/db mice. To gain more insights into the mechanism of action of MFG-E8, we investigated the effects of MFG-E8 on the signal pathway involving the extracellular signal-regulated kinase (ERK) and monocyte chemoattractant protein-1 (MCP-1). Treatment with recombinant MFG-E8 led to increased whereas inhibition of MFG-E8 to decreased expression of MCP-1 and phosphorylation of ERK1/2. CONCLUSION: Our data suggests that MFG-E8 plays an important role in atherogenesis in diabetes through both ERK and MCP-1 signaling pathways. GSPB2, a well-studied antioxidant, significantly inhibited the arterial wall changes favoring atherogenesis in db/db mice by down-regulating MFG-E8 expression in aorta and its serum level. Measuring MFG-E8 serum level could be a useful clinical surrogate prognosticating atherogenesis in DM patients.


Subject(s)
Antigens, Surface/metabolism , Aorta/metabolism , Aorta/pathology , Biflavonoids/therapeutic use , Catechin/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Milk Proteins/metabolism , Proanthocyanidins/therapeutic use , Proteomics , Aged , Animals , Antigens, Surface/blood , Aorta/drug effects , Aorta/ultrastructure , Biflavonoids/pharmacology , Blood Glucose/drug effects , Body Weight/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Catechin/pharmacology , Cholesterol/blood , Computational Biology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fasting/blood , Glycation End Products, Advanced/blood , Grape Seed Extract/pharmacology , Grape Seed Extract/therapeutic use , Humans , Isotope Labeling , Male , Mice , Mice, Inbred C57BL , Milk Proteins/blood , Proanthocyanidins/pharmacology , Proteome/metabolism , RNA Interference/drug effects , Triglycerides/blood
4.
Arterioscler Thromb Vasc Biol ; 32(3): 745-55, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22199367

ABSTRACT

OBJECTIVE: Age-related aortic stiffness is an independent risk factor for cardiovascular diseases. Although oxidative stress is implicated in aortic stiffness, the underlying molecular mechanisms remain unelucidated. Here, we examined the source of oxidative stress in aging and its effect on smooth muscle cell (SMC) function and aortic compliance using mutant mouse models. METHODS AND RESULTS: Pulse wave velocity, determined using Doppler, increased with age in superoxide dismutase 2 (SOD2)+/- but not in wild-type, p47phox-/- and SOD1+/- mice. Echocardiography showed impaired cardiac function in these mice. Increased collagen I expression, impaired elastic lamellae integrity, and increased medial SMC apoptosis were observed in the aortic wall of aged SOD2+/- versus wild-type (16-month-old) mice. Aortic SMCs from aged SOD2+/- mice showed increased collagen I and decreased elastin expression, increased matrix metalloproteinase-2 expression and activity, and increased sensitivity to staurosporine-induced apoptosis versus aged wild-type and young (4-month-old) SOD2+/- mice. Smooth muscle α-actin levels were increased with age in SOD2+/- versus wild-type SMCs. Aged SOD2+/- SMCs had attenuated insulin-like growth factor-1-induced Akt and Forkhead box O3a phosphorylation and prolonged tumor necrosis factor-α-induced Jun N-terminal kinase 1 activation. Aged SOD2+/- SMCs had increased mitochondrial superoxide but decreased hydrogen peroxide levels. Finally, dominant-negative Forkhead box O3a overexpression attenuated staurosporine-induced apoptosis in aged SOD2+/- SMCs. CONCLUSION: Mitochondrial oxidative stress over a lifetime causes aortic stiffening, in part by inducing vascular wall remodeling, intrinsic changes in SMC stiffness, and aortic SMC apoptosis.


Subject(s)
Aging/metabolism , Aortic Diseases/metabolism , Mitochondria/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oxidative Stress , Actins/metabolism , Age Factors , Aging/pathology , Animals , Aorta/metabolism , Aorta/physiopathology , Aortic Diseases/diagnostic imaging , Aortic Diseases/genetics , Aortic Diseases/physiopathology , Apoptosis , Cells, Cultured , Collagen Type I/metabolism , Compliance , Disease Models, Animal , Elastin/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genotype , Hydrogen Peroxide/metabolism , Insulin-Like Growth Factor I/metabolism , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/pathology , Mitogen-Activated Protein Kinase 8/metabolism , Muscle, Smooth, Vascular/diagnostic imaging , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/pathology , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Pulsatile Flow , Stroke Volume , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Superoxides/metabolism , Transfection , Ultrasonography, Doppler, Pulsed , Vasodilation , Ventricular Function, Left , Ventricular Pressure
5.
Apoptosis ; 16(7): 732-45, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21505870

ABSTRACT

One of characteristics of diabetes mellitus (DM) is endothelial cell (EC) dysfunction and apoptosis which contributes to the development of vasculopathy. Advanced glycation end products (AGEs) continuously produced in the setting of DM play an important role in causing EC dysfunction and apoptosis. However, the underlying molecular mechanism remains largely elusive. Lactadherin, a secreted glycoprotein of milk-fat globule, is expressed by multiple cell types of arterial wall including ECs. Our previous proteomic studies showed that the expression of lactadherin was significantly increased in the aorta of diabetic rats as compared with control rats and treatment with grape seed procyanidin extracts significantly inhibited the lactadherin expression in diabetic rats. We hypothesized that lactadherin plays a critical role in AGEs-induced EC apoptosis; grape seed procyanidin B2 (GSPB2) and resveratrol protect against AGEs-induced EC apoptosis through lactadherin regulation. Our results showed that AGEs upregulated lactadherin expression and lactadherin RNA interference significantly attenuated AGEs-induced EC apoptosis. Overexpression of lactadherin increased EC apoptosis with up-regulation of Bax/Bcl-2 ratio, cytochrome c release, caspase-9 and caspase-3 activation suggesting the involvement of mitochondria apoptosis pathway. Mechanistically, overexpression of lactadherin reduced the phosphorylation of GSK3beta at baseline. Our study also revealed nine proteins interacting with lactadherin in HUVEC and study of these candidate proteins could unveil further underlying molecular mechanisms. In summary, our study identified lactadherin as a key player responsible for AGEs-induced EC apoptosis and antioxidants GSPB2 and resveratrol protect against AGEs-induced EC apoptosis by inhibiting lactadherin. Targeting lactadherin with antioxidant could be translated into clinical application in the fighting against DM complications.


Subject(s)
Antigens, Surface/metabolism , Apoptosis/drug effects , Biflavonoids/pharmacology , Catechin/pharmacology , Endothelial Cells/cytology , Glycation End Products, Advanced/pharmacology , Milk Proteins/metabolism , Proanthocyanidins/pharmacology , Stilbenes/pharmacology , Vitis/chemistry , Amino Acid Sequence , Caspases/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Cytochromes c/metabolism , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mass Spectrometry , Molecular Sequence Data , Peptides/chemistry , Phosphorylation/drug effects , Plasmids , Protective Agents/pharmacology , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Resveratrol , Seeds/chemistry , Transduction, Genetic , Umbilical Veins/cytology , bcl-2-Associated X Protein/metabolism
6.
Br J Pharmacol ; 162(3): 712-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20958287

ABSTRACT

BACKGROUND AND PURPOSE: There is much evidence supporting the role of ß2-adrenoceptors (ß2AR) in angiogenesis but the mechanisms underlying their effects have not been elucidated. Hence, we studied post-ischaemic angiogenesis in the hindlimb (HL) of ß2AR knock-out mice (ß2AR-/-) in vivo and explored possible molecular mechanisms in vitro. EXPERIMENTAL APPROACH: Femoral artery resection (FAR) was performed in wild-type and ß2AR-/- mice and adaptive responses to chronic HL ischaemia were explored; blood flow was measured by ultrasound and perfusion of dyed beads, bone rarefaction, muscle fibrosis and skin thickness were evaluated by immunoflourescence and morphometric analysis. Intrafemoral delivery of an adenovirus encoding the human ß2AR (ADß2AR) was used to reinstate ß2ARs in ß2AR-/- mice. Molecular mechanisms were investigated in mouse-derived aortic endothelial cells (EC) in vitro, focusing on NFκB activation and transcriptional activity. RESULTS: Angiogenesis was severely impaired in ß2AR-/- mice subjected to FAR, but was restored by gene therapy with ADß2AR. The proangiogenic responses to a variety of stimuli were impaired in ß2AR-/- EC in vitro. Moreover, removal of ß2ARs impaired the activation of NFκB, a transcription factor that promotes angiogenesis; neither isoprenaline (stimulates ßARs) nor TNFα induced NFκB activation in ß2AR(-/-) EC. Interestingly, cAMP response element binding protein (CREB), a transcription factor that counter regulates NFκB, was constitutively increased in ß2AR(-/-) ECs. ADß2AR administration restored ß2AR membrane density, reduced CREB activity and reinstated the NFκB response to isoprenaline and TNFα. CONCLUSIONS AND IMPLICATIONS: Our results suggest that ß2ARs control angiogenesis through the tight regulation of nuclear transcriptional activity.


Subject(s)
CREB-Binding Protein/metabolism , Genetic Therapy , Ischemia/physiopathology , NF-kappa B/metabolism , Neovascularization, Physiologic , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Animals , Blood Flow Velocity , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Genetic Vectors , Humans , Ischemia/therapy , Luciferases/metabolism , Male , Mice , Mice, Knockout , Radioligand Assay , Receptors, Adrenergic, beta-2/deficiency , Signal Transduction , Transfection , Vascular Endothelial Growth Factor A/metabolism
8.
Cardiol Rev ; 18(4): 198-203, 2010.
Article in English | MEDLINE | ID: mdl-20539103

ABSTRACT

Organic nitrates have been used for over a century in cardiovascular therapy and are still widely used in the treatment of acute coronary syndromes, chronic angina pectoris, and congestive heart failure. Nitrates, together with sodium nitroprusside, generally referred to as nitrovasodilators, exert their biologic effects via the release of nitric oxide. They are also known as nitric oxide donors. The mechanism of action of these drugs is traditionally believed to lie in their arterial vasodilation and venodilation effects, resulting in an improvement of coronary artery blood supply and/or reduction of cardiac workload in the treatment of coronary artery disease and congestive heart failure. Recently it has been recognized that these drugs also have intrinsic antiplatelet and antithrombotic effects, demonstrated both in vitro and in vivo, which would add further rationale for the use of these drugs in atherothrombotic diseases. Research has shown that nitrovasodilators can nonselectively inhibit platelet aggregation induced by multiple stimuli. However, clinical trials have yielded conflicting results regarding clinical outcome, especially with long-term nitrate use. The potentially beneficial effects of nitrates could be negated by the development of tolerance and the generation of deleterious oxidative stress causing endothelial dysfunction during continuous nitrate administration. Much progress has been made in the development of new nitric oxide donors devoid of oxidant-generating properties. Novel combination therapies with nitrovasodilators plus antioxidants or agents with antioxidant properties have shown promise in reducing or reversing tolerance, potentiating antiplatelet effects, and improving clinical outcome. It is expected that clinical introduction of novel nitrovasodilator regimens will provide a new approach to the prevention and treatment of atherothrombotic diseases. Large-scale clinical trials will ultimately provide the evidence-based answers.


Subject(s)
Cardiovascular Diseases/drug therapy , Nitrates/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Blood Platelets/drug effects , Cardiovascular Diseases/physiopathology , Drug Tolerance , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Humans , Oxidative Stress , Thrombosis/drug therapy , Treatment Outcome , Vasodilator Agents/therapeutic use
9.
Zhonghua Bing Li Xue Za Zhi ; 38(1): 50-4, 2009 Jan.
Article in Chinese | MEDLINE | ID: mdl-19489226

ABSTRACT

OBJECTIVE: To study the activation of sterol regulatory element binding protein (SREBP) and its critical role in endothelial cell migration. METHODS: Bovine aortic endothelial cells (ECs) were cultured. The expression of SREBP and Cdc42 were determined by Western blot and quantitative real-time PCR. Moreover, outward growth migration model and transwell chamber assay were used to detect ECs migration. RESULTS: (1) SREBP was activated during ECs migration. Western blot analysis demonstrated increased active form SREBP in migrating as compared to non-migrating ECs population. SREBP activation decreased as ECs migration slowed;(2) Coincidental with SREBP activation, mRNA expression of its target genes such as low density lipoprotein receptor, HMG-CoA reductase, and fatty acid synthase also increased in migrating ECs population as detected by real-time PCR; (3) Migration induced SREBP activation in ECs was inhibited by SREBP-acting protein RNAi and pharmacologically by 25-hydroxycholesterol; (4) Inhibition of SREBP led to decreased ECs migration in various models; (5) Cells genetically deficient in SREBP-acting protein, S1P, or S2P, phenotypically exhibited impaired migration; (6) SREBP inhibition in ECs suppressed the activity of small GTPase Cdc42, a key molecule for ECs motility. CONCLUSIONS: SREBP is activated during and plays a critical role in ECs migration. Targeting SREBP could become a novel approach in fighting diseases involving abnormal ECs migration.


Subject(s)
Cell Movement , Fatty Acid Synthases/metabolism , Hydroxymethylglutaryl CoA Reductases/metabolism , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Aorta/cytology , CHO Cells , Cattle , Cells, Cultured , Cricetinae , Cricetulus , Endothelial Cells , Fatty Acid Synthases/genetics , Hydroxycholesterols/pharmacology , Hydroxymethylglutaryl CoA Reductases/genetics , RNA Interference , RNA, Messenger/metabolism , Receptors, LDL/genetics , Sterol Regulatory Element Binding Proteins/physiology
10.
Clin Transl Sci ; 2(1): 57-61, 2009 Feb.
Article in English | MEDLINE | ID: mdl-20443868

ABSTRACT

G protein-coupled receptor kinase 5 (GRK5) is present in endothelial cells (ECs) and has the potential to regulate EC function through seven transmembrane-spanning receptor (7TMR) signaling. Recently, it has been appreciated that GRKs can affect receptor tyrosine kinases (RTKs). VEGF, an RTK, is one of the most potent mediators for EC function and angiogenesis; therefore, we determined the role GRK5 plays in VEGF signaling in human coronary artery ECs (HCAECs). GRK5 levels were increased by VEGF treatment in HCAECs. Adenoviral overexpression of GRK5 inhibited migration and proliferation of HCAECs in response to VEGF. GRK5 overexpression in HCAECs significantly suppressed both acute and late activation of Akt and extracellular signal-related kinase (ERKs) as well as the phosphorylation of GSK-3beta, an endogenous substrate of Akt. Coimmunoprecipitations revealed that GRK5 is physically associated with Akt. This study shows for the first time that GRK5 negatively regulates VEGF signaling in HCAECs and suggests that targeted intervention of GRK5 in ECs might be a novel therapeutic strategy to prevent and treat disorders involving altered EC function.


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/enzymology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cell Movement/drug effects , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , G-Protein-Coupled Receptor Kinase 5/genetics , G-Protein-Coupled Receptor Kinase 5/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Immunoprecipitation , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
11.
Am J Physiol Heart Circ Physiol ; 295(4): H1695-704, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18723764

ABSTRACT

G protein-coupled receptor kinase 2 (GRK2) is a serine/theorinine kinase that phosphorylates and desensitizes agonist-bound G protein-coupled receptors. GRK2 is increased in expression and activity in lymphocytes and vascular smooth muscle (VSM) in human hypertension and animal models of the disease. Inhibition of GRK2 using the carboxyl-terminal portion of the protein (GRK2ct) has been an effective tool to restore compromised beta-adrenergic receptor (AR) function in heart failure and improve outcome. A well-characterized dysfunction in hypertension is attenuation of betaAR-mediated vasodilation. Therefore, we tested the role of inhibition of GRK2 using GRK2ct or VSM-selective GRK2 gene ablation in a renal artery stenosis model of elevated blood pressure (BP) [the two-kidney, one-clip (2K1C) model]. Use of the 2K1C model resulted in a 30% increase in conscious BP, a threefold increase in plasma norepinephrine levels, and a 50% increase in VSM GRK2 mRNA levels. BP remained increased despite VSM-specific GRK2 inhibition by either GRK2 knockout (GRK2KO) or peptide inhibition (GRK2ct). Although betaAR-mediated dilation in vivo and in situ was enhanced, alpha(1)AR-mediated vasoconstriction was also increased. Further pharmacological experiments using alpha(1)AR antagonists revealed that GRK2 inhibition of expression (GRK2KO) or activity (GRK2ct) enhanced alpha(1D)AR vasoconstriction. This is the first study to suggest that VSM alpha(1D)ARs are a GRK2 substrate in vivo.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , Hypertension, Renovascular/enzymology , Muscle, Smooth, Vascular/enzymology , Receptors, Adrenergic, alpha-1/metabolism , Renal Artery Obstruction/complications , Vasoconstriction , Adrenergic beta-Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Angiotensin II/metabolism , Animals , Aorta/enzymology , Blood Pressure , Cattle , Disease Models, Animal , Dose-Response Relationship, Drug , G-Protein-Coupled Receptor Kinase 2/genetics , Hypertension, Renovascular/etiology , Hypertension, Renovascular/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Norepinephrine/blood , Receptors, Adrenergic, alpha-1/drug effects , Renal Artery Obstruction/enzymology , Renal Artery Obstruction/physiopathology , Vasoconstriction/drug effects
12.
Biochim Biophys Acta ; 1784(11): 1857-64, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18692161

ABSTRACT

Although digitalis has been used in clinical treatment extensively, the precise mechanism of its toxic actions on cardiovascular system remained unclear, it would be of interest to study the differential proteomic analysis of vascular endothelial cells in response to toxic concentrations of digitalis thus to provide new agents for treatment of digitalis-induced cytotoxicity. We employed human umbilical vein endothelial cells (HUVEC) as our model system. HUVEC were exposed to increasing concentrations (0.1 nM-10 microM) of digoxin at 12-96 h intervals. Cell viability tests revealed that digoxin played dual effects on cell growth. Apoptosis detection confirmed that apoptosis was primarily responsible for digoxin-induced cell death. Proteomics analysis further revealed that the digoxin-induced apoptosis was accompanied by regulated expression of ATP synthase beta chain, cystatin A, electron transfer flavoprotein, heterogeneous nuclear ribonucleoproteins H3, lamin A, profilin-1, proteasome subunit 5, succinyl-CoA ligase beta chain and heat shock protein 60 (HSP60). Deep study on the overexpression of HSP60 confirmed that HSP60 exerted a protective role in digoxin-induced apoptosis through inhibition of caspase-3 activity in HUVEC. These results provided an impetus for further delineation of mechanism of digoxin-induced cytotoxicity and offered new agents that help attenuate its toxicity.


Subject(s)
Cell Proliferation/drug effects , Chaperonin 60/physiology , Digoxin/pharmacology , Endothelial Cells/drug effects , Proteomics , Apoptosis/drug effects , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Chaperonin 60/genetics , Chaperonin 60/metabolism , Cytoprotection/drug effects , Cytoprotection/genetics , Cytotoxins/pharmacology , Endothelial Cells/metabolism , Humans , Proteomics/methods , Transfection
13.
Life Sci ; 82(3-4): 174-81, 2008 Jan 16.
Article in English | MEDLINE | ID: mdl-18068195

ABSTRACT

Postintervention restenosis (PIRS) after balloon angioplasty or stent implantation is a limitation for these interventional procedures even with the advent of new drug-eluting stents. Sterol regulatory element-binding proteins (SREBP) are transcription factors governing cellular lipid biosynthesis and thus critical in the regulation of the lipid-rich cell membranes. PIRS following injury results partially from newly proliferating cells expressing vascular smooth muscle cell (VSMC) markers. Platelet-derived growth factor (PDGF), lysophosphatidic acid (LPA) and alpha(1)-adrenergic receptor stimulation are well recognized diverse mitogens for VSMC activation in PIRS. We examined whether PDGF, LPA and alpha(1)-adrenergic receptor stimulation with phenylephrine (PE) regulate SREBP expression and subsequently, VSMC proliferation. Our results show that PDGF, LPA and PE upregulate SREBP-1 in a time- and dose-dependent manner. PDGF, LPA and PE-mediated proliferation is dependent on SREBP since inhibition of SREBP expression using targeted knockdown of the SREBP precursor SREBP activating protein (SCAP) by siRNA led to an attenuation of SREBP expression and decreased PDGF, LPA and PE induced proliferation. In two different in vivo PIRS models we found that SREBP-1 was enhanced in the injured blood vessel wall, especially within the neointima and co-localized with alpha-smooth muscle actin positive cells. Thus, SREBP is enhanced in the vessel wall following PIRS and is important in the regulation of pro-hyperplasia molecular signaling. SREBP inhibition may be a powerful tool to limit PIRS.


Subject(s)
Blood Vessels/metabolism , Coronary Restenosis/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Biomarkers/metabolism , Blood Vessels/drug effects , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Intracellular Signaling Peptides and Proteins/genetics , Lysophospholipids/pharmacology , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phenylephrine/pharmacology , Platelet-Derived Growth Factor/pharmacology , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1 , Stents
14.
Am J Physiol Heart Circ Physiol ; 293(5): H3072-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17873012

ABSTRACT

More than 30% of the US population has high blood pressure (BP), and less than a third of people treated for hypertension have it controlled. In addition, the etiology of most high BP is not known. Having a better understanding of the mechanisms underlying hypertension could potentially increase the effectiveness of treatment. Because G(q) signaling mediates vasoconstriction and vascular function can cause BP abnormalities, we were interested in determining the role of vascular smooth muscle (VSM) G(q) signaling in two divergent models of hypertension: a renovascular model of hypertension through renal artery stenosis and a genetic model of hypertension using mice with VSM-derived high BP. Inhibition of VSM G(q) signaling attenuated BP increases induced by renal artery stenosis to a similar extent as losartan, an ANG II receptor blocker and current antihypertensive therapy. Inhibition of G(q) signaling also attenuated high BP in our genetic VSM-derived hypertensive model. In contrast, BP remained elevated 25% following treatment with losartan, and prazosin, an alpha(1)-adrenergic receptor antagonist, only decreased BP by 35%. Inhibition of G(q) signaling attenuated VSM reactivity to ANG II and resulted in a 2.4-fold rightward shift in EC(50). We also determined that inhibition of G(q) signaling was able to reverse VSM hypertrophy in the genetic VSM-derived hypertensive model. These results suggest that G(q) signaling is an important signaling pathway in two divergent models of hypertension and, perhaps, optimization of antihypertensive therapy could occur with the identification of particular G(q)-coupled receptors involved.


Subject(s)
Blood Pressure , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypertension/congenital , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Signal Transduction , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hypertension, Renal/physiopathology , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
Am J Physiol Heart Circ Physiol ; 293(1): H60-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17384131

ABSTRACT

Recent studies from our lab and others have shown that the hematopoietic cytokine erythropoietin (EPO) can protect the heart from ischemic damage in a red blood cell-independent manner. Here we examined any protective effects of the long-acting EPO analog darbepoetin alfa (DA) in a rat model of ischemia-reperfusion (I/R) injury. Rats were subjected to 30-min ischemia followed by 72-h reperfusion. In a dose-response study, DA (2, 7, 11, and 30 mug/kg) or vehicle was administered as a single bolus at the start of ischemia. To determine the time window of potential cardioprotection, a single high dose of DA (30 mug/kg) was given at either the initiation or the end of ischemia or at 1 or 24 h after reperfusion. After 3 days, cardiac function and infarct size were assessed. Acute myocyte apoptosis was quantified by TUNEL staining on myocardial sections and by caspase-3 activity assays. DA significantly reduced infarct size from 32.8 +/- 3.5% (vehicle) to 11.0 +/- 3.3% in a dose-dependent manner, while there was no difference in ischemic area between groups. Treatment with DA as late as 24 h after the beginning of reperfusion still demonstrated a significant reduction in infarct size (17.0 +/- 1.6%). Consistent with infarction data, DA improved in vivo cardiac reserve compared with vehicle. Finally, DA significantly decreased myocyte apoptosis and caspase-3 activity after I/R. These data indicate that DA protects the heart against I/R injury and improves cardiac function, apparently through a reduction of myocyte apoptosis. Of clinical importance pointing toward a relevant therapeutic utility, we report that even if given 24 h after I/R injury, DA can significantly protect the myocardium.


Subject(s)
Cardiotonic Agents/administration & dosage , Disease Models, Animal , Erythropoietin/analogs & derivatives , Myocardial Ischemia/drug therapy , Myocardial Ischemia/physiopathology , Animals , Darbepoetin alfa , Dose-Response Relationship, Drug , Erythropoietin/administration & dosage , Hematinics/administration & dosage , Humans , Male , Myocardial Ischemia/diagnosis , Rats , Rats, Sprague-Dawley , Treatment Outcome
16.
Biol Pharm Bull ; 30(2): 247-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17268060

ABSTRACT

Digitalis has been used to treat congestive heart failure for more than 200 years, although the dual effects (proliferation and death) induced by digitalis on cell growth have been known for many years, the mechanisms by which digitalis causes the actions were not completely known. The aim of this work was to characterize the proliferative effect of ouabain on cell growth in endothelial cells, and, to do the differential proteomic analysis of human umbilical vein endothelial cells (HUVEC) in response to ouabain and examine changes in protein expression. HUVEC were exposed to different concentrations (0.1-100 nM) of ouabain at 12-48 h intervals. Cell growth and morphological changes of HUVEC treated with ouabain were compared with cells under nontreated conditions. Ouabain stimulated HUVEC cell proliferation at low concentrations and induced cell death at higher concentrations. Using proteomics study, we identified 32 proteins of HUVEC with various important cellular functions and revealed 8 proteins such as Annexin A1, Annexin A2, Malate dehydrogenase, Myosin regulatory light chain 2 (MRLC2), Profilin-1, S100 calcium-binding protein A13, Triosephosphate isomerase and Translationally controlled tumor protein, regulated by low-dose ouabain treatment and MRLC2 was subsequently confirmed by Western blot. Our results give new insights into the cellular and molecular mechanisms of the proliferation action of low-dose ouabain on HUVEC and provide new avenues for the treatment of cardiovascular diseases.


Subject(s)
Cardiotonic Agents/pharmacology , Endothelial Cells/drug effects , Ouabain/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Humans , Proteins/metabolism , Proteomics , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Umbilical Veins/cytology
17.
J Leukoc Biol ; 80(3): 608-20, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16849610

ABSTRACT

Angiogenesis is essential in many physiological and pathological processes and can be stimulated by many different factors. To better understand and to manipulate this process more effectively, it would be beneficial to identify molecules common to the signaling pathways stimulated by different classes of angiogenic factors. Sterol regulatory element-binding proteins (SREBPs) are involved in the metabolism of cholesterol and fatty acids, molecules that are critical in membrane biology, and hence, many of the processes involved in angiogenesis. Here, we show that angiogenic factors of different families, such as basic fibroblast growth factor, thrombin, and interleukin (IL)-8, stimulate SREBP activation, whereas nonangiogenic factors, such as transforming growth factor-beta1, do not. We focused our detailed studies on IL-8 in vitro and in vivo, as this chemokine is also involved in inflammation and hence, has the potential to be critical in inflammation-induced angiogenesis, a process common to many diseases. Using human microvascular endothelial cells, a rabbit skin wound-healing model, and the chorioallantoic membrane assay, we show that IL-8 stimulates the activation of SREBP-1 and -2, and this activation is specific and receptor-mediated. SREBP activation leads to activation of RhoA through 3-hydroxy-3-methylglutaryl CoA reductase. RhoA is a small guanosinetriphosphatase, important in cytoskeletal functions, which in turn, are critical in many of the cellular processes needed for angiogenesis. Given that diverse, angiogenic factors use different cell-surface receptors, identification of this common step in the signal-transduction pathway provides the opportunity for novel approaches for prevention and treatment of diseases involving abnormal angiogenesis.


Subject(s)
Interleukin-8/physiology , Neovascularization, Physiologic/immunology , Sterol Regulatory Element Binding Proteins/metabolism , Cell Movement/immunology , Cell Proliferation , Down-Regulation , Endothelial Cells/drug effects , Endothelial Cells/immunology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/physiology , Humans , Interleukin-8/pharmacology , Sterol Regulatory Element Binding Proteins/drug effects , Thrombin/pharmacology , Thrombin/physiology , Time Factors , Transcription Factors/genetics , Transcription Factors/immunology , rhoA GTP-Binding Protein/metabolism
18.
Mol Ther ; 12(6): 1120-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16168714

ABSTRACT

Myocardial infarction (MI) represents an enormous clinical challenge as loss of myocardium due to ischemic injury is associated with compromised left ventricular (LV) function often leading to acute cardiac decompensation or chronic heart failure. S100A1 was recently identified as a positive inotropic regulator of myocardial contractility in vitro and in vivo. Here, we explore the strategy of myocardial S100A1 gene therapy either at the time of, or 2 h after, MI to preserve global heart function. Rats underwent cryothermia-induced MI and in vivo intracoronary delivery of adenoviral transgenes (4 x 10(10) pfu). Animals received saline (MI), the S100A1 adenovirus (MI/AdS100A1), a control adenovirus (MI/AdGFP), or a sham operation. S100A1 gene delivery preserved global in vivo LV function 1 week after MI. Preservation of LV function was due mainly to S100A1-mediated gain of contractility of the remaining, viable myocardium since contractile parameters and Ca(2+) transients of isolated MI/AdS100A1 myocytes were significantly enhanced compared to myocytes isolated from both MI/AdGFP and sham groups. Moreover, S100A1 gene therapy preserved the cardiac beta-adrenergic inotropic reserve, which was associated with the attenuation of GRK2 up-regulation. Also, S100A1 overexpression reduced cardiac hypertrophy 1 week post-MI. Overall, our data indicate that S100A1 gene therapy provides a potential novel treatment strategy to maintain contractile performance of the post-MI heart.


Subject(s)
Calcium-Binding Proteins/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Myocardial Infarction/pathology , Myocardium/pathology , Adenoviridae/genetics , Animals , Blotting, Western , Calcium/metabolism , Cold Temperature , Echocardiography , Green Fluorescent Proteins/metabolism , Heart Ventricles/pathology , Hemodynamics , Immunohistochemistry , In Vitro Techniques , Ischemia , Male , Models, Statistical , Muscle Contraction , Myocardium/metabolism , RNA/metabolism , Rats , Receptors, Adrenergic, beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , S100 Proteins , Time Factors , Transgenes
19.
Circulation ; 112(8): 1145-53, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16103237

ABSTRACT

BACKGROUND: Essential hypertension involves an increase in sympathetic nervous system activity and an associated decrease in beta-adrenergic receptor (AR)-mediated dilation. In addition, increased levels of G protein-coupled receptor (GPCR) kinases (GRKs), which regulate GPCR signaling, are associated with increased blood pressure (BP). METHODS AND RESULTS: We generated transgenic mice with approximately 2-fold vascular smooth muscle (VSM)-specific overexpression of GRK5 to recapitulate a selective aspect of hypertension and understand the impact on GPCR regulation of BP. VSM-GRK5 mice were hypertensive, with a 25% to 35% increase in BP, whereas there was no concomitant cardiac or VSM hypertrophy. BP elevations were segregated with sex, with male mice having higher levels than female mice, and ovariectomy did not alter this phenotype. BP was restored to control values with pertussis toxin Gi-signaling inhibition or chronic beta1AR inhibition after 7 days of CGP20712A, whereas the beta2AR antagonist ICI 118,551 was ineffective. Alpha1AR response was not altered, nor was betaAR-mediated dilation in male blood vessels, whereas norepinephrine sensitivity was increased. In contrast, female VSM-GRK5 blood vessels have diminished betaAR-mediated dilation and enhanced sensitivity to angiotensin II (Ang II). CONCLUSIONS: Our data suggest that in both male and female mice, VSM-specific overexpression of GRK5 elevates BP mediated by Gi and, at least in part, by beta1AR in males and Ang II receptors in females. Understanding mechanisms underlying an increase in VSM-GRK5 may have a profound influence on the use and development of antihypertensive therapeutics.


Subject(s)
Blood Pressure/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiology , Protein Serine-Threonine Kinases/genetics , Sex Characteristics , Angiotensin II/pharmacology , Animals , Aorta/drug effects , Aorta/physiology , Cyclic AMP/metabolism , Female , G-Protein-Coupled Receptor Kinase 5 , Hypertension/genetics , Hypertension/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Ovariectomy , Potassium Channels/metabolism , Potassium Chloride/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Receptors, Adrenergic, beta-2/metabolism , Signal Transduction/physiology , Transgenes/physiology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilation/physiology
20.
Circ Res ; 95(5): 471-8, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15271857

ABSTRACT

By stimulating the migration and proliferation of endothelial cells (ECs), vascular endothelial growth factor (VEGF) is a potent angiogenic factor. However, the molecular mechanism involved in the VEGF-induced angiogenesis remains elusive. We hypothesized that sterol regulatory element binding proteins (SREBPs), transcription factors governing cellular lipid homeostasis, play an important role in regulating angiogenesis in response to VEGF. VEGF activated SREBP1 and SREBP2 in ECs, as demonstrated by the increased SREBPs, their cleavage products, and the upregulation of the targeted genes. VEGF-induced SREBP activation depended on SREBP cleavage-activating protein (SCAP), because knocking down SCAP by RNA interference (RNAi) inhibited SREBP activation in response to VEGF. SREBP activation was also blocked by 25-hydroxycholesterol (25-HC). To verify the functional implication of SREBPs in VEGF-induced angiogenesis, we tested the role of SREBPs in EC migration and proliferation. SCAP RNAi or 25-HC inhibited VEGF-induced pseudopodia extension and migration of ECs. Both treatments inhibited VEGF-induced EC proliferation, with cell growth arrested at the G(0)/G(1) phase and a concomitant decrease of the S phase. Blocking the PI3K-Akt pathway inhibited the VEGF-activated SREBPs, demonstrating that PI3K-Akt regulates SREBPs. Consistent with our in vitro data, SREBP1 was detected in newly developed microvasculatures in a rabbit skin partial-thickness wound-healing model. SREBP inhibition also markedly suppressed VEGF-induced angiogenesis in chick embryos. In summary, this study identifies SREBPs as the key molecules in regulating angiogenesis in response to VEGF.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Endothelium, Vascular/physiology , Neovascularization, Physiologic , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Chick Embryo , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Hydroxycholesterols/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins/physiology , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Skin/blood supply , Skin Physiological Phenomena , Sterol Regulatory Element Binding Protein 1 , Sterol Regulatory Element Binding Protein 2 , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...