Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
J Immunol Res ; 2022: 8433464, 2022.
Article in English | MEDLINE | ID: mdl-36300016

ABSTRACT

Adiponectin is a cytokine produced by adipocytes and acts as a potential cardioprotective agent and plays an important role in myocardial ischemia/reperfusion injury. In a myocardial hypoxia/reoxygenation model using neonatal rat ventricular myocytes, we investigated the contribution of adiponectin-mediated autophagy to its cardioprotective effects. Cardiomyocytes were exposed to hypoxia/reoxygenation pretreated with or without adiponectin in the presence of absence of rapamycin. Cell viability was analyzed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method. Western blotting assay was used to determine the expression levels of microtubule-associated proteins 1A/1B light chain 3B (LC3B), adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), p62/sequestosome 1, unc-51 like autophagy activating kinase 1 (ULK1), and Beclin-1. Autophagosome formation was detected by monodansylcadaverine staining. We found that hypoxia induced a time dependent decline in cardiomyocyte viability, and increase in autophagy and reoxygenation further augmented hypoxia-induced autophagy induction and consequently reduced cell viability. Adiponectin treatment alleviated hypoxia/reoxygenation-induced cellular damage and autophagy in cardiomyocytes. Adiponectin treatment also attenuated hypoxia/reoxygenation-promoted cardiomyocyte autophagy even in the presence of another autophagy stimulator rapamycin in part by inhibiting vacuolar hydron-adenosine triphosphatase. Additionally, autophagy suppression by adiponectin during hypoxia/reoxygenation was associated with the attenuated phosphorylation of AMPK and ULK1, augmented phosphorylation of mTOR, and the reduced protein expression levels of Beclin-1 in cardiomyocytes. Taken together, these results suggest that adiponectin protects ischemia/reperfusion-induced cardiomyocytes by suppressing autophagy in part through AMPK/mTOR/ULK1/Beclin-1 signaling pathway.


Subject(s)
Adiponectin , Myocytes, Cardiac , Rats , Animals , Myocytes, Cardiac/metabolism , Beclin-1/metabolism , Beclin-1/pharmacology , Autophagy-Related Protein-1 Homolog/metabolism , AMP-Activated Protein Kinases/metabolism , Cardiotonic Agents/metabolism , Cardiotonic Agents/pharmacology , Apoptosis , Autophagy , TOR Serine-Threonine Kinases/metabolism , Hypoxia/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/pharmacology , Sirolimus/pharmacology , Cytokines/metabolism , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/pharmacology , Mammals/metabolism
2.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 38(5): 504-509, 2022 Sep.
Article in Chinese | MEDLINE | ID: mdl-37088760

ABSTRACT

OBJECTIVE: To investigate the effects of p53 upregulated modulator of apoptosis (PUMA) on the apoptosis of H9C2 cardiomyocytes induced by high glucose and its mechanisms. METHODS: H9C2 cardiomyocytes were treated with 5.5mmol/L (control group) or 35 mmol/L glucose (HG group) for 6 h, 12 h, 24 h or 48 h respectively to induce apoptosis, each group sets 5 multiple wells. Apoptosis was tested by TUNEL assay. PUMA mRNA was measured by RT-PCR and protein expression was measured by Western blot assay. The mitochondrial membrane potential was detected by JC-1 method. The expressions of cleaved caspase-3 and cytochrome C (Cyt C) protein in mitochondria and cytoplasm were determined by Western blot assay. H9C2 cardiomyocytes were randomly divided into four groups, control group (5.5 mmol/L), HG (35 mmol/L) group, HG+si-scramble group(si-scramble treatment for 24 h, then 35 mmol/L high glucose treatment for 24 h) and HG-si-PUMA group (si-PUMA treatment for 24 h, then 35mmol/L high glucose treatment for 24 h). Si-PUMA was transfected into cardiomyocytes and the effects of PUMA on high glucose-induced apoptosis were studied. RESULTS: Compared with the control group, high glucose increased cardiomyocyte apoptosis and enhanced PUMA mRNA and protein expressions significantly (P<0.05 or P<0.01). Cell injury and increased PUMA expression were time-dependent and there was no significant difference between the high glucose 24 h group and the high glucose 48h group. The following experiment used high glucose 24 h as the stimulation time. The cardiomyocytes transfected with si-PUMA to inhibit PUMA expression had decreased apoptotic rate and cleaved caspase-3, increased mitochondria membrane potential and decreased Cyt C release (P<0.05 or P<0.01). There were no significant differences between the HG+si-scramble group and the high glucose group (P>0.05). CONCLUSION: PUMA mediates high glucose-induced cardiomyocyte apoptosis suggesting PUMA may be an important target gene of diabetic cardiomyopathy.


Subject(s)
Apoptosis , Myocytes, Cardiac , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Glucose/adverse effects , Glucose/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Rats
3.
Clin Appl Thromb Hemost ; 27: 10760296211029710, 2021.
Article in English | MEDLINE | ID: mdl-34254531

ABSTRACT

This study aims to determine whether dysfunctional High Density Lipoprotein (HDL) influenced the expression of scavenger receptor class B type Ⅰ (SR-B1) to determine reverse cholesterol transport. Blood samples obtained from coronary heart disease patients confirmed by angiography were collected. HDL was extracted from the blood via ultracentrifugation. Then, the HDL was injected into apoE-/- mice, and the HepG2 cells cultured with Dulbecco's modified eagle medium (DMEM) were added the HDL extracted from coronary heart disease patients. As controls, normal cases without coronary heart disease (CHD) and patients with angina pectoris and acute myocardial infarction were used. The protein expression levels of SR-B1 were detected by western blot, and the lipid accumulation levels were detected by Oil Red O staining in both tissues and cell levels. These results revealed that the HDL obtained from CHD patients downregulate the SR-B1 expression in ex vitro and in vitro studies. In addition, dysfunctional HDL may result in lower SR-B1 expression levels. The degree of SR-B1 expression levels could be relative to the degree of coronary congestion. Along with the increase in severe coronary congestion, such as myocardial infarction, the SR-B1 expression levels were lower. The dysfunctional HDL derived from coronary heart disease patients decreased the expression of SR-B1, and promoted lipid accumulation.


Subject(s)
Atherosclerosis/genetics , Coronary Artery Disease/genetics , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/metabolism , Animals , Humans , Male , Mice
4.
J Immunol Res ; 2021: 1815098, 2021.
Article in English | MEDLINE | ID: mdl-34307691

ABSTRACT

Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.


Subject(s)
Adiponectin/metabolism , Myocardial Reperfusion Injury/immunology , Myocytes, Cardiac/pathology , Animals , Animals, Newborn , Apoptosis/immunology , Cell Hypoxia/immunology , Cell Survival , Cells, Cultured , Culture Media/metabolism , Disease Models, Animal , Female , Humans , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/immunology , Necroptosis/immunology , Oxidative Stress/immunology , Pregnancy , Primary Cell Culture , Rats , Reactive Oxygen Species/metabolism
6.
Exp Anim ; 70(3): 293-301, 2021 Aug 06.
Article in English | MEDLINE | ID: mdl-33583872

ABSTRACT

This study was designed to investigate the effects of the Angiotensin II type I receptor (AT1R) shRNA on blood pressure and left ventricular remodeling in spontaneously hypertensive rats. Ten Wistar Kyoto (WKY) rats were used as a normal blood pressure control group, and 20 spontaneously hypertensive rats (SHR) were randomly divided into the experimental and hypertension control groups. The rats in the experimental group were injected with AT1R shRNA recombinant adenovirus (Ad5-AT1R-shRNA) via a tail vein, and the rats in the other two groups were injected with recombinant adenovirus (Ad5-EGFP). The systolic blood pressure (SBP) at rat arteria caudalis was measured before and after the injection, and the heart, kidney, aorta, and adrenal tissues were obtained two days after repeated injection to observe the distribution of Ad5-AT1R-shRNA under a fluorescence microscope. Before the injection of Ad5-AT1R-shRNA, the blood pressure of the experimental group and the hypertension control group was significantly higher than that of the normal blood pressure control group (P<0.01). After two injections, the blood pressure in the experimental group decreased significantly, and the duration of blood pressure reduction reached 19 days. In the experimental group, the kidney, heart, aorta, and adrenal gland tissues showed vigorous fluorescence expression under the fluorescence microscope. Repeated administration of Ad5-AT1R-shRNA has a long-lasting hypotensive effect on SHR and can significantly improve ventricular remodeling.


Subject(s)
Blood Pressure/genetics , RNA, Small Interfering/genetics , Receptor, Angiotensin, Type 1/genetics , Ventricular Remodeling/genetics , Animals , Male , RNA, Small Interfering/metabolism , Random Allocation , Rats, Inbred SHR , Rats, Inbred WKY , Receptor, Angiotensin, Type 1/metabolism
7.
Hum Cell ; 34(1): 49-59, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32959354

ABSTRACT

ATF3 (activating transcription factor 3) is a member of the mammalian activation transcription factor/cAMP-responsive element-binding (CREB) family. It plays a role in inflammation and innate immunity, and suggests that ATF3 is associated with atherosclerosis. In our study, we analyzed datasets of atherosclerosis from the NCBI-GEO (Gene Expression Omnibus) database and found that expression levels of ATF3 were lower in macrophages from ruptured atherosclerotic plaques than from stable atherosclerotic plaques. Expression levels of ATF3 correlated with the stability of atherosclerotic plaques. KEGG analysis of different expression genes (DEGs) between ruptured and stable atherosclerotic plaques was performed by Metascape database. The PI3K-AKT pathway may be a potential pathway of the formation of ruptured atherosclerotic plaques. High-fat diet-induced atherosclerosis apoE-/- mice were divided into two groups: a model group and an ATF3 overexpression (OE)-group. Tests on atherosclerotic plaques in the aortic root suggested that absence of ATF3 and increase of macrophages may be risk factors for the formation of ruptured atherosclerotic plaques. We found decreased areas of lesions in aortic roots and branches of aortic arch, as well as increased lesional content of macrophages as well as TUNEL-positive areas. Consistent with these results, we found reduced degradation and incidence of elastic plate cracks accompanied by suppressed MMPs expression and transduction pathway protein PI3K/AKT activation. These data suggest that ATF3 is a signaling molecule that mediates the progression and stability of atherosclerotic plaques. ATF3 could be a potential new biomarker for the prognosis of atherosclerosis and may be a therapeutic target to reduce atherosclerosis.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/physiology , Atherosclerosis/diagnosis , Atherosclerosis/genetics , Plaque, Atherosclerotic/genetics , Activating Transcription Factor 3/metabolism , Animals , Atherosclerosis/immunology , Atherosclerosis/therapy , Biomarkers , Gene Expression , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Plaque, Atherosclerotic/etiology , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
8.
Biochem Cell Biol ; 98(5): 583-590, 2020 10.
Article in English | MEDLINE | ID: mdl-32413267

ABSTRACT

Research has shown that some circular RNAs (circRNA) are abnormally expressed in the process of myocardial fibrosis, but the mechanism behind this was unknown. In the process of inducing cardiac fibroblast (CF) activation with TGF-ß1 or Ang II, the expression of circRNA circ_BMP2K and miR-455-3p were significantly inhibited, whereas the expression of SUMO1 was promoted. The results from our dual luciferase reporter gene assays, RIP assays, and pull-down assays show that miR-455-3p directly binds circ_BMP2K, thereby mutually promoting their expression levels. SUMO1 is a target gene of miR-455-3p, and circ_BMP2K enhances the inhibitory effects of miR-455-3p on the expression of SUMO1. Further study showed that both circ_BMP2K and miR-455-3p inhibited the expression of alpha-SMA as well as type I and type III collagen, whereas SUMO1 promoted their expression, and miR-455-3p inhibitors or overexpression of SUMO1 reversed the effects of circ_BMP2K and miR-455-3p. Circ_BMP2K and miR-455-3p inhibits cell proliferation and migration and promotes the apoptosis of CFs, but SUMO1 has the opposite effects; miR-455-3p inhibitors or overexpression of SUMO1 reverses the effects of circ_BMP2K and miR-455-3p. Thus, circ_BMP2K promotes the expression of miR-455-3p, down-regulates the expression of SUMO1, and finally, inhibits the activation, growth, and migration of CFs. These results could provide important therapeutic targets and a theoretical basis for regulating the process of myocardial fibrosis.


Subject(s)
Fibroblasts/metabolism , MicroRNAs/metabolism , RNA, Circular/metabolism , SUMO-1 Protein/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Fibroblasts/pathology , Humans , MicroRNAs/genetics , RNA, Circular/genetics , SUMO-1 Protein/genetics
9.
Exp Ther Med ; 13(6): 2757-2762, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28587337

ABSTRACT

The objective of the present study was to investigate the effect of adiponectin (APN) on macrophage reverse cholesterol transport (RCT) in adiponectin-/- knockout mice (APN-/-mice) and its possible anti-atherosclerotic mechanism. A total of 30 male APN-/-mice were randomly divided into the control group and four intervention groups. The intervention groups were treated with intraperitoneal injections of APN, at doses of 50, 150, 200 and 250 µg/(kg/day), respectively, for 4 weeks. The control group received normal saline. After 4 weeks, serum lipid levels were measured, the degree of severity of atherosclerotic lesions was observed by light microscopy, the 3H-TC (APN-/-mice treated with intraperitoneal injections of 3H-TC-labeled macrophages) radioactivity in serum, liver, and feces, and the expression of ABCA1 mRNA and protein in liver were determined. Compared with the control group, serum triglycerides, total cholesterol, and low-density lipoproteins levels in the intervention groups were significantly decreased, while high-density lipoprotein was increased. The severity of aortic atherosclerotic lesions in the intervention groups was milder than in the control group, which had obvious aortic atherosclerotic lesions, large lipid deposition on vessel walls, and the formation of atheromatous plaques. In the intervention groups, serum 3H-TC content was significantly decreased (P<0.05), but the 3H-TC content in liver and feces was significantly increased (P<0.05). The levels of ABCA1 mRNA in liver of the intervention groups were significantly increased in a dose-dependent manner. In conclusion, APN can promote RCT and intracellular cholesterol efflux by upregulating the expression of ABCA1, to delay the occurrence and development of atherosclerosis.

10.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 929-938, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28602962

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) plays a crucial role in reverse cholesterol transport and exhibits anti-atherosclerosis effects. Some microRNAs (miRs) regulate ABCA1 expression, and recent studies have shown that miR-20a/b might play a critical role in atherosclerotic diseases. Here, we attempted to clarify the potential contribution of miR-20a/b in post-transcriptional regulation of ABCA1, cholesterol efflux, and atherosclerosis. We performed bioinformatics analysis and found that miR-20a/b was highly conserved and directly bound to ABCA1 mRNA with low binding free energy. Luciferase-reporter assay also confirmed that miR-20a/b significantly reduced luciferase activity associated with the ABCA1 3' untranslated region reporter construct. Additionally, miR-20a/b decreased ABCA1 expression, which, in turn, decreased cholesterol efflux and increased cholesterol content in THP-1 and RAW 264.7 macrophage-derived foam cells. In contrast, miR-20a/b inhibitors increased ABCA1 expression and cholesterol efflux, decreased cholesterol content, and inhibited foam-cell formation. Consistent with our in vitro results, miR-20a/b-treated ApoE-/- mice showed decreased ABCA1expression in the liver and reductions of reverse cholesterol transport in vivo. Furthermore, miR-20a/b regulated the formation of nascent high-density lipoprotein and promoted atherosclerotic development, whereas miR-20a/b knockdown attenuated atherosclerotic formation. miR-20 is a new miRNA capable of targeting ABCA1 and regulating ABCA1 expression. Therefore, miR-20 inhibition constitutes a new strategy for ABCA1-based treatment of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional/physiology , 3' Untranslated Regions , Animals , Atherosclerosis/metabolism , Cell Line , Foam Cells/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Lipoproteins, HDL/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells
11.
Cell Physiol Biochem ; 38(5): 1906-14, 2016.
Article in English | MEDLINE | ID: mdl-27160732

ABSTRACT

BACKGROUND/AIMS: To detect the changes of high density lipoprotein (HDL) and its subtypes in serum of patients with coronary heart disease (CHD). METHODS: 337 hospitalized patients were selected from our hospital during August, 2014 - January, 2015, and divided into CHD group (n = 190) and control group (n = 127). Lipoprint lipoprotein analyzer was used to classify low density lipoprotein (LDL) particle size and its sub-components, as well as HDL particle size and its sub-components. The changes of the subtypes in patients with CHD were statistically analyzed. The possible mechanism was explored. RESULTS: (1) Compared with the control group, the concentration of HDL in CHD patients reduced, HDLL significantly decreased (P < 0.001), while HDLS increased (P < 0.001); (2) In the patients with HDL less than 1.04 mmol/L among CHD, all HDL subtypes reduced, but HDLL had the most significant decreased; (3) HDL and all HDL subtypes were positively correlated with apolipoprotein A-I (apoA-I), of which, HDLL had the biggest correlation with apoA-I (P < 0.001); (4) HDL subtypes had good correlation with HDL, of which, HDLM had a maximum correlation with HDL (P < 0.001). CONCLUSION: HDL maturation disorders existed in the serum of CHD patients, HDLL may be protected factor for CHD, whose decrease was closely related wit the risk increase of CHD. The cardiovascular protection function of HDLL may be related with apoA-I content.


Subject(s)
Coronary Artery Disease/pathology , Aged , Apolipoprotein A-I/blood , Body Mass Index , Case-Control Studies , Coronary Artery Disease/blood , Female , Humans , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Middle Aged , Particle Size , Triglycerides/blood
12.
Mol Med Rep ; 13(6): 5021-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27121797

ABSTRACT

Platelet activation is important in hypertension­induced cardiac inflammation and fibrosis. P-selectin expression significantly (P<0.05) increases when platelets are activated during hypertension. Although P­selectin recruits leukocytes to sites of inflammation, the role of P­selectin in cardiac inflammation and fibrosis remains to be elucidated. The present study aimed to investigate whether platelet­derived P­selectin promotes hypertensive cardiac inflammation and fibrosis. P­selectin knockout (P­sel KO) mice and wild­type (WT) C57BL/6 littermates were infused with angiotensin II (Ang II) at 1,500 ng/kg/min for 7 days and then cross­transplanted with platelets originating from either WT or P­sel KO mice. P­selectin expression was increased in the myocardium and plasma of hypertensive mice, and the P­sel KO mice exhibited significantly (P<0.05) reduced cardiac fibrosis. The fibrotic areas were markedly smaller in the hearts of P­sel KO mice compared with WT mice, as assessed by Masson's trichrome staining. In addition, α­smooth muscle actin and transforming growth factor ß1 (TGF­ß1) expression levels were decreased in the P­sel KO mice, as assessed by immunohistochemistry. Following platelet transplantation into P­sel KO mice, the number of Mac­2 (galectin­3)­ and TGF­ß1­positive cells was increased in mice that received WT platelets compared with those that received P­sel KO platelets, and the mRNA expression levels of collagen I and TGF­ß1 were also increased. The results from the present study suggest that activated platelets secrete P­selectin to promote cardiac inflammation and fibrosis in Ang II­induced hypertension.


Subject(s)
Angiotensin II/metabolism , Blood Platelets/metabolism , Myocarditis/etiology , Myocarditis/metabolism , P-Selectin/metabolism , Platelet Activation , Angiotensin II/blood , Animals , Blood Platelets/drug effects , Disease Models, Animal , Endothelial Cells/metabolism , Fibrosis , Gene Expression , Hypertension/chemically induced , Hypertension/complications , Immunohistochemistry , Male , Mice , Mice, Knockout , Myocarditis/diagnosis , Myocardium/metabolism , Myocardium/pathology , P-Selectin/genetics
13.
Free Radic Biol Med ; 89: 473-85, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26453924

ABSTRACT

Reduced levels of adiponectin (APN) contribute to cardiovascular injury in the diabetic population. Recent studies demonstrate elevated circulating APN levels are associated with endothelial dysfunction during pre-diabetes, suggesting the development of APN resistance. However, mechanisms leading to, and the role of, vascular APN resistance in endothelial dysfunction remain unidentified. The current study determined whether diabetes cause endothelial APN resistance, and by what mechanisms. Under high glucose/high lipids (HG/HL), APN-stimulated nitric oxide production by HUVEC was decreased, phosphorylation of eNOS, AMPK, and Akt was attenuated (P<0.01), and APN's anti-TNFα effect was blunted (P<0.01). APN receptor expression remained normal, whereas Cav1 expression was reduced in HG/HL cells (P<0.01). The AdipoR1/Cav1 signaling complex was dissociated in HG/HL cells. Knock-down of Cav1 inhibited APN's anti-oxidative and anti-inflammatory actions. Conversely, preventing HG/HL-induced Cav1 downregulation by Cav1 overexpression preserved APN signaling in HG/HL cells. Knock-in of a wild type Cav1 in Cav1 knock-down cells restored caveolae structure and rescued APN signaling. In contrast, knock-in of a mutated Cav1 scaffolding domain restored caveolae structure, but failed to rescue APN signaling in Cav1 knock-down cells. Finally, AdipoR1/Cav1 interaction was significantly reduced in diabetic vascular tissue, and the vasorelaxative response to APN was impaired in diabetic animals. The current study demonstrates for the first time the interaction between AdipoR1 and Cav1 is critical for adiponectin-mediated vascular signaling. The AdipoR1/Cav1 interaction is adversely affected by HG/HL, due largely to reduced Cav1 expression, supporting a potential mechanism for the development of APN resistance, contributing to diabetic endothelial dysfunction.


Subject(s)
Adiponectin/metabolism , Caveolin 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Receptors, Adiponectin/metabolism , Signal Transduction , Animals , Blood Glucose/metabolism , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Gene Knockdown Techniques , Glucose/metabolism , Humans , Immunoblotting , Immunoprecipitation , Lipids/blood , Mice , Mice, Inbred C57BL , Microscopy, Confocal , RNA, Small Interfering , Signal Transduction/physiology , Transfection
14.
Mol Med Rep ; 12(4): 5335-41, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26165515

ABSTRACT

Induction of oxidative stress has a causal role in atherosclerosis. The aim of the present study was to examine the role of lectin­like oxidized low­density lipoprotein receptor­1 (LOX­1) in oxidized low­density lipoprotein (OxLDL)­induced oxidative stress in atherosclerosis. Small interfering RNA (siRNA) technology was employed to decrease the expression of LOX­1 in mouse RAW264.7 macrophages and the effects of LOX­1 silencing on OxLDL­induced reactive oxygen species (ROS) generation and NADPH oxidase (NOX) expression were investigated. The in vivo effects of reducing LOX­1 were also examined in a mouse model (ApoE­/­) of high­fat diet­induced atherosclerosis. Compared with the control cells, OxLDL exposure led to a significant (P<0.05) increase in the intracellular levels of malondialdehyde and ROS and a significant decrease in the activity of superoxide dismutase. Delivery of LOX­1­targeting siRNA significantly (P<0.05) reversed the alterations in oxidative stress parameters induced by OxLDL. LOX­1 silencing downregulated the expression of NOX2, Rac1, p47phox and p22phox and impaired the activation of mitogen­activated protein kinases in OxLDL­treated cells. Adenoviral delivery of LOX­1 siRNA caused a significant increase in the size of the fibrous cap and a decrease in the macrophage content in lesions, compared with the control mice. Western blot analysis demonstrated that the protein expression levels of NOX1, Rac1, p47phox and p22phox in aortic lesions were significantly lower in the LOX­1 siRNA group than in the control group. LOX­1 is implicated in OxLDL­induced oxidative stress of macrophages in atherosclerosis, which in part, involves the regulation of NADPH oxidases.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Oxidative Stress , Scavenger Receptors, Class E/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cell Line , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Disease Models, Animal , Gene Expression Regulation , Gene Silencing , Lipids/blood , Lipoproteins, LDL/pharmacology , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , RNA, Small Interfering/genetics , Scavenger Receptors, Class E/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
15.
Int J Clin Exp Pathol ; 8(3): 2545-54, 2015.
Article in English | MEDLINE | ID: mdl-26045760

ABSTRACT

OBJECTIVE: This study aims to explore the mechanism of globular adiponectin inhibiting vascular calcification. METHODS: We established drug-induced rat vascular calcification model, globular adiponectin was given to observe the effect of globular Adiponectin on the degree of calcification. The markers of vascular calcification and apoptosis were also investigated. Meanwhile, the in vitro effect of globular Adiponectin on vascular calcification was also evaluated using primary cultured rat vascular smooth muscle cells. RESULTS: We found that globular adiponectin could inhibit drug-induced rat vascular calcification significantly in vivo. The apoptosis of vascular smooth muscle cells was also reduced. The possible mechanism could be the down-regulation of endoplasmic reticulum stress by globular adiponectin. Experiments in primary cultured vascular smooth muscle cells also confirmed that globular adiponectin could reduce cell apoptosis to suppress vascular calcification via inhibition of endoplasmic reticulum stress. CONCLUSIONS: This study confirmed that globular adiponectin could suppress vascular calcification; one of the mechanisms could be inhibition of endoplasmic reticulum stress to reduce cell apoptosis. It could provide an effective method in the therapy of vascular calcification-associated diseases.


Subject(s)
Adiponectin/metabolism , Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Muscle, Smooth, Vascular/pathology , Vascular Calcification/physiopathology , Adiponectin/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , Male , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Physiol Biochem ; 36(2): 599-610, 2015.
Article in English | MEDLINE | ID: mdl-25997853

ABSTRACT

BACKGROUND: Atherosclerosis is widely recognized as a complex inflammatory disease involving pathogenic immune response of T cells and antigen-presenting cells (APCs) such as dendritic cells (DCs) and macrophages. Accumulating evidence has revealed that mature DCs play critical roles in the differentiation of effector T cells into CD4+ T cells, which effectively participate in the onset of acute coronary syndrome (ACS). IFN regulatory factor (IRF)-1 has been shown to be involved in various immune processes. The role of IRF-1 in DCs in the pathogenesis of ACS has not been investigated. METHODS AND RESULTS: We examined the relative mRNA and protein expression of IRF-1 in human monocyte-derived DCs in patients with coronary artery disease (CAD). The overexpression or silencing of IRF-1 expression in DCs in patients with ACS was performed to explore the possible role of IRF-1 in the maturation and function of DCs involved in ACS. The results showed that the relative expression of IRF-1 in DCs is obviously increased in patients with ACS. The overexpression or silencing of IRF-1 expression could effectively promote or attenuate the maturation and function of DCs. In addition, we revealed that the MAPK pathway (phosphorylation of JNK, p38 and ERK1/2) might be downstream of IRF-1 signalling pathway in activation of circulating DCs in ACS patients. CONCLUSION: The present data demonstrate that IRF-1 could effectively promote the immune maturation and function of DCs in ACS patients.


Subject(s)
Acute Coronary Syndrome/immunology , Dendritic Cells/immunology , Interferon Regulatory Factor-1/immunology , Acute Coronary Syndrome/genetics , Aged , Dendritic Cells/metabolism , Female , Humans , Interferon Regulatory Factor-1/genetics , Male , Middle Aged , RNA, Messenger/genetics , Up-Regulation
17.
Cell Physiol Biochem ; 35(6): 2472-82, 2015.
Article in English | MEDLINE | ID: mdl-25967876

ABSTRACT

BACKGROUND/AIMS: Atherosclerosis is a chronic inflammatory disease. Intracellular adhesion molecule-1 (ICAM-1), vascular cellular adhesion molecule-1 (VCAM-1), and monocyte chemoattractant protein-1 (MCP-1) play important roles in inflammatory processes. P38 mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB signaling regulate ICAM-1, VCAM-1, and MCP-1 expression. Angiotensin (Ang) II upregulates ICAM-1, VCAM-1, and MCP-1 expression through the P38 MAPK and NF-κB pathways. Ang-(1-7) may oppose the actions of Ang II. We investigated whether Ang-(1-7) prevents Ang II-induced ICAM-1, VCAM-1, and MCP-1 expression in human umbilical vein endothelial cells (HUVECs). METHODS: ICAM-1, VCAM-1, and MCP-1 expression was estimated by real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA); P38, NF-κB, and p-IκB-α expression was estimated by western blotting. RESULTS: Ang-(1-7) inhibited Ang II-induced ICAM-1, VCAM-1, and MCP-1 expression and secretion in HUVECs. Ang II sharply increased P38 MAPK phosphorylation, which was inhibited by pretreatment with Ang-(1-7). Moreover, Ang-(1-7) significantly inhibited Ang II-induced IκB-α phosphorylation and NF-κB P65 nuclear translocation. The MAS receptor antagonist A-779 abolished the suppressive effects of Ang-(1-7). CONCLUSION: Ang-(1-7) attenuates Ang II-induced ICAM-1, VCAM-1, and MCP-1 expression via the MAs receptor by suppressing the P38 and NF-κB pathways in HUVECs. Ang-(1-7) might delay the progression of inflammatory diseases, including atherosclerosis.


Subject(s)
Angiotensin II/pharmacology , Angiotensin I/pharmacology , Chemokine CCL2/metabolism , Intercellular Adhesion Molecule-1/metabolism , NF-kappa B/metabolism , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , I-kappa B Proteins/metabolism , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Proto-Oncogene Mas , Signal Transduction/drug effects
18.
Mol Med Rep ; 12(1): 1387-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25779847

ABSTRACT

Angiotensin II (Ang II) and Ang-(1-7) are key effector peptides of the renin-angiotensin system. The present study aimed to investigate the effects of Ang-(1-7) on Ang II-stimulated cholesterol efflux and the associated molecular mechanisms. Differentiated THP-1 macrophages were treated with Ang II (1 µM) and/or Ang-(1-7) (10 and 100 nM) for 24 h and the cholesterol efflux and gene expression levels were assessed. Pharmacological inhibition of peroxisome proliferator-activated receptor (PPAR)γ and mitogen-activated protein kinases (MAPKs) were performed to identify the signaling pathways involved. The results demonstrated that Ang II significantly inhibited the cholesterol efflux from cholesterol-loaded THP-1 macrophages. Treatment with Ang-(1-7) led to a dose-dependent restoration of cholesterol efflux in the Ang II-treated cells. The co-treatment with Ang-(1-7) and Ang II significantly increased the expression levels of adenosine triphosphate (ATP)-binding cassette (ABC)A1 and ABCG1 compared with treatment with Ang II alone. This was coupled with increased expression levels of PPARγ and liver X receptor (LXR)α. The pharmacological inhibition of PPARγ significantly (P<0.05) eliminated the Ang-(1-7)-mediated induction of ABCA1 and ABCG1 mRNA expression. Treatment with Ang-(1-7) caused the inactivation of c-Jun N-terminal kinases (JNK) and p38 MAPK signaling in the Ang II-treated THP-1 macrophages. In addition, the inhibition of JNK or p38 MAPK signaling using specific pharmacological inhibitors mimicked the Ang-(1-7)-induced expression of PPARγ and LXRα. In conclusion, the data demonstrated that treatment with Ang-(1-7) promoted cholesterol efflux in Ang II-treated THP-1 macrophages, partly through inactivation of p38 and JNK signaling and by inducing the expression of PPARγ and LXRα. Ang (1-7) may, therefore, have therapeutic benefits for the treatment of atherosclerosis.


Subject(s)
Atherosclerosis/genetics , Cholesterol/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , Orphan Nuclear Receptors/biosynthesis , p38 Mitogen-Activated Protein Kinases/biosynthesis , ATP Binding Cassette Transporter 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/biosynthesis , Angiotensin I/administration & dosage , Angiotensin II/administration & dosage , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Gene Expression Regulation, Enzymologic , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Liver X Receptors , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Orphan Nuclear Receptors/genetics , PPAR gamma/antagonists & inhibitors , Peptide Fragments/administration & dosage , RNA, Messenger/biosynthesis , Renin-Angiotensin System , p38 Mitogen-Activated Protein Kinases/genetics
19.
JAMA ; 313(13): 1325-35, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25771069

ABSTRACT

IMPORTANCE: Uncertainty remains about the efficacy of folic acid therapy for the primary prevention of stroke because of limited and inconsistent data. OBJECTIVE: To test the primary hypothesis that therapy with enalapril and folic acid is more effective in reducing first stroke than enalapril alone among Chinese adults with hypertension. DESIGN, SETTING, AND PARTICIPANTS: The China Stroke Primary Prevention Trial, a randomized, double-blind clinical trial conducted from May 19, 2008, to August 24, 2013, in 32 communities in Jiangsu and Anhui provinces in China. A total of 20,702 adults with hypertension without history of stroke or myocardial infarction (MI) participated in the study. INTERVENTIONS: Eligible participants, stratified by MTHFR C677T genotypes (CC, CT, and TT), were randomly assigned to receive double-blind daily treatment with a single-pill combination containing enalapril, 10 mg, and folic acid, 0.8 mg (n = 10,348) or a tablet containing enalapril, 10 mg, alone (n = 10,354). MAIN OUTCOMES AND MEASURES: The primary outcome was first stroke. Secondary outcomes included first ischemic stroke; first hemorrhagic stroke; MI; a composite of cardiovascular events consisting of cardiovascular death, MI, and stroke; and all-cause death. RESULTS: During a median treatment duration of 4.5 years, compared with the enalapril alone group, the enalapril-folic acid group had a significant risk reduction in first stroke (2.7% of participants in the enalapril-folic acid group vs 3.4% in the enalapril alone group; hazard ratio [HR], 0.79; 95% CI, 0.68-0.93), first ischemic stroke (2.2% with enalapril-folic acid vs 2.8% with enalapril alone; HR, 0.76; 95% CI, 0.64-0.91), and composite cardiovascular events consisting of cardiovascular death, MI, and stroke (3.1% with enalapril-folic acid vs 3.9% with enalapril alone; HR, 0.80; 95% CI, 0.69-0.92). The risks of hemorrhagic stroke (HR, 0.93; 95% CI, 0.65-1.34), MI (HR, 1.04; 95% CI, 0.60-1.82), and all-cause deaths (HR, 0.94; 95% CI, 0.81-1.10) did not differ significantly between the 2 treatment groups. There were no significant differences between the 2 treatment groups in the frequencies of adverse events. CONCLUSIONS AND RELEVANCE: Among adults with hypertension in China without a history of stroke or MI, the combined use of enalapril and folic acid, compared with enalapril alone, significantly reduced the risk of first stroke. These findings are consistent with benefits from folate use among adults with hypertension and low baseline folate levels. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00794885.


Subject(s)
Antihypertensive Agents/therapeutic use , Enalapril/therapeutic use , Folic Acid/therapeutic use , Hypertension/drug therapy , Stroke/prevention & control , Vitamin B Complex/therapeutic use , China , Double-Blind Method , Drug Therapy, Combination , Folic Acid/blood , Humans , Kaplan-Meier Estimate , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Polymorphism, Genetic , Primary Prevention , Risk , Stroke/epidemiology
20.
Int J Clin Exp Pathol ; 8(1): 450-7, 2015.
Article in English | MEDLINE | ID: mdl-25755733

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) plays a crucial role in reverse cholesterol transport and anti-atherosclerosis. Liver X receptor alpha (LXRα) can stimulate cholesterol efflux through ABCA1. It has been well known that adiponectin has cardiovascular protection. In this study, we attempted to clarify the effect of adiponectin on expression of ABCA1, and explored the role of LXRα in the regulation of ABCA1 in RAW 264.7 macrophages. Our results showed that adiponectin increased ABCA1 expression at both the mRNA and protein levels in a dose-dependent and time-dependent manner. Consequently, adiponectin promoted cholesterol efflux and decreased cholesterol content in RAW 264.7 macrophages. Moreover, adiponectin up-regulated the expression of LXRα in a dose-dependent and time-dependent manner in RAW 264.7 macrophages. LXRα small interfering RNA completely abolished the promotion effects of adiponectin. In summary, adiponectin up-regulates ABCA1 expression via the LXRα pathway in RAW 264.7 macrophages. This novel insight could prove useful for developing new treatment strategies for cardiovascular diseases.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Adiponectin/pharmacology , Macrophages/drug effects , Orphan Nuclear Receptors/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , ATP Binding Cassette Transporter 1/genetics , Animals , Cell Line , Cholesterol/metabolism , Dose-Response Relationship, Drug , Liver X Receptors , Macrophages/metabolism , Mice , RNA, Small Interfering
SELECTION OF CITATIONS
SEARCH DETAIL
...