Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Cell Endocrinol ; 576: 111996, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37406985

ABSTRACT

BACKGROUND: Myocardial interstitial fibrosis is an important manifestation of diabetic heart disease, and insulin resistance is one of the mechanisms of myocardial interstitial fibrosis. Some studies have found that miR-543 is associated with insulin resistance, but whether it plays a role in diabetic myocardial interstitial fibrosis remains unclear. This study aimed to investigate the role of miR-543 in diabetic myocardial interstitial fibrosis. METHODS: The combination of high glucose and high insulin was used to establish an insulin-resistant myocardial fibroblast model. The expression levels of miR-543, α-SMA, collagen Ⅰ, collagen Ⅲ and PTEN were detected. Cell proliferation and migration were detected. Luciferase reporter gene assay was used to verify the targeting relationship between miR-543 and PTEN. RESULTS: The expression of miR-543 was up-regulated in myocardial fibroblasts with insulin resistance, which was consistent with the results of bioinformatics analysis. The proliferation and migration levels of myocardial fibroblasts in insulin-resistant states were increased, and the expression levels of α-SMA, collagen Ⅰ and collagen Ⅲ were also increased. Inhibition of miR-543 expression could reverse the above changes. Target gene prediction and dual luciferase reporter assay demonstrated that miR-543 could bind to the 3'UTR region of PTEN. Moreover, the effect of miR-543 on insulin-resistant myocardial fibroblasts is mediated by targeting PTEN. CONCLUSIONS: Inhibition of miR-543 can reduce myocardial fibroblast-myofibroblast transformation and collagen expression in insulin-resistant states by targeting PTEN.


Subject(s)
Insulin Resistance , Insulins , MicroRNAs , Cell Proliferation/genetics , Collagen/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Fibroblasts/metabolism , Fibrosis , Insulin Resistance/genetics , Insulins/metabolism , Luciferases/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Myofibroblasts/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Animals , Mice
2.
Angiology ; 73(6): 579-587, 2022 07.
Article in English | MEDLINE | ID: mdl-34330158

ABSTRACT

Circulating levels of microRNA-221 and 222 (miR-221/222) in patients with coronary artery disease (CAD) are elevated, yet the relationship between circulating miR-221/222 and the severity of coronary lesions in patients with acute coronary syndrome (ACS) remains unknown. In this study, the relative expression levels of circulating miR-221/222 in patients with ACS (n = 267) and controls (n = 71) were compared by real-time fluorescence quantitative-polymerase chain reaction (RT-qPCR). The ACS group was further divided into unstable angina pectoris (UA) group (n = 191) and acute myocardial infarction (AMI) group (n = 76). Significant upregulation of circulating miR-221/222 was observed in ACS. A positive linear correlation between circulating miR-221/222 and Gensini scores was demonstrated. The area under the curve (AUC) of circulating miR-221/222 in the diagnosis of coronary artery stenosis ≥50% was 0.605 and 0.643, respectively. The circulating miRNA-221/222 expression levels in ACS patients were elevated and positively associated with the severity of the coronary artery lesions. Circulating miR-221/222 may be novel biomarkers for the diagnosis of coronary artery stenosis ≥50% and the occurrence of ACS.


Subject(s)
Acute Coronary Syndrome , Circulating MicroRNA , Coronary Artery Disease , Coronary Stenosis , MicroRNAs , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/genetics , Biomarkers , Circulating MicroRNA/genetics , Coronary Artery Disease/complications , Coronary Artery Disease/genetics , Coronary Stenosis/complications , Coronary Stenosis/diagnosis , Coronary Stenosis/genetics , Coronary Vessels , Humans
3.
J Cardiovasc Pharmacol ; 75(5): 475-482, 2020 05.
Article in English | MEDLINE | ID: mdl-32141988

ABSTRACT

Nitrate esters have been used in clinical practice for more than one century for the treatment of angina. Their clinical effectiveness is due to vasodilator activity in arteries through a method of delivering nitric oxide or a nitric oxide-like compound. Recently, an increasing numbers of functions of this molecule in biology and pathophysiology have been discovered. Macrophage polarization shift in epicardial adipose tissue (EAT) has been demonstrated to be correlated with the severity of coronary artery disease (CAD). In this study, we aimed to investigate whether nitrate esters could improve coronary atherosclerosis through inhibition of macrophage polarization shift in EAT. A case-control study enrolled 48 subjects in 2 groups: CAD patients with or without nitrate esters treatment. Infiltration of M1/M2 macrophages and the expressions of pro-inflammatory and anti-inflammatory cytokines in EAT and subcutaneous white adipose tissue were investigated by immunohistochemical stain among subjects undergoing coronary artery bypass graft surgery. The expression levels of metabolic genes were investigated by real-time reverse transcription-polymerase chain reaction (RT-PCR). We found that nitrate ester treatment significantly inhibited NF-кB activity and decreased macrophage infiltration and M1/M2 macrophage ratio in EAT in patients with CAD. The expressions of pro-inflammatory cytokines were significantly decreased, along with significantly elevated expressions of anti-inflammatory cytokines in CAD patients with nitrate ester treatment, corresponding EAT dysfunction was ameliorated and the severity of patients with CAD (Gensini score) was significantly decreased. The protective effects on macrophage polarization and EAT function through NF-кB activity inhibition suggested a potential mechanism of nitrate esters in alleviating the severity of CAD.


Subject(s)
Adipose Tissue, White/drug effects , Anti-Inflammatory Agents/therapeutic use , Coronary Artery Disease/drug therapy , Esters/therapeutic use , Macrophages/drug effects , NF-kappa B/metabolism , Nitrates/therapeutic use , Pericardium/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Aged , Case-Control Studies , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Pericardium/metabolism , Pericardium/pathology , Severity of Illness Index , Signal Transduction
4.
Cell Physiol Biochem ; 48(4): 1416-1432, 2018.
Article in English | MEDLINE | ID: mdl-30064125

ABSTRACT

BACKGROUND/AIMS: Adipocyte-derived exosomes (ADEs) stimulate the activation of macrophages and contribute to the development of insulin resistance. Sonic Hedgehog (Shh) is an exosome-carrying protein and stimulates macrophages to secrete inflammatory cytokines. However, the impact of ADEs carrying Shh on the pro-inflammatory activation of macrophages and consequently, adipocyte insulin resistance is unclear. METHODS: 3T3-L1 adipocytes were cultured with high glucose and insulin to imitate the pathogeny of insulin resistance. ADEs were isolated from conditioned media of 3T3-L1 adipocytes via differential ultracentrifugation. We explored the role of ADEs carrying Shh in the polarization of macrophages by flow cytometry. Western blot and electrophoretic mobility shift assay (EMSA) were performed to determine the activation of Shh-mediated signalling pathways. The effects of ADE-treated macrophages on adipocyte insulin signalling were studied by Western blot. RESULTS: We found that circulating Shh-positive exosomes were increased in type 2 diabetes patients. High glucose and insulin increased the secretion of Shh-positive ADEs. The ADEs carrying Shh induced pro-inflammatory or M1 polarization of bone marrow-derived macrophages (BMDM) and RAW 264.7 macrophages. Inhibitors of Ptch and PI3K blocked the M1 polarization induced by ADEs, which suggests that ADEs carrying Shh mediated M1 macrophage polarization through the Ptch/PI3K signalling pathway. ADE-treated RAW 264.7 macrophages were subsequently used to assess the effect on insulin signalling in adipocytes. Using a co-culture assay, we showed that both ADE-treated macrophages and exosomes from these macrophages could decrease the expression of insulin-resistant substrate-1 (IRS-1) and hormone-sensitive lipase (HSL) in adipocytes. Inhibitors of Ptch and PI3K blocked the down-regulation of IRS-1 and HSL induced by ADE-treated macrophages. CONCLUSION: Together, these data indicate that ADEs carrying Shh induce the M1 polarization of macrophages, which contributes to insulin resistance in adipocytes through the Ptch/PI3K pathway.


Subject(s)
Diabetes Mellitus, Type 2/pathology , Exosomes/metabolism , Hedgehog Proteins/metabolism , Patched-1 Receptor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Patched-1 Receptor/antagonists & inhibitors , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction
5.
J Cachexia Sarcopenia Muscle ; 9(1): 106-118, 2018 02.
Article in English | MEDLINE | ID: mdl-29152896

ABSTRACT

BACKGROUND: Exercise rehabilitation is demonstrated to improve the prognosis of patients with coronary heart disease (CHD). Statins, as the key medicine to lower cholesterol in CHD, result in skeletal muscle injury and impair exercise training adaptation. Energy metabolism dysfunction is identified as the potential mechanism underlying statin-induced skeletal muscle injury. In this study, we investigated the effects of the metabolic modulator trimetazidine on skeletal muscle energy metabolism and statin-associated exercise intolerance. METHODS: High-fat fed apolipoprotein E knockout (ApoE-/- ) mice were given aerobic exercise and administrated simvastatin, trimetazidine, or simvastatin plus trimetazidine by gavage. Exercise capacity was evaluated at the end of the treatment by hanging grid test, forelimb grip strength, and running tolerance test. Plasma glucose, lipid, and creatine kinase concentrations were measured at the end of the treatment. After sacrifice, gastrocnemii were stored for assessment of muscle morphology and fibre type. Energy metabolism was estimated by plasma lactic acid concentration, ragged red fibres, and glycogen stores. Activities of mitochondrial complex III, citrate synthase activity, and membrane potential were measured to assess mitochondrial function. Oxidative stress was also evaluated by superoxide in mitochondria, superoxide dismutase activity, and glutathione redox state. RESULTS: In high-fat fed ApoE-/- mice, exercise training had no effect on lipid concentrations. Lower lipid concentrations with increased creatine kinase were observed with additional simvastatin treatment. Exercise capacity increased significantly in response to exercise training alone but was blunted by the addition of simvastatin. Similarly, cross-sectional area of muscle fibres and the proportion of slow-twitch fibres increased in the exercise group but decreased in the simvastatin plus exercise group. Additionally, simvastatin increased centronucleated fibres and induced energy metabolism dysfunction by inhibiting complex III activity and thus promoted oxidative stress in gastrocnemius. We demonstrated that trimetazidine could reverse simvastatin-induced exercise intolerance and muscle damages. We also found the ability of trimetazidine in restoration of muscle fibre hypertrophy and facilitating fast-to-slow type shift. The energy metabolism dysfunction and oxidative stress in gastrocnemii were rescued by trimetazidine. CONCLUSIONS: Trimetazidine alleviated statin-related skeletal muscle injury by restoration of oxidative phenotype and increasing fibre cross-sectional areas in response to exercise training. Correspondingly, the exercise training adaptation were improved in high-fat fed ApoE-/- mice. Moreover, trimetazidine is able to exert its positive effects without affecting the beneficial lipid-lowering properties of the statins. Thus, trimetazidine could be prescribed to remedy the undesirable statins-induced exercise intolerance during cardiac rehabilitation in patients with CHD.


Subject(s)
Exercise Therapy/methods , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Muscle Weakness/chemically induced , Muscle, Skeletal/pathology , Simvastatin/adverse effects , Trimetazidine/therapeutic use , Vasodilator Agents/therapeutic use , Animals , Female , Humans , Male , Mice , Trimetazidine/pharmacology , Vasodilator Agents/pharmacology
6.
J Cell Mol Med ; 21(12): 3298-3308, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28631352

ABSTRACT

The aim of this study was to investigate whether overexpression of STAMP2 improves insulin resistance by regulating angiogenesis in adipose tissues. The characteristics of diabetic mice were measured by serial metabolite and pathology tests. Samples were obtained from epididymal, subcutaneous and brown adipose tissues. Histological and morphological analysis demonstrated that STAMP2 gene overexpression reduced adipocyte size, angiogenesis in epididymal and brown adipose tissues. On aortic ring assay, microvessels sprouting from aortas were significantly inhibited after STAMP2 gene overexpression. The cellular effect of STAMP2 on angiogenesis was explored in human umbilical vein endothelial cells (HUVECs) model. Correlation of STAMP2 and angiogenesis was validated by Ad-STAMP2 transfection and STAMP2 siRNA inhibition. In vitro, overexpression of STAMP2 significantly inhibited endothelial cell migration, tube formation. The effects of Ad-STAMP2 transfection on HUVECs were abolished by treatment with PPARγ antagonist GW9662 (2.5 µM), and the roles of STAMP2 siRNA on HUVECs were also reversed by treatment with PPARγ agonist rosiglitazone (RSG) (0.1 mM). RT-PCR indicated that STAMP2 could regulate levels of adhesion molecules, vascular endothelial growth factor A and CD36. The expression of PPARγ and CD36 was decreased when STAMP2 was inhibited by siRNA, while PPARγ and CD36 were highly expressed after overexpression of STAMP2. Our results suggested that STAMP2 gene overexpression may improve insulin resistance via attenuating angiogenesis in epididymal and brown adipose tissues through the PPARγ/CD36 signalling pathway.


Subject(s)
Adipose Tissue/metabolism , CD36 Antigens/genetics , Diabetes Mellitus, Experimental/therapy , Membrane Proteins/genetics , Neovascularization, Pathologic/prevention & control , PPAR gamma/genetics , Adipose Tissue/blood supply , Adipose Tissue/pathology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , CD36 Antigens/metabolism , Cell Movement , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Insulin Resistance , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , PPAR gamma/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Streptozocin
7.
Mol Immunol ; 87: 152-160, 2017 07.
Article in English | MEDLINE | ID: mdl-28445787

ABSTRACT

BACKGROUND: Atherosclerosis (AS) is the most common and serious complication of type 2 diabetes mellitus (T2DM) and is accelerated via chronic systemic inflammation rather than hyperglycemia. Adipose tissue is the major source of systemic inflammation in abnormal metabolic state. Pro-inflammatory CD4+T cells play pivotal role in promoting adipose inflammation. Adipose-derived stem cells (ADSCs) for fat regeneration have potent ability of immunosuppression and restricting CD4+T cells as well. Whether T2DM ADSCs are impaired in antagonizing CD4+T cell proliferation and polarization remains unclear. METHODS: We constructed type 2 diabetic ApoE-/- mouse models and tested infiltration and subgroups of CD4+T cell in stromal-vascular fraction (SVF) in vivo. Normal/T2DM ADSCs and normal splenocytes with or without CD4 sorting were separated and co-cultured at different scales ex vivo. Immune phenotypes of pro- and anti-inflammation of ADSCs were also investigated. Flow cytometry (FCM) and ELISA were applied in the experiments above. RESULTS: CD4+T cells performed a more pro-inflammatory phenotype in adipose tissue in T2DM ApoE-/- mice in vivo. Restriction to CD4+T cell proliferation and polarization was manifested obviously weakened after co-cultured with T2DM ADSCs ex vivo. No obvious distinctions were found in morphology and growth type of both ADSCs. However, T2DM ADSCs acquired a pro-inflammatory immune phenotype, with secreting less PGE2 and expressing higher MHC-II and co-stimulatory molecules (CD40, CD80). Normal ADSCs could also obtain the phenotypic change after cultured with T2DM SVF supernatant. CONCLUSION: CD4+T cell infiltration and pro-inflammatory polarization exist in adipose tissue in type 2 diabetic ApoE-/- mice. T2DM ADSCs had impaired function in restricting CD4+T lymphocyte proliferation and pro-inflammatory polarization due to immune phenotypic changes.


Subject(s)
Adipose Tissue/pathology , Apolipoprotein E2/metabolism , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation/physiology , Diabetes Mellitus, Type 2/pathology , Stem Cells/pathology , Adipocytes/pathology , Animals , Cells, Cultured , Coculture Techniques/methods , Disease Models, Animal , Lymphocyte Activation/physiology , Mice , Obesity/metabolism , Obesity/pathology
11.
Age (Dordr) ; 38(3): 60, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27206970

ABSTRACT

Testosterone deficiency is associated with a higher incidence of cardiovascular diseases in men. However, its effect on cell senescence, which plays a causal role in vascular aging, remains unclear. Here, we tested the hypothesis that testosterone alleviated vascular smooth muscle cell (VSMC) senescence and collagen synthesis via growth arrest-specific protein 6 (Gas6)/Axl- and Akt/FoxO1a-dependent pathways. Testosterone significantly ameliorated angiotensin II-induced VSMC senescence and collagen overexpression. In addition, testosterone inhibited angiotensin II-induced matrix metalloproteinase-2 (MMP-2) activity, which played a pivotal role in facilitating age-related collagen deposition. Testosterone increased the expression of tissue inhibitor of metalloproteinase-2 but decreased the expression of MMP-2 and membrane type-1 metalloproteinase which contributed to increase MMP-2 activity. The effects on VSMCs senescence and collagen synthesis were mediated by restoration of angiotensin II-induced downregulation of Gas6 and Axl expression and a subsequent reduction of Akt and FoxO1a phosphorylation. The effects of testosterone were reversed by a Gas6 blocker, Axl-Fc, and a specific inhibitor of Axl, R428. Treatment of VSMCs with PI3K inhibitor LY294002 abrogated the downregulating effect of testosterone on MMP-2 activity. Furthermore, when FoxO1a expression was silenced by using a specific siRNA, the inhibitory effect of testosterone on MMP-2 activity was revered as well, that indicated this process was Akt/FoxO1a dependence. Taken together, Gas6/Axl and Akt/FoxO1a were involved in protective effects of testosterone on VSMCs senescence and collagen synthesis. Our results provide a novel mechanism underlying the protective effect of testosterone on vascular aging and may serve as a theoretical basis for testosterone replacement therapy.


Subject(s)
Aging/genetics , Collagen/biosynthesis , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Testosterone/pharmacology , Androgens/pharmacology , Animals , Apoptosis , Blotting, Western , Cattle , Cells, Cultured , Cellular Senescence , Collagen/antagonists & inhibitors , DNA/genetics , Enzyme-Linked Immunosorbent Assay , Intercellular Signaling Peptides and Proteins/biosynthesis , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Signal Transduction/drug effects , Axl Receptor Tyrosine Kinase
12.
J Transl Med ; 14(1): 109, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27121077

ABSTRACT

BACKGROUND: Trimetazidine, as an anti-ischemic and antioxidant agent, has been demonstrated to have many cardioprotective effects. However, whether early administration of trimetazidine has an effect on diabetic cardiomyopathy and the mechanisms underlying the effect have not yet been elucidated. METHODS: We established a type 2 DCM rat model by high-fat diet and low-dose streptozotocin. Rats were separated into different groups: control, diabetes, and diabetes + trimetazidine (n = 6, each). Cardiac autophagy, cardiac functions, and cardiomyocyte apoptosis were monitored. RESULTS: Rats with type 2 DCM showed severe insulin resistance, left ventricular dysfunction, increased cardiomyocyte apoptosis, and reduced cardiac autophagy. Collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) ratio were significantly higher in the diabetic group than the control group. We found that trimetazidine treatment ameliorated metabolic disturbance and insulin resistance, reduced cardiomyocyte apoptosis, and restored cardiac autophagy. CVF and PVCA/LA ratio were also lower in the diabetes + trimetazidine group than the diabetic group (CVF, 4.75 ± 0.52 % vs. 11.04 ± 1.67 %, p < 0.05; PVCA/LA, 8.37 ± 0.51 vs. 17.97 ± 2.66, p < 0.05). Furthermore, trimetazidine inhibited phosphorylation of ERK and P38 MAPK to reduce myocardial fibrosis. Inhibited phosphorylation of AMPK was restored and the interaction between Bcl-2 and Beclin1 was enhanced in diabetes + trimetazidine group, resulting in the initiation of autophagy and alleviation of apoptosis. CONCLUSIONS: Early administration of trimetazidine could ameliorate diabetic cardiomyopathy by inhibiting myocardial fibrosis and cardiomyocyte apoptosis and enhancing autophagy. Therefore, trimetazidine may be a good choice in the prevention of diabetic cardiomyopathy if applied at the early stage of diabetes.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/pathology , Trimetazidine/administration & dosage , Trimetazidine/therapeutic use , Adenylate Kinase/metabolism , Animals , Blood Glucose/metabolism , Catheterization , Collagen/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/blood , Diabetic Cardiomyopathies/metabolism , Diastole , Disease Models, Animal , Fibrosis , Glucose Intolerance/blood , Glucose Intolerance/complications , Glucose Intolerance/pathology , Insulin/metabolism , Male , Phosphorylation/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects , Trimetazidine/pharmacology , Ventricular Dysfunction, Left/physiopathology
13.
Exp Mol Pathol ; 99(3): 517-23, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26407758

ABSTRACT

BACKGROUND: Vascular remodeling is an important feature of diabetic macrovascular complications. The prostaglandin F2α receptor (FP), the expression of which is upregulated by insulin resistance and diabetes, is reportedly involved in myocardial remodeling. In this study, we aimed to investigate whether the FP receptor is implicated in diabetes-induced vascular remodeling. METHODS: A type 2 diabetic rat model was induced through a high-fat diet and low-dose streptozotocin (STZ). Thirty-two rats were randomized into four groups: control, diabetes, diabetes treated with empty virus and diabetes treated with FP receptor-shRNA. Then, we evaluated the metabolic index, FP receptor expression and vascular remodeling. We used FP receptor gene silencing in vivo to investigate the role that the FP receptor plays in the pathophysiologic features of vascular remodeling. RESULTS: Diabetic rats displayed increased levels of blood glucose, cholesterol, and triglycerides, as well as severe insulin resistance and FP receptor overexpression. In addition, increased medial thickness, excessive collagen deposition and diminished elastic fibers were observed in the diabetic rats, resulting in vascular remodeling. In the FP receptor-shRNA group, the medial thickness, collagen content, elastin/collagen ratio, and collagen I/collagen III content ratio were markedly decreased. Additionally, with FP receptor gene silencing, the JNK phosphorylation level was markedly decreased. CONCLUSIONS: Silencing of the FP receptor exerts a protective effect on diabetes-induced vascular remodeling, thereby suggesting a new therapeutic target for vascular remodeling in diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Receptors, Prostaglandin/metabolism , Vascular Remodeling , Animals , Blood Glucose/physiology , Gene Silencing , Myocardium/metabolism , RNA, Small Interfering/genetics , Rats, Sprague-Dawley , Receptors, Prostaglandin/genetics , Vascular Remodeling/physiology
14.
Thromb Res ; 136(1): 45-51, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25976449

ABSTRACT

OBJECTIVE: This study aimed to assess the association between single nucleotide polymorphisms in thrombospondin-1 (THBS1), thrombospondin-2 (THBS2), thrombospondin-4 (THBS4) and coronary artery disease (CAD) risk. METHODS: Electronic databases were searched before June, 2014 to obtain articles associated with thrombospondin polymorphisms and CAD risk. After identifying case-control studies, odds ratios (ORs) and 95% confidence intervals (95% CIs) were used to pool effect sizes. Different effect models were used according to heterogeneity. Meta-regression and sensitivity analyses were performed to examine the heterogeneity source. Begg's funnel plot and Egger's test were conducted for publication bias. RESULTS: 13 studies involving 10,801 cases and 9,381 controls were included. Associations were observed between the THBS1 N700S polymorphism and CAD risk in general population(heterozygote model: OR=1.14, 95% CI: 1.03-1.26; dominant model: OR=1.13, 95% CI: 1.00-1.29), European population (heterozygote model: OR=1.13, 95% CI: 1.00-1.27) and Asian population (heterozygote model: OR=1.57, 95% CI: 1.01-2.44; dominant model: OR=1.56, 95% CI: 1.00-2.43). The THBS2 3' untranslated region (UTR) polymorphism and THBS4 A387P polymorphism were not associated with overall CAD risk. However, an association was observed between the THBS4 A387P polymorphism and CAD risk in the American population (allele model: OR=1.09, 95% CI: 1.00-1.18; homozygote model: OR=1.29, 95% CI: 1.04-1.61; recessive model: OR=1.27, 95% CI: 1.02-1.58). CONCLUSIONS: The THBS1 N700S polymorphism was associated with increased CAD risk, especially in Asian and European populations. No association was observed between the THBS2 3' UTR polymorphism and CAD risk. The THBS4 A387P polymorphism was associated with increased CAD risk in the American population.


Subject(s)
Coronary Artery Disease/genetics , Polymorphism, Single Nucleotide , Thrombospondin 1/genetics , Thrombospondins/genetics , Genetic Predisposition to Disease , Humans
15.
J Zhejiang Univ Sci B ; 16(5): 380-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25990055

ABSTRACT

BACKGROUND: Statins have proven efficacy in inhibiting the onset and progress of atherosclerosis. The effectiveness of pitavastatin in reversing carotid atherosclerosis associated with hypercholesterolemia (HC) is unknown. OBJECTIVES: To explore the simultaneous effects of pitavastatin calcium on brachial arterial flow-mediated vasodilatation (FMD), carotid intima-media thickness (IMT), and arterial stiffness (ß), three surrogate markers of atherosclerosis were studied in HC patients. METHODS: A randomized, double-blind trial was performed with 40 HC subjects who fulfilled the inclusion/exclusion criteria. Patients were given pitavastatin calcium 1 mg/d (Group 1) or 2 mg/d (Group 2) for 8 weeks. There were 20 patients in each group, and 30 gender- and age-matched healthy subjects as controls were recruited. FMD of the brachial artery, carotid IMT, and arterial stiffness indicated by ß were measured at baseline and at 8 weeks after starting pitavastatin calcium therapy using ultrasound techniques. Biochemical tests were also made on all subjects. RESULTS: At baseline, higher total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C), reduced FMD, and increased ß and IMT were observed in HC patients (P<0.001 for all) compared with controls. After 8 weeks, TC was decreased by 20.59%/27.56% and LDL-C 30.92%/35.64%, respectively, in comparison to baseline groups; the HC groups had reduced ß and improved endothelial function over the 8-week follow-up (P<0.05-0.001); nonetheless, no significant alterations of IMT were found (P>0.05). Significant negative interactions between TC/LDL and FMD (P<0.05-0.001), positive interactions between TC and IMT (P=0.003) and between TC/LDL and ß (P<0.001-0.000) were found. CONCLUSIONS: Treatment with pitavastatin calcium exerted favorable effects on endothelial function and arterial stiffness. It also improved carotid atherosclerosis in patients with HC.


Subject(s)
Atherosclerosis/drug therapy , Calcium/therapeutic use , Endothelium, Vascular/drug effects , Hypercholesterolemia/drug therapy , Quinolines/therapeutic use , Aged , Body Mass Index , Brachial Artery/drug effects , Brachial Artery/pathology , Carotid Arteries/diagnostic imaging , Carotid Arteries/drug effects , Carotid Intima-Media Thickness , Cholesterol, LDL/blood , Double-Blind Method , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Male , Middle Aged , Vascular Stiffness/drug effects , Vasodilation/drug effects
16.
Cell Physiol Biochem ; 35(3): 1151-66, 2015.
Article in English | MEDLINE | ID: mdl-25766527

ABSTRACT

BACKGROUND/AIMS: Growth arrest-specific protein 6 (Gas6) is a cytokine that can be synthesized by a variety of cell types and secreted into the extracellular matrix. Previous studies have confirmed that Gas6 is involved in certain pathophysiological processes of the cardiovascular system through binding to its receptor, Axl. In the present study, we investigated the role of Gas6 in cellular senescence and explored the mechanisms underlying its activity. METHODS: We used vascular smooth muscle cells (VSMCs) to create two cellular senescence models, one for replicative senescence (RS) and one for induced senescence (IS), to test the hypothesis that Gas6 delays senescence. RESULTS: Gas6-treated cells appear relatively younger compared with non-Gas6-treated cells. In particular, Gas6-treated cells displayed decreased staining for SA-ß-Gal, fewer G1 phase cells, and decreased levels of p16(INK4a) and p21(Cip1) expression; conversely, Gas6-treated cells displayed more S phase cells and significantly increased proliferation indexes. Furthermore, in both the IS and RS models with Gas6 treatment, the levels of PI3K, p-Akt, and p-FoxO3a decreased following Axl inhibition by R428; similarly, the levels of p-Akt and p-FoxO3a also decreased following PI3K inhibition by LY294002. CONCLUSION: Gas6/Axl signaling is essential for delaying the cellular senescence process regulated by the PI3K/Akt/FoxO signaling pathway.


Subject(s)
Cellular Senescence/genetics , Forkhead Transcription Factors/biosynthesis , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Animals , Benzocycloheptenes/administration & dosage , Cellular Senescence/drug effects , Chromones/administration & dosage , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Intercellular Signaling Peptides and Proteins/administration & dosage , Mice , Morpholines/administration & dosage , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Triazoles/administration & dosage , Axl Receptor Tyrosine Kinase
17.
Mol Med Rep ; 12(1): 463-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25738901

ABSTRACT

MicroRNA-21 (miR-21) has been found to promote cell proliferation and survival. It has also been shown to exhibit an increased expression in a number of forms of cardiovascular disease. However, the mechanisms underlying the involvement of miR-21 in atherosclerosis remain to be elucidated. In the present study, it was demonstrated that miR-21 was upregulated in a time-dependent manner in response to high-concentration glucose stimulation in Raw 264.7 macrophages. High concentrations of glucose induce macrophage apoptosis. miR-21-inhibited macrophages treated with a normal concentration of glucose exhibited increased levels of cell apoptosis and augmented levels of activated caspase-3, while cells treated with an miR-21 inhibitor and a high concentration of glucose, revealed significantly increased levels of apoptosis. In addition, inhibition of miR-21 increased mRNA and protein levels of programmed cell death 4 (PDCD4), which, by contrast, were reduced in miR-21-inhibited cells that had been treated with a high concentration of glucose. In conclusion, miR-21 is sensitive to high-concentration glucose treatment in macrophages, and appears to have a protective effect in macrophage apoptosis induced by high concentrations of glucose via PDCD4.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Apoptosis/drug effects , Atherosclerosis/genetics , MicroRNAs/biosynthesis , RNA-Binding Proteins/biosynthesis , Animals , Apoptosis Regulatory Proteins/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Glucose/administration & dosage , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , MicroRNAs/metabolism , RNA, Messenger , RNA-Binding Proteins/metabolism , Signal Transduction
18.
Acta Diabetol ; 52(4): 717-26, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25577243

ABSTRACT

AIM: Arterial stiffness is an important feature of diabetic macrovascular complications. Activin receptor-like kinase 7 (ALK7), a member of type I transforming growth factor-ß (TGF-ß) receptors, is correlated with pathogenic risks of type 2 diabetes mellitus and cardiovascular diseases and may be involved in cardiovascular remodeling. We aimed to investigate whether ALK7 is implicated in diabetes-induced aortic stiffness. METHODS: Type 2 diabetes was induced by high-fat diet and low-dose streptozotocin (STZ; 27.5 mg/kg). Forty rats were separated into four groups: control, diabetes, diabetes with empty virus and diabetes treated with ALK7-shRNA. The metabolic index, ALK 7 expression and aortic stiffness were evaluated. We used gene silencing method to investigate the role of ALK7 in the pathological development. RESULTS: Diabetic rats showed increased blood glucose, cholesterol, triglyceride levels, severe insulin resistance and ALK7 overexpression. Diabetes enhanced aortic stiffness, as demonstrated by the loss and disruption of elastic fibers as well as by an increase in collagen fibers in the aortic media. ALK7 gene silencing ameliorated metabolic hyperlipidemia and insulin resistance. With ALK7 gene silencing, collagen content, elastin to collagen ratio, as well as collagen I-to-collagen III content ratio in diabetic rats were significantly decreased. Moreover, the phosphorylation level of Smad2/3 was markedly decreased after ALK7 gene silencing. CONCLUSIONS: ALK7 gene silencing has a protective effect on diabetes-induced aortic stiffness, insulin resistance and hyperlipidemia, thus implicating a new potential therapeutic approach to diabetic macrovascular stiffness.


Subject(s)
Activin Receptors, Type I/genetics , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , RNA Interference/physiology , Vascular Stiffness/genetics , Activin Receptors, Type I/metabolism , Animals , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Angiopathies/genetics , Diabetic Angiopathies/physiopathology , Diet, High-Fat , Hyperlipidemias/complications , Hyperlipidemias/genetics , Hyperlipidemias/physiopathology , Insulin Resistance/genetics , Male , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Streptozocin , Vascular Stiffness/drug effects
19.
J Mol Med (Berl) ; 92(6): 629-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24500109

ABSTRACT

UNLABELLED: Prostaglandin F2(α)-F-prostanoid (PGF2(α)-FP) receptor is closely related to insulin resistance, which plays a causal role in the pathogenesis of diabetic cardiomyopathy (DCM). We sought to reveal whether PGF2(α)-FP receptor plays an important part in modulating DCM and the mechanisms involved. We established the type 2 diabetes rat model by high-fat diet and low-dose streptozotocin (STZ) and then evaluated its characteristics by metabolite tests, Western blot analysis for FP-receptor expression, histopathologic analyses of cardiomyocyte density and fibrosis area. Next, we used gene silencing to investigate the role of FP receptor in the pathophysiologic features of DCM. Our study showed elevated cholesterol, triglyceride, glucose, and insulin levels, severe insulin resistance, and FP-receptor overexpression in diabetic rats. The collagen volume fraction (CVF) and perivascular collagen area/luminal area (PVCA/LA) were higher in the diabetic group than the control group (CVF% 10.99 ± 0.99 vs 1.59 ± 0.18, P < 0.05; PVCA/LA% 17.07 ± 2.61 vs 2.86 ± 0.69, P < 0.05). We found that the silencing of FP receptor decreased cholesterol, triglyceride, glucose, and insulin levels and ameliorated insulin resistance. The CVF and PVCF/LA were significantly downregulated in FP-receptor short hairpin RNA (shRNA) treatment group (FP-receptor shRNA group vs vehicle group: CVF% 5.59 ± 0.92 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 4.74 ± 1.57 vs 14.79 ± 2.22, P < 0.05; FP-receptor shRNA + PGF2(α) group vs vehicle group : CVF% 5.19 ± 0.79 vs 10.97 ± 1.33, P < 0.05, PVCA/LA% 5.96 ± 1.15 vs 14.79 ± 2.22, P < 0.05, respectively). Furthermore, with FP-receptor gene silencing, the activated protein kinase C (PKC) and Rho kinase were significantly decreased, and the blunted phosphorylation of Akt was restored. FP-receptor gene silencing may exert a protective effect on DCM by improving myocardial fibrosis, suggesting a new therapeutic approach for human DCM. KEY MESSAGES: FP-receptor gene silencing improves glucose tolerance and insulin resistance in type 2 diabetes (T2D). FP-receptor gene silencing modulates the activities of PKC/Rho and Akt signaling pathways in T2D. FP-receptor gene silencing decreases collagen expression and ameliorates myocardial fibrosis in T2D. FP-receptor gene silencing protects from diabetic cardiomyopathy in T2D.


Subject(s)
Diabetic Cardiomyopathies/prevention & control , Gene Silencing/physiology , Myocardium/pathology , Receptors, Prostaglandin/metabolism , Receptors, Prostaglandin/physiology , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/therapy , Diabetic Cardiomyopathies/blood , Diet, High-Fat , Male , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin/genetics
20.
J Cell Mol Med ; 18(4): 735-48, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24467451

ABSTRACT

Our research aims to evaluate the function of the STAMP2 gene, an important trigger in insulin resistance (IR), and explore its role in macrophage apoptosis in diabetic atherosclerotic vulnerable plaques. The characteristics of diabetic mice were measured by serial metabolite and pathology tests. The level of STAMP2 was measured by RT-PCR and Western blot. The plaque area, lipid and collagen content of brachiocephalic artery plaques were measured by histopathological analyses, and the macrophage apoptosis was measured by TUNEL. Correlation of STAMP2/Akt signaling pathway and macrophage apoptosis was validated by Ad-STAMP2 transfection and STAMP2 siRNA inhibition. The diabetic mice showed typical features of IR, hyperglycaemia. Overexpression of STAMP2 ameliorated IR and decreased serum glucose level. In brachiocephalic lesions, lipid content, macrophage quantity and the vulnerability index were significantly decreased by overexpression of STAMP2. Moreover, the numbers of apoptotic cells and macrophages in lesions were both significantly decreased. In vitro, both mRNA and protein expressions of STAMP2 were increased under high glucose treatment. P-Akt was highly expressed and caspase-3 was decreased after overexpression of STAMP2. However, expression of p-Akt protein was decreased and caspase-3 was increased when STAMP2 was inhibited by siRNA. STAMP2 overexpression could exert a protective effect on diabetic atherosclerosis by reducing IR and diminishing macrophage apoptosis.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/therapy , Diabetes Mellitus, Type 2/genetics , Membrane Proteins/genetics , Plaque, Atherosclerotic/genetics , Animals , Apoptosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus, Type 2/pathology , Diet, High-Fat , Gene Expression Regulation , Insulin Resistance/genetics , Macrophages/pathology , Membrane Proteins/biosynthesis , Mice , Oncogene Protein v-akt/genetics , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/therapy , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...