Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Am Heart Assoc ; 13(9): e033474, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700020

ABSTRACT

BACKGROUND: Copper exposure is reported to be associated with increased risk of stroke. However, the association of copper exposure with subclinical carotid atherosclerosis remains unclear. METHODS AND RESULTS: This observational study included consecutive participants from Xinqiao Hospital between May 2020 and August 2021. Blood metals were measured using inductively coupled plasma mass spectrometry and carotid atherosclerosis was assessed using ultrasound. Modified Poisson regression was performed to evaluate the associations of copper and other metals with subclinical carotid plaque presence. Blood metals were analyzed as categorical according to the quartiles. Multivariable models were adjusted for age, sex, body mass index, education, smoking, drinking, hypertension, diabetes, dyslipidemia, estimated glomerular filtration rate, and coronary artery disease history. Bayesian Kernel Machine Regression was conducted to evaluate the overall association of metal mixture with subclinical carotid plaque presence. One thousand five hundred eighty-five participants were finally enrolled in our study, and carotid plaque was found in 1091 subjects. After adjusting for potential confounders, metal-progressively-adjusted models showed that blood copper was positively associated with subclinical carotid plaque (relative risk according to comparing quartile 4 to quartile 1 was 1.124 [1.021-1.238], relative risk according to per interquartile increment was 1.039 [1.008-1.071]). Blood cadmium and lead were also significantly associated with subclinical carotid plaque. Bayesian Kernel Machine Regression analyses suggested a synergistic effect of copper-cadmium-lead mixture on subclinical carotid plaque presence. CONCLUSIONS: Our findings identify copper as a novel risk factor of subclinical carotid atherosclerosis and show the potential synergistic proatherogenic effect of copper, cadmium, and lead mixture.


Subject(s)
Carotid Artery Diseases , Copper , Humans , Female , Male , Carotid Artery Diseases/blood , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/epidemiology , Copper/blood , Middle Aged , Risk Factors , Aged , Plaque, Atherosclerotic/blood , Cadmium/blood , Risk Assessment , China/epidemiology , Biomarkers/blood , Asymptomatic Diseases , Lead/blood
2.
Cytokine ; 167: 156213, 2023 07.
Article in English | MEDLINE | ID: mdl-37121091

ABSTRACT

BACKGROUND: Anti-aging protein Klotho has been reported to be associated with atherosclerosis, which was considered as a chronic inflammatory disease. However, the relationship between Klotho and senile inflammation remained unclear. The present study aims to ascertain the correlation of Klotho with inflammation in middle-aged and elderly coronary atherosclerotic disease (CAD). METHODS: A total of 302 patients with CAD were included in this study. Coronary atherosclerosis was confirmed and quantified for all patients by coronary angiography. Serum Klotho was detected by enzyme linked immunosorbent assay. Serum concentrations of IL-6 and IL-8 were quantified by chemiluminescence assay. T-lymphocyte subsets were measured using flow cytometry. RESULTS: Multivariate linear regression analysis showed that serum Klotho was an independent predictor for circulating monocytes (standard ß = -0.321, P < 0.001) and CD4+/CD8+ ratio (standard ß = -0.522, P < 0.001). After adjustment, serum Klotho was still independently associated with IL-6 (standard ß = -0.395, P < 0.001) and IL-8 (standard ß = -0.296, P < 0.001). Moreover, circulating monocytes, CD4+ and CD8+ lymphocytes were correlated with increased serum concentrations of IL-6 and IL-8, independent of CRP (P < 0.05). In receiver operating characteristic curve analysis, CD4+/CD8+ ratio (AUC = 0.863, P < 0.001), IL-6 (AUC = 0.893, P < 0.001) and IL-8 (AUC = 0.884, P < 0.001) presented the excellent predictive performance for significant CAD. CONCLUSIONS: Decreased concentrations in serum Klotho reflect senile inflammation, which is related to the severity of CAD in middle-aged and elderly patients.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Aged , Humans , Middle Aged , Aging , Coronary Angiography , Inflammation , Interleukin-6 , Interleukin-8
3.
Mech Ageing Dev ; 211: 111789, 2023 04.
Article in English | MEDLINE | ID: mdl-36764463

ABSTRACT

OBJECTIVE: We aimed to evaluate the prognostic performance of circulating Klotho in coronary atherosclerotic disease (CAD), and to further explore the effect of Klotho on stress-mediated endothelial senescence and underlying mechanism. METHODS: A cohort of 295 patients had a 12-month follow-up for major adverse cardiovascular events (MACE). Serum Klotho was detected by enzyme linked immunosorbent assay. Cell viability, SA-ß-Gal staining, the expression of P53 and P16 were analyzed for endothelial senescence. Oxidative stress was evaluated by measurement of reactive oxygen species, superoxide dismutase and malondialdehyde. LC3, P62, Wnt3a, GSK-3ß and mTOR were analyzed by western blotting. Autophagosome formation was detected by adenovirus transfection. RESULTS: In epidemiological analysis, low Klotho (≤295.9 pg/ml) was significantly associated with MACE risk (HR=2.266, 95 %CI 1.229-4.176). In experimental analysis, Klotho alleviated endothelial senescence and oxidative stress caused by Ang-II exposure; Klotho restored impaired autophagic flux to ameliorate Ang-II induced endothelial senescence; Ang-II activated Wnt3a/GSK-3ß/mTOR signaling to inhibit autophagy, whereas Klotho restored autophagy through blockade of Wnt3a/GSK-3ß/mTOR signaling; Klotho ameliorated endothelial senescence by suppressing Wnt3a/GSK-3ß/mTOR pathway under Ang-II exposure. CONCLUSIONS: Prognostic significance of Klotho in CAD is potentially ascribed to its anti-endothelial senescence effect via autophagic flux restoration by inhibiting Wnt3a/ GSK-3ß/mTOR signaling.


Subject(s)
Atherosclerosis , Signal Transduction , Humans , Autophagy , Cellular Senescence , Glycogen Synthase Kinase 3 beta/metabolism , Prognosis , TOR Serine-Threonine Kinases/metabolism , Wnt3A Protein/pharmacology , Klotho Proteins/metabolism , Angiotensin II/pharmacology
4.
Sci Rep ; 7(1): 18067, 2017 12 22.
Article in English | MEDLINE | ID: mdl-29273744

ABSTRACT

Endothelial progenitor cells (EPC) participate in vessel recovery and maintenance of normal endothelial function. Therefore, pitavastatin-nanoparticles (NPs)-engineered EPC may be effective in repairing injured vasculature. Pitavastatin-loaded poly(lactic-co-glycolic) acid (PLGA) NPs were obtained via ultrasonic emulsion solvent evaporation with PLGA as the carrier encapsulating pitavastatin. The effects and mechanism of pitavastatin-NPs on EPC proliferation in vitro were evaluated. Then, EPC that internalized pitavastatin-NPs were transplanted into rats after carotid artery injury. EPC homing, re-endothelialization, and neointima were evaluated by fluorescence labeling, evans Blue and hematoxylin/eosin (H&E) staining. Pitavastatin-NPs significantly improved EPC proliferation compared with control and pitavastatin group. Those effects were blocked by pretreatment with the pharmacological phosphoinositide 3-kinase (PI3K) blockers LY294002. After carotid artery injury, more transplanted EPC were detected in target zone in Pitavastatin-NPs group than pitavastatin and control group. Re-endothelialization was promoted and intimal hyperplasia was inhibited as well. Thus, pitavastatin-NPs promote EPC proliferation via PI3K signaling and accelerate recovery of injured carotid artery.


Subject(s)
Carotid Artery Injuries/drug therapy , Cell Proliferation/drug effects , Endothelial Progenitor Cells/drug effects , Nanoparticles , Quinolines/therapeutic use , Animals , Chromones/pharmacology , Drug Delivery Systems , Endothelial Progenitor Cells/cytology , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Quinolines/pharmacology , Rats
5.
Stem Cell Res Ther ; 8(1): 237, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29065929

ABSTRACT

BACKGROUND: Endothelial progenitor cell (EPC) differentiation is considered crucial for vascular repair. Vascular endothelial growth factor (VEGF) induces EPC differentiation, but the underlying mechanism of this phenomenon remains unclear. Connexin 43 (Cx43) is reported to be involved in the regulation of stem cell differentiation. Therefore, we sought to determine whether Cx43 is involved in VEGF-induced EPC differentiation and vascular repair. METHODS: Rat spleen-derived EPCs were cultured and treated with various concentrations of VEGF (0, 10, or 50 ng/mL), and the relationship between EPC differentiation and Cx43 expression was evaluated. Thereafter, fluorescence redistribution after photobleaching was performed to assess the relationship between adjacent EPC differentiation and Cx43-induced gap junction intercellular communication (GJIC). After carotid artery injury, EPCs pretreated with VEGF were injected into the tail veins, and the effects of Cx43 on vascular repair were evaluated. RESULTS: EPCs cultured with VEGF exhibited accelerated differentiation and increased expression of Cx43. However, inhibition of Cx43 expression using short interfering RNA (siRNA) attenuated EPC GJIC and consequent EPC differentiation. VEGF-pretreated EPC transplantation promoted EPC homing and reendothelialization, and inhibited neointimal formation. These effects were attenuated by siRNA inhibition of Cx43. CONCLUSIONS: Our results from in vivo and in vitro experiments indicated that VEGF promotes EPC differentiation and vascular repair through Cx43.


Subject(s)
Carotid Artery Injuries/therapy , Connexin 43/genetics , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/transplantation , Regeneration/physiology , Vascular Endothelial Growth Factor A/pharmacology , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Communication/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Connexin 43/antagonists & inhibitors , Connexin 43/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Gene Expression Regulation , Male , Neointima/prevention & control , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Spleen/cytology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
6.
Sci Rep ; 6: 30053, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27530451

ABSTRACT

The DNA methyltransferase-mediated proinflammatory activation of macrophages is causally linked to the development of atherosclerosis (AS). However, the role of DNMT1, a DNA methylation maintenance enzyme, in macrophage polarization and AS development remains obscure. Here, we established transgenic mice with macrophage-specific overexpression of DNMT1 (Tg(DNMT1)) or PPAR-γ (Tg(PPAR-γ)) to investigate their effects on AS progression in ApoE-knockout mice fed an atherogenic diet. Primary macrophages were extracted to study the role of the DNMT1/PPAR-γ pathway in regulating inflammatory cytokine production. We demonstrated that Tg(DNMT1) significantly increased proinflammatory cytokine production in macrophages and plasma, and it accelerated the progression of AS in the atherogenic diet-treated ApoE-knockout mice. Further, we found that the DNA methylation status of the proximal PPAR-γ promoter was regulated by DNMT1 in macrophages. Notably, additional Tg(PPAR-γ) or pharmacological activation of PPAR-γ effectively prevented Tg(DNMT1)-induced proinflammatory cytokine production in macrophages and AS development in the mouse model. Finally, we demonstrated that elevated DNMT1 was correlated with decreased PPAR-γ, and increased proinflammatory cytokine production in the peripheral blood monocytes isolated from the patients with AS, compared to those of healthy donors. Our findings shed light on a novel strategy for the prevention and therapy of AS.


Subject(s)
Atherosclerosis/immunology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/genetics , Macrophage Activation/immunology , Macrophages/immunology , PPAR gamma/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Cytokines/biosynthesis , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Humans , Hypoglycemic Agents/pharmacology , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , PPAR gamma/agonists , PPAR gamma/genetics , Promoter Regions, Genetic/genetics , Rosiglitazone , Thiazolidinediones/pharmacology
7.
Exp Biol Med (Maywood) ; 240(12): 1752-63, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26069270

ABSTRACT

Hypercholesterolemia impairs the quantity and function of endothelial progenitor cell. We hypothesized that glycogen synthase kinase 3ß activity is involved in regulating biological function of endothelial progenitor cells in hypercholesterolemia microenvironment. For study, endothelial progenitor cells derived from apolipoprotein E-deficient mice fed with high-fat diet were used. Glycogen synthase kinase 3ß activity was interfered with glycogen synthase kinase 3ß inhibitor lithium chloride or transduced with replication defective adenovirus vector expressing catalytically inactive glycogen synthase kinase 3ß (GSK3ß-KM). Functions of endothelial progenitor cells, proliferation, migration, secretion and network formation of endothelial progenitor cells were assessed in vitro. The expression of phospho-glycogen synthase kinase 3ß, ß-catenin and cyclinD1 in endothelial progenitor cells was detected by Western blot. The in vivo function re-endothelialization and vasodilation were also analyzed by artery injury model transplanted with glycogen synthase kinase 3ß-inhibited endothelial progenitor cells. We demonstrated that while the proliferation, migration, network formation as well as VEGF and NO secretion were impaired in apolipoprotein E-deficient endothelial progenitor cells, glycogen synthase kinase 3ß inhibition significantly improved all these functions. Apolipoprotein E-deficient endothelial progenitor cells showed decreased phospho-glycogen synthase kinase 3ß, ß-catenin and cyclinD1 expression, whereas these signals were enhanced by glycogen synthase kinase 3ß inhibition and accompanied with ß-catenin nuclear translocation. Our in vivo model showed that glycogen synthase kinase 3ß inhibition remarkably increased re-endothelial and vasodilation. Taken together, our data suggest that inhibition of glycogen synthase kinase 3ß is associated with endothelial progenitor cell biological functions both in vitro and in vivo. It might be an important interference target in hypercholesterolemia microenvironment.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Endothelial Progenitor Cells/physiology , Glycogen Synthase Kinase 3/physiology , Hypercholesterolemia/physiopathology , Animals , Blotting, Western , Cell Cycle/physiology , Cells, Cultured , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Hypercholesterolemia/enzymology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Vascular Endothelial Growth Factor A/metabolism
8.
Stem Cell Res Ther ; 6: 37, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25956351

ABSTRACT

INTRODUCTION: Transplantation of endothelial progenitor cells (EPCs) restores endothelial function in patients with endothelial dysfunction and initial denudation. The goal of the present study was to determine the effect of cryopreserved human umbilical cord blood (UCB)-derived EPC infusion on the repair of carotid artery injury in nude rats. METHODS: Mononuclear cells (MNCs) from human cryopreserved UCB and peripheral blood (PB) of patients with cardiovascular diseases and healthy volunteers were cultured in a conditioned medium. The in vitro migration, proliferation, adhesion, and survival capacities, as well as paracrine cytokine release of EPCs were investigated. EPC homing, induced reendothelialization, and the effect on neointima formation were also assessed in vivo. RESULTS: Patient-derived PB EPCs (PPB-EPCs) displayed decreased migration, proliferation, adhesion, and survival capabilities as compared to PB-EPCs from healthy volunteers (HPB-EPCs) and cryopreserved UCB-EPCs. However, there was no difference in the release of vascular endothelial growth factor (VEGF) and stromal cell derived factor 1 (SDF-1) between the three groups. Two weeks after transplantation, more labeled UCB-EPCs and HPB-EPCs than PPB-EPCs were found by cell tracking in the injury zone. Administration of PPB-EPCs, HPB-EPCs, and UCB-EPCs enhanced reendothelialization and inhibited neointima formation compared to the saline control. However, UCB-EPC and HPB-EPC infusion showed a greater improvement than PPB-EPCs. CONCLUSIONS: Cryopreserved UCB-MNCs derived EPCs and HPB-EPCs show better responses to cytokines and vascular injury than PPB-EPCs. Thus, cryopreservation and delivery of cryopreserved autogenous UCB-EPCs or HPB-EPCs may be a promising vasculoprotective approach for patients with multiple cardiovascular risk factors.


Subject(s)
Carotid Artery Injuries/therapy , Endothelial Progenitor Cells/transplantation , Fetal Blood/cytology , Aged , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Cryopreservation , Cytokines/metabolism , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Female , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/transplantation , Male , Middle Aged , Rats , Rats, Nude , Transplantation, Heterologous
9.
Stem Cells Dev ; 24(13): 1582-90, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25753987

ABSTRACT

The dysfunction of endothelial progenitor cells (EPCs) has been shown to prevent endothelial repair during the development of atherosclerosis (AS). Previous studies have revealed that store-operated calcium entry (SOCE) is an important factor in regulating EPC functions. However, whether this is also the mechanism in AS has not been elucidated. Therefore, we evaluated the role of SOCE in EPCs isolated from an atherosclerotic mouse model. Atheromatous plaques were more frequent in the aortas of ApoE(-/-) mice fed a high-fat diet for 16 weeks compared with controls, and the proliferative and migratory activities of atherosclerotic EPCs were significantly decreased. Accordingly, SOCE amplitude, as well as spontaneous or VEGF-induced Ca(2+) oscillations, decreased in atherosclerotic EPCs. These results may be associated with the downregulated expression of Stim1, Orai1, and TRPC1, which are major mediators of SOCE. In addition, eNOS expression and phosphorylation at Ser(1177), which are critical regulators of EPC function, were markedly reduced in the atherosclerotic EPCs. The impairment of eNOS activity could also be induced by using an SOCE inhibitor or by Stim1 gene silencing, indicating a link between the activities of eNOS and SOCE in AS. Furthermore, decreased SOCE function inhibited EPC proliferation and migration in vitro. In conclusion, our results showed that the reduction of SOCE induced EPC dysfunction during AS, potentially through downregulation of store-operated calcium channel (SOCC) components and impaired eNOS activity. Approaches aimed at reestablishing SOCE activity may thus improve the function of EPCs during AS.


Subject(s)
Atherosclerosis/metabolism , Calcium Signaling , Endothelial Progenitor Cells/metabolism , Animals , Aorta/pathology , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/pathology , Calcium Channels/genetics , Calcium Channels/metabolism , Cell Movement , Cell Proliferation , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
10.
Zhonghua Xin Xue Guan Bing Za Zhi ; 42(3): 214-8, 2014 Mar.
Article in Chinese | MEDLINE | ID: mdl-24831478

ABSTRACT

OBJECTIVE: To observe the effect of platelet derived growth factor receptor ß (PDGFR-ß) transfected endothelial progenitor cells (EPCs) on vascular regeneration. METHODS: Spleen-derived mononuclear cells (MNCs) were isolated using density gradient centrifugation and induced with special culture medium. EPCs transfection was performed with Lipofectamine(TM) 2000 reagent according to the instruction manual. Carotid artery injury was induced in splenectomized mice. EPCs were injected by tail vein immediately and at 24 h after endothelial injury of the carotid artery. Evans blue staining was performed to evaluate reendothelialization at 7 days after endothelial injury of the carotid artery. RESULTS: Most adherent cells were LDL and UEA-I double positive. Laser scanning confocal microscopy showed that transfection efficiency was about 50%-60%. The reendothelialized area in the PDGFR-ß-EPCs group was significantly larger than that in EGFP-EPCs group. CONCLUSION: Transplantation of PDGFR-ß over-expressed EPCs can promote reendothelialization in the early phase after carotid artery injury.


Subject(s)
Carotid Artery Injuries/surgery , Endothelial Cells/cytology , Receptors, Platelet-Derived Growth Factor/genetics , Stem Cells/cytology , Animals , Carotid Artery Injuries/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Endothelium, Vascular/cytology , Male , Mice , Mice, Inbred C57BL , Receptors, Platelet-Derived Growth Factor/metabolism , Regeneration , Stem Cell Transplantation , Stem Cells/metabolism
11.
PLoS One ; 7(9): e43922, 2012.
Article in English | MEDLINE | ID: mdl-22984452

ABSTRACT

Cardiac stem cells (CSCs) can home to the infarcted area and regenerate myocardium. Stromal cell-derived factor-1α/C-X-C chemokine receptor type 4 (SDF-1α/CXCR4) axis is pivotal in inducing CSCs migration. However, the mechanisms remain unclear. This study set out to detect if SDF-1α promotes migration and engraftment of CSCs through the CXCR4/PI3K (phosphatidylinositol 3-kinase) pathway. In the in vitro experiment, c-kit+ cells were isolated from neonatal mouse heart fragment culture by magnetic cell sorting. Fluorescence-activated cell sorting results demonstrated that a few c-kit+ cells expressed CD45 (4.54%) and Sca-1 (2.58%), the hematopoietic stem cell marker. Conditioned culture could induce c-kit+ cells multipotent differentiation, which was confirmed by cardiac troponin I (cTn-I), α-smooth muscle actin (α-SMA), and von Willebrand factor (vWF) staining. In vitro chemotaxis assays were performed using Transwell cell chambers to detect CSCs migration. The results showed that the cardiomyocytes infected with rAAV1-SDF-1α-eGFP significantly increased SDF-1α concentration, 5-fold more in supernatant than that in the control group, and subsequently attracted more CSCs migration. This effect was diminished by administration of AMD3100 (10 µg/ml, CXCR4 antagonist) or LY294002 (20 µmol/L, PI3K inhibitor). In myocardial infarction mice, overexpression of SDF-1α in the infarcted area by rAAV1-SDF-1α-eGFP infection resulted in more CSCs retention to the infarcted myocardium, a higher percentage of proliferation, and reduced infarcted area which was attenuated by AMD3100 or ly294002 pretreatment. These results indicated that overexpression of SDF-1α enhanced CSCs migration in vitro and engraftment of transplanted CSCs and reduced infarcted size via CXCR4/PI3K pathway.


Subject(s)
Cell Movement , Chemokine CXCL12/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Signal Transduction , Stem Cell Transplantation , Stem Cells/cytology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation , Cell Membrane/metabolism , Cell Proliferation , Cells, Cultured , Flow Cytometry , Mice , Mice, Inbred BALB C , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Myocardial Infarction/enzymology , Myocardial Infarction/therapy , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, CXCR4/metabolism , Stem Cells/metabolism
12.
Zhonghua Xin Xue Guan Bing Za Zhi ; 39(7): 649-53, 2011 Jul.
Article in Chinese | MEDLINE | ID: mdl-22088247

ABSTRACT

OBJECTIVE: To investigate the effect of stromal interaction molecule 1 (STIM1) silencing on EPCs cell cycle. METHODS: Rat bone marrow derived endothelial progenitor cells (EPCs) were isolated and cultured in L-DMEM with 20% FBS. Ad-si/rSTIM1 and Ad-hSTIM1 were then transfected into EPCs and the expression of STIM1 mRNA was detected by RT-PCR. The cell cycle was determined using flow cytometry analysis and intracellular free Ca2+ was measured using LSCM. Co-immunoprecipitation was performed to examine the interaction between STIM1 and TRPC1. Protein levels of inositol 1, 4, 5-trisphosphate were analyzed with ELISA assay. RESULTS: Forty-eight hours after transfection, the expression of STIM1 mRNA was significantly downregulated (0.37 +/- 0.02 vs. 1.00 +/- 0.02, P < 0.05) and intracellular free Ca2+ level was significantly reduced (34.07 +/- 4.10 vs. 86.51 +/- 14.12, P < 0.05) in Ad-si/rSTIM1 group compared with control group. The cell cycle was arrested at G1 phase [(90.91 +/- 1.10)% vs. (77.10 +/- 0.56)%, P < 0.05] and the store-operated channel entry was strikingly inhibited in EPCs after treatment with Ad-si/rSTIM1. However, cotransfection of Ad-hSTIM1 with Ad-si/rSTIM1 significantly reversed these responses. Interestingly, co-immunoprecipitation study showed that STIM1 co-precipitated with TRPC1, and IP3 levels measured by ELISA were similar among three groups (P > 0.05). CONCLUSION: siRNA-mediated knockdown of STIM1 inhibited EPCs proliferation by reducing intracellular free Ca2+ through TRPC1-SOC signaling pathway.


Subject(s)
Endothelial Cells/cytology , Gene Silencing , Membrane Proteins/genetics , Neoplasm Proteins/genetics , RNA, Small Interfering , Stem Cells/cytology , Adenoviridae/genetics , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Genetic Vectors , Rats , Stromal Interaction Molecule 1 , Transfection , Transient Receptor Potential Channels/metabolism
13.
Pharmacology ; 87(5-6): 265-73, 2011.
Article in English | MEDLINE | ID: mdl-21494058

ABSTRACT

Tetramethylpyrazine (TMP) is a reactive oxygen species (ROS) antagonist that has potent properties for the treatment of a variety of vascular diseases, such as ischemic stroke and pulmonary hypertension secondary to chronic obstructive pulmonary diseases. However, there are few data about the role of TMP in hypoxia-induced pulmonary vascular leakage. This study examined the effect of TMP on hypoxia-induced pulmonary vascular leakage and the underlying mechanisms. Rat pulmonary microvascular endothelial cells (RPMVECs) treated with TMP or not were subjected to hypoxic or normoxic conditions for 24 h, and the monolayer permeability, intracellular ROS, hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) proteins levels were determined. Additionally, rats administrated TMP were exposed to hypobaric hypoxia to evaluate the effect of TMP in vivo by measuring lung water content, pulmonary vascular leakage into the lungs and immunohistochemistry for HIF-1α and VEGF. Hypoxia was found to cause a significant increase in RPMVEC monolayer permeability and intracellular ROS, HIF-1α and VEGF protein levels. Treatment with TMP decreased the hypoxia-induced RPMVEC monolayer permeability and attenuated the elevation of ROS, HIF-1α and VEGF protein levels. TMP-treated animals showed less pulmonary vascular leakage and HIF-1α and VEGF expression compared with those exposed to hypoxia alone. These observations supported that TMP inhibited the increase in pulmonary vascular permeability induced by hypoxia. The underlying mechanisms may be related to the scavenging of intracellular ROS and the suppression of hypoxia-induced upregulation of HIF-1α and VEGF proteins.


Subject(s)
Endothelial Cells/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/blood supply , Pyrazines/pharmacology , Reactive Oxygen Species/metabolism , Vascular Endothelial Growth Factors/metabolism , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Hypoxia/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/metabolism , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Signal Transduction/drug effects , Vascular Endothelial Growth Factors/antagonists & inhibitors , Vascular Endothelial Growth Factors/genetics
14.
Biochem Biophys Res Commun ; 391(1): 261-6, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19912987

ABSTRACT

Chronic renal failure (CRF) markedly accelerates the development of atherosclerosis, but the pathogenesis of uremic atherosclerosis remains to be elucidated. The klotho gene, predominantly expressed in the kidney, plays a key role in regulating aging and the development of age-related diseases in mammals. A loss of klotho results in multiple aging-like phenotypes including atherosclerosis. This study examines the relationship between the klotho expression and the development of accelerated atherosclerosis in uremic state. Eight-week-old apolipoprotein E-deficient (apo-E(-/-)) male mice underwent 5/6 partial kidney ablation to induce CRF or sham-operation. At 6 wk after nephrectomy, CRF mice showed significantly increased aortic plaque area fraction, aortic root plaque area and aortic cholesterol content as compared with non-CRF mice. Serum urea, total cholesterol and triglyceride concentrations were significantly higher in CRF apo-E(-/-) mice compared with non-CRF controls. Moreover, the expression of renal klotho gene and the serum levels of klotho protein were markedly decreased in CRF mice compared with controls. These results suggested that CRF favored atherosclerosis in apo-E(-/-) mice and uremic atherosclerosis was accompanied by down-regulation of klotho expression.


Subject(s)
Aortic Diseases/etiology , Atherosclerosis/etiology , Glucuronidase/genetics , Kidney Failure, Chronic/complications , Uremia/complications , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Down-Regulation , Gene Expression Regulation , Glucuronidase/blood , Kidney Failure, Chronic/blood , Klotho Proteins , Male , Mice , Mice, Knockout , Uremia/blood
15.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 19(12): 705-7, 2007 Dec.
Article in Chinese | MEDLINE | ID: mdl-18093422

ABSTRACT

OBJECTIVE: To explore the relationship between human umbilical vascular endothelial cells (HUVECs) and endothelial lipase (EL), and the effect of EL on expression of endothelial cell adhesion molecule (ICAM). METHODS: HUVECs was treated with tumor necrosis factor-alpha(TNF-alpha) 10 microg/L and the mRNA of adhesion molecules [intercellular adhesion molecule-1 (ICAM-1), vascular cellular adhesion molecule-1 (VCAM-1) and E-selectin] were detected by reverse transcription-polymerase chain reaction (RT-PCR). Then the effect of 50 microg/L anti-endothelial lipase (anti-EL) antibody on the influence of TNF-alpha on these adhesion molecules was observed. RESULTS: After being treated with TNF-alpha, the mRNA of adhesion molecules expressed by HUVECs were significant up-regulated, there was significant difference compared with control group (all P<0.01). These effects of TNF-alpha were significantly abolished by 50 microg/L anti-EL antibody (P<0.05 or P<0.01). CONCLUSION: EL can affect the expression of adhesion molecules on endothelial cell adhesion molecule. This effect of EL may play a role in the pathophysiologic process in the pathogenesis progress of atherosclerosis.


Subject(s)
E-Selectin/metabolism , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipase/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Humans , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins/cytology
16.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 19(11): 644-6, 2007 Nov.
Article in Chinese | MEDLINE | ID: mdl-17996128

ABSTRACT

OBJECTIVE: To evaluate the significance of the ratio of circulating endothelial cell expressing endothelial lipase (EL(+)/CECs) and supersensitivity C-reactive protein (hsCRP) in prognosis of patients with coronary artery disease (CAD). METHODS: one hundred and seven patients with acute coronary syndrome (ACS), 69 patients with stable angina pain (SAP), and 82 patients in whom CAD was excluded to serve as control were included for study. Blood samples were collected from ulnar vein, and hsCRP was detected, circulating endothelial cells (CECs) were isolated, and the ratio of CEC (EL(+)/CECs) which expressed endothelial lipase (EL) was determined by immunohistochemistry. Over a follow-up period of 6 months, the incidence of cardiac event of all patients was recorded. RESULTS: In patients with CAD, the EL(+)/CECs and hsCRP were significantly different among groups (P<0.05 or P<0.01). Among them, hsCRP and EL(+)/CECs were higher in ACS group than SAP group patients, whose hsCRP and EL(+)/CECs higher than the control group. The incidence rate of cardiovascular event was significantly higher (all P<0.01) in those whose hsCRP or EL(+)/CECs was higher than those whose with lower average values of these two parameters. Regression analysis indicated that the EL(+)/CECs and hsCRP could be used as the prognostic factor of CAD. The prognostic value of combined determination of EL(+)/CECs and hsCRP was higher. CONCLUSION: The expression of EL in endothelial cells may play a role in the progression of CAD. The EL(+)/CECs may be a good prognostic factor. EL(+)/CECs together with hsCRP may increase the prognostic value.


Subject(s)
C-Reactive Protein/metabolism , Coronary Artery Disease/blood , Endothelial Cells/enzymology , Lipase/blood , Female , Follow-Up Studies , Humans , Male , Prognosis
17.
Physiol Meas ; 27(6): N27-33, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16603795

ABSTRACT

Left ventricular catheterization is a key step for hemodynamic studies. At present, the right common carotid artery is a popular entrance, but right common carotid artery punctuation and ligation primarily leads to obstruction and second left ventricular catheterization in the same animal cannot be performed. In this note, we will describe an improved method of catheterization in rats using guide wires of percutaneous transluminal coronary angioplasty (PTCA) through the right external carotid artery and right common carotid artery, respectively. 30 rats anesthetized with sodium pentobarbital (50 mg kg(-1)) underwent the first operation and, 2 weeks later, 29 were successfully catheterized again. One rat died of left ventricle perforation while the hemodynamic monitor was performing. Therefore, our method proves able to be performed two times in one animal. Moreover, it inflicts less injury during the operation, because the tube was advanced through a soft guide wire.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Angioplasty, Balloon, Coronary/methods , Cardiac Catheterization/methods , Heart Ventricles/surgery , Animals , Cardiac Catheterization/adverse effects , Equipment Design , Equipment Failure Analysis , Female , Heart Injuries/etiology , Heart Injuries/prevention & control , Heart Ventricles/injuries , Rats , Rats, Wistar , Wounds, Penetrating/etiology , Wounds, Penetrating/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...