Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Med Rep ; 29(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38275129

ABSTRACT

Hyperphosphatemia or severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) infection can promote cardiovascular adverse events in patients with chronic kidney disease. Hyperphosphatemia is associated with elevated inflammation and sterol regulatory element binding protein 2 (SREBP2) activation, but the underlying mechanisms in SARS­CoV­2 that are related to cardiovascular disease remain unclear. The present study aimed to elucidate the role of excess inorganic phosphate (PI) in SARS­CoV­2 N protein­induced NLRP3 inflammasome activation and the underlying mechanisms in vascular smooth muscle cells (VSMCs). The expression levels of SARS­CoV­2 N protein, SREBP cleavage­activating protein (SCAP), mature N­terminal SREBP2, NLRP3, procaspase­1, cleaved caspase­1, IL­1ß and IL­18 were examined by western blotting. The expression levels of SREBP2, HMG­CoA reductase, HMGCS1, low density lipoprotein receptor, proprotein convertase subtilisin/kexin type 9 (PCSK9), SREBP1c, fatty acid synthase, stearyl coenzyme A desaturase 1, acetyl­CoA carboxylase α and ATP­citrate lyase were determined by reverse transcription­quantitative PCR. The translocation of SCAP or NLRP3 from the endoplasmic reticulum to the Golgi was detected by confocal microscopy. The results showed that excess PI promoted SCAP­SREBP and NLRP3 complex translocation to the Golgi, potentially leading to NLRP3 inflammasome activation and lipogenic gene expression. Furthermore, PI amplified SARS­CoV­2 N protein­induced inflammation via the SCAP­SREBP pathway, which facilitates NLRP3 inflammasome assembly and activation. Inhibition of phosphate uptake with phosphonoformate sodium alleviated NLRP3 inflammasome activation and reduced SREBP­mediated lipogenic gene expression in VSMCs stimulated with PI and with SARS­CoV­2 N protein overexpression. Inhibition of SREBP2 or small interfering RNA­induced silencing of SREBP2 effectively suppressed the effect of PI and SARS­CoV­2 N protein on NLRP3 inflammasome activation and lipogenic gene expression. In conclusion, the present study identified that PI amplified SARS­CoV­2 N protein­induced NLRP3 inflammasome activation and lipogenic gene expression via the SCAP­SREBP signaling pathway.


Subject(s)
COVID-19 , Hyperphosphatemia , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proprotein Convertase 9/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , SARS-CoV-2/metabolism , Phosphates , Sterol Regulatory Element Binding Protein 1/metabolism , Signal Transduction , Inflammation
2.
Tissue Cell ; 86: 102276, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37979395

ABSTRACT

The sterol regulatory element-binding protein (SREBP) activation and cytokine level were significantly increased in coronavirus disease-19. The NLRP3 inflammasome is an amplifier for cellular inflammation. This study aimed to elucidate the modulatory effect of SARS-CoV-2 nucleocapsid protein (SARS-CoV-2 NP) on trimethylamine N-oxide (TMAO)-induced lipogenesis and NLRP3 inflammasome activation and the underlying mechanisms in vascular smooth muscle cells (VSMCs). Our data indicated that SARS-CoV-2 NP activates the dissociation of the SREBP cleavage activating protein (SCAP) from the endoplasmic reticulum, resulting in SREBP activation, increased lipogenic gene expression, and NLRP3 inflammasome activation. TMAO was applied to VSMC-induced NLRP3 inflammasome by promoting the SCAP-SREBP complex endoplasmic reticulum-to-Golgi translocation, which facilitates directly binding of SARS-CoV-2 NP to the NLRP3 protein for NLRP3 inflammasome assembly. SARS-CoV-2 NP amplified the TMAO-induced lipogenic gene expression and NLRP3 inflammasome. Knockdown of SCAP-SREBP2 can effectively reduce lipogenic gene expression and alleviate NLRP3 inflammasome-mediated systemic inflammation in VSMCs stimulated with TMAO and SARS-CoV-2 NP. These results reveal that SARS-CoV-2 NP amplified TMAO-induced lipogenesis and NLRP3 inflammasome activation via priming the SCAP-SREBP signaling pathway.


Subject(s)
COVID-19 , Methylamines , Sterol Regulatory Element Binding Proteins , Humans , Sterol Regulatory Element Binding Proteins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , SARS-CoV-2 , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction , Inflammation , Nucleocapsid Proteins
3.
Mol Med Rep ; 28(1)2023 Jul.
Article in English | MEDLINE | ID: mdl-37203402

ABSTRACT

Chronic inflammation is a key factor that accelerates the progression of inflammatory vascular disease. Hydrogen sulfide (H2S) has potent anti­inflammatory effects; however, its underlying mechanism of action has not been fully elucidated. The present study aimed to investigate the potential effect of H2S on sirtuin 1 (SIRT1) sulfhydration in trimethylamine N­oxide (TMAO)­induced macrophage inflammation, and its underlying mechanism. Pro­inflammatory M1 cytokines (MCP­1, IL­1ß, and IL­6) and anti­inflammatory M2 cytokines (IL­4 and IL­10) were detected by RT­qPCR. CSE, p65 NF­κB, p­p65 NF­κB, IL­1ß, IL­6 and TNF­α levels were measured by Western blot. The results revealed that cystathionine γ­lyase protein expression was negatively associated with TMAO­induced inflammation. Sodium hydrosulfide (a donor of H2S) increased SIRT1 expression and inhibited the expression of inflammatory cytokines in TMAO­stimulated macrophages. Furthermore, nicotinamide, a SIRT1 inhibitor, antagonized the protective effect of H2S, which contributed to P65 NF­κB phosphorylation and upregulated the expression of inflammatory factors in macrophages. H2S ameliorated TMAO­induced activation of the NF­κB signaling pathway via SIRT1 sulfhydration. Moreover, the antagonistic effect of H2S on inflammatory activation was largely eliminated by the desulfhydration reagent dithiothreitol. These results indicated that H2S may prevent TMAO­induced macrophage inflammation by reducing P65 NF­κB phosphorylation via the upregulation and sulfhydration of SIRT1, suggesting that H2S may be used to treat inflammatory vascular diseases.


Subject(s)
Hydrogen Sulfide , Humans , Cystathionine gamma-Lyase/metabolism , Hydrogen Sulfide/pharmacology , Inflammation/metabolism , Interleukin-6 , Macrophages/metabolism , NF-kappa B , Sirtuin 1/metabolism
4.
Int J Mol Med ; 43(3): 1321-1330, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30664197

ABSTRACT

Fibroblast growth factor (FGF)­21, a member of the family of FGFs, exhibits protective effects against myocardial ischemia and ischemia/reperfusion injury; it is also an enhancer of autophagy. However, the mechanisms underlying the protective role of FGF­21 against cardiomyocyte hypoxia/reoxygenation (H/R) injury remain unclear. The present study aimed to investigate the effect of FGF­21 on H9c2 cardiomyocyte injury induced by H/R and the mechanism associated with changes in autophagy. Cultured H9c2 cardiomyocytes subjected to hypoxia were treated with a vehicle or FGF­21 during reoxygenation. The viability of H9c2 rat cardiomyocytes was measured using Cell Counting Kit­8 and trypan blue exclusion assays. The contents of creatine kinase (CK) and creatine kinase isoenzymes (CK­MB), cardiac troponin I (cTnT), cardiac troponin T (cTnI) and lactate dehydrogenase (LDH) in culture medium were detected with a CK, CK­MB, cTnT, cTnI and LDH assay kits. The protein levels were examined by western blot analysis. Autophagic flux was detected by Ad­mCherry­GFP­LC3B autophagy fluorescent adenovirus reagent. The results indicated that FGF­21 alleviated H/R­induced H9c2 myocardial cell injury and enhanced autophagic flux during H/R, and that this effect was antagonized by co­treatment with 3­methyladenine, an autophagy inhibitor. Furthermore, FGF­21 increased the expression levels of Beclin­1 and Vps34 proteins, but not of mechanistic target of rapamycin. These data indicate that FGF­21 treatment limited H/R injury in H9c2 cardiomyocytes by promoting autophagic flux through upregulation of the expression levels of Beclin­1 and Vps34 proteins.


Subject(s)
Autophagy , Fibroblast Growth Factors/metabolism , Hypoxia/metabolism , Myocytes, Cardiac/metabolism , Oxygen/metabolism , Animals , Biomarkers , Cell Line , Cell Survival , Cytoprotection , Fibroblast Growth Factors/pharmacology , Genes, Reporter , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Rats , Signal Transduction
5.
Biomed Pharmacother ; 104: 36-44, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29758414

ABSTRACT

Elevated plasma low-density lipoprotein cholesterol (LDL-C) is an important risk factor for cardiovascular diseases. Statins are the most widely used therapy for patients with hyperlipidemia. However, a significant residual cardiovascular risk remains in some patients even after maximally tolerated statin therapy. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a new pharmacologically therapeutic target for decreasing LDL-C. PCSK9 reduces LDL intake from circulation by enhancing LDLR degradation and preventing LDLR recirculation to the cell surface. Moreover, PCSK9 inhibitors have been approved for patients with either familial hypercholesterolemia or atherosclerotic cardiovascular disease, who require additional reduction of LDL-C. In addition, PCSK9 inhibition combined with statins has been used as a new approach to help reduce LDL-C levels in patients with either statin intolerance or unattainable LDL goal. This review will discuss the emerging anti-PCSK9 therapies in the regulation of cholesterol metabolism and atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Lipid Metabolism/physiology , Proprotein Convertase 9/metabolism , Animals , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Atherosclerosis/drug therapy , Cholesterol, LDL/metabolism , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/metabolism , Lipid Metabolism/drug effects
6.
Mol Med Rep ; 18(1): 675-683, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29845269

ABSTRACT

Autophagy is a maintenance process for recycling long-lived proteins and cytoplasmic organelles. The level of this process is enhanced during ischemia/reperfusion (I/R) injury. Autophagy can trigger survival signaling in myocardial ischemia, whereas defective autophagy during reperfusion is detrimental. Autophagy can be regulated through multiple signaling pathways in I/R, including Beclin­1/class III phosphatidylinositol­3 kinase (PI­3K), adenosine monophosphate activated protein kinase/mammalian target of rapamycin (mTOR), and PI­3K/protein kinase B/mTOR pathways, which consequently lead to different functions. Thus, autophagy has both protective and detrimental functions, which are determined by different signaling pathways and conditions. Targeting the activation of autophagy can be a promising new therapeutic strategy for treating cardiovascular disease.


Subject(s)
Autophagy , Myocardial Reperfusion Injury/physiopathology , Signal Transduction , Animals , Humans , Myocardial Reperfusion Injury/metabolism
8.
Exp Ther Med ; 12(2): 1113-1118, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27446329

ABSTRACT

Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it can induce severe cytotoxicity, which limits its clinical application. In the present study, the effects of resveratrol (RES) on sirtuin 1 (SIRT1) activation in mediating DOX-induced cytotoxicity in H9c2 cardiac cells was investigated. H9c2 cells were exposed to 5 µM DOX for 24 h to establish a model of DOX cardiotoxicity. Apoptosis of H9c2 cardiomyocytes was assessed using the MTT assay and Hoechst nuclear staining. The results demonstrated that pretreating H9c2 cells with RES prior to the exposure of DOX resulted in increased cell viability and a decreased quantity of apoptotic cells. Western blot analysis demonstrated that DOX decreased the expression level of SIRT1. These effects were significantly alleviated by co-treatment with RES. In addition, the results demonstrated that DOX administration amplified forkhead box O1 (FoxO1) and P53 expression levels in H9c2 cells. RES was also found to protect against DOX-induced increases of FoxO1 and P53 expression levels in H9c2 cells. Furthermore, the protective effects of RES were arrested by the SIRT1 inhibitor nicotinamide. In conclusion, the results demonstrated that RES protected H9c2 cells against DOX-induced injuries via SIRT1 activation.

9.
Int J Mol Med ; 37(6): 1661-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27081862

ABSTRACT

Doxorubicin (DOX) is an efficient drug used in cancer therapy that also produces reactive oxygen species (ROS) that induces severe cytotoxicity, which limits its clinical application. Hydrogen sulfide (H2S), a novel gasotransmitter, has been shown to exert cardioprotective effects. The present study aimed to determine whether exogenous H2S protects H9c2 cardiac cells against DOX-induced cytotoxicity and whether these protective effects are mediated through the PI3K/Akt/FoxO3a pathway. The H9c2 cardiac cells were exposed to 5 µM DOX for 24 h to establish a model of DOX-induced cardiotoxicity. The results showed that the treatment of H9c2 cardiac cells with sodium hydrosulfide (NaHS) for 30 min prior to DOX exposure markedly attenuated the phosphorylation of Akt and FoxO3a. Notably, pre-treatment of the H9c2 cells with NaHS significantly attenuated the nuclear localization of FoxO3a as well as the apoptosis of H9c2 cells induced by DOX. The treatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of ROS, prior to DOX exposure, also markedly increased the phosphorylation of Akt and FoxO3a which was inhibited by DOX alone. Furthermore, pre-treatment with LY294002, a selective inhibitor of PI3K/Akt, reversed the protective effect of H2S against DOX-induced injury of cardiomyocytes, as demonstrated by an increased number of apoptotic cells, a decrease in cell viability and the reduced phosphorylation of Akt and FoxO3a. These findings suggested that exogenous H2S attenuates DOX-induced cytotoxic effects in H9c2 cardiac cells through the PI3K/Akt/FoxO3a pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Doxorubicin/antagonists & inhibitors , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/drug effects , Sulfides/pharmacology , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Cardiotoxicity/prevention & control , Cell Line , Chromones/pharmacology , Doxorubicin/pharmacology , Forkhead Box Protein O3/genetics , Forkhead Box Protein O3/metabolism , Gene Expression Regulation , Models, Biological , Morpholines/pharmacology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction , Sulfides/chemistry
10.
Mol Cell Biochem ; 414(1-2): 57-66, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26899709

ABSTRACT

Cardiovascular disease is a growing major global public health problem. Oxidative stress is regarded as one of the key regulators of pathological physiology, which eventually leads to cardiovascular disease. However, mechanisms by which FGF-2 rescues cells from oxidative stress damage in cardiovascular disease is not fully elucidated. Herein this study was designed to investigate the protective effects of FGF-2 in H2O2-induced apoptosis of H9c2 cardiomyocytes, as well as the possible signaling pathway involved. Apoptosis of H9c2 cardiomyocytes was induced by H2O2 and assessed using methyl thiazolyl tetrazolium assay, Hoechst, and TUNEL staining. Cells were pretreated with PI3K/Akt inhibitor LY294002 to investigate the possible PI3K/Akt pathways involved in the protection of FGF-2. The levels of p-Akt, p-FoxO3a, and Bim were detected by immunoblotting. Stimulation with H2O2 decreased the phosphorylation of Akt and FoxO3a, and induced nuclear localization of FoxO3a and apoptosis of H9c2 cells. These effects of H2O2 were abrogated by pretreatment with FGF-2. Furthermore, the protective effects of FGF-2 were abolished by PI3K/Akt inhibitor LY294002. In conclusion, our data suggest that FGF-2 protects against H2O2-induced apoptosis of H9c2 cardiomyocytes via activation of the PI3K/Akt/FoxO3a pathway.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 2/physiology , Forkhead Box Protein O3/metabolism , Hydrogen Peroxide/toxicity , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Bcl-2-Like Protein 11/metabolism , Cell Line , Phosphorylation , Protein Transport , Rats
11.
Oncol Lett ; 11(2): 1499-1505, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26893768

ABSTRACT

The hepatocyte growth factor (HGF)/c-Met signaling pathway results in cancer cell scattering and invasion, and has been reported to participate in several types of cancer, including prostate and colorectal cancer. The downstream phosphorylation cascade of HGF, particularly the mitogen-activated protein kinase and phosphoinositide 3-kinase/AKT signaling pathway, regulates epithelial-mesenchymal transition (EMT). However, the mechanism by which these signaling pathways govern EMT, and whether certain kinases are able to respond to specific EMT effectors, remains to be elucidated. In the present study, an increase in the levels of vimentin, rather than co-regulation of certain EMT marker proteins, was observed in response to HGF-induced EMT in DU145 prostate cancer cells. In addition, it was observed that curcumin abrogated HGF-induced DU145 cell scattering and invasion. Furthermore, curcumin was able to effectively inhibit the HGF-induced increase in the levels of vimentin by downregulating the expression of phosphorylated c-Met, extracellular signal-regulated kinase and Snail. In conclusion, the results of the present study demonstrated that curcumin was able to reverse HGF-induced EMT, possibly by inhibiting c-Met expression in DU145 prostate cancer cells.

12.
Mol Med Rep ; 13(2): 1281-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26675978

ABSTRACT

Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it has severe cardiotoxic side effects. The aim of the present study was to investigate the effects of resveratrol on the adenosine monophosphate-activated protein kinase (AMPK)/P53 pathway in mediating DOX-induced cytotoxicity. H9c2 cells were exposed to 5 µM DOX for 24 h to establish a model of DOX-induced cardiotoxicity. DOX administration amplified P53 and B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax) expression and decreased Bcl-2 expression in H9c2 cells. Resveratrol increased the cell viability and decreased the apoptotic rate. In addition, resveratrol markedly increased the phosphorylation of AMPK. Of note, resveratrol protected against DOX-induced increases of P53 and Bax and also prevented the downregulation of Bcl-2 in H9c2 cells. Furthermore, AMPK inhibitor Compound C abolished the protective effects of resveratrol. The results of the present study therefore indicated that resveratrol protected H9c2 cells from DOX-induced apoptosis via the AMPK/P53 pathway.


Subject(s)
AMP-Activated Protein Kinases/genetics , Apoptosis/drug effects , Myocytes, Cardiac/drug effects , Tumor Suppressor Protein p53/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Doxorubicin/administration & dosage , Gene Expression Regulation/drug effects , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Resveratrol , Signal Transduction/drug effects , Stilbenes/administration & dosage , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
13.
Mol Med Rep ; 13(1): 367-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26573464

ABSTRACT

Doxorubicin (DOX) is a widely used chemotherapeutic agent, which can give rise to severe cardiotoxicity, limiting its clinical use. Preliminary evidence suggests that hydrogen sulfide (H2S) may exert protective effects on DOX­induced cardiotoxicity. Therefore, the aim of the present study was to investigate whether peroxiredoxin III is involved in the cardioprotection of H2S against DOX­induced cardiotoxicity. The results demonstrated that DOX not only markedly induced injuries, including cytotoxicity and apoptosis, it also increased the expression levels of peroxiredoxin III. Notably, pretreatment with sodium hydrosulfide significantly attenuated the DOX­induced decrease in cell viability and increase in apoptosis, and also reversed the increased expression levels of peroxiredoxin III in H9c2 cardiomyocytes. In addition, pretreatment of the H9c2 cells with N­acetyl­L­cysteine, a scavenger of reactive oxygen species, prior to exposure to DOX markedly decreased the expression levels of peroxiredoxin III. In conclusion, the results of the present study suggested that exogenous H2S attenuates DOX­induced cardiotoxicity by inhibiting the expression of peroxiredoxin III in H9c2 cells. In the present study, the apoptosis of H9c2 cardiomyocytes was assessed using an methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and cystathionine-γ-lyase were examined by western blotting.


Subject(s)
Cardiotoxicity/metabolism , Doxorubicin/toxicity , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/metabolism , Peroxiredoxin III/metabolism , Acetylcysteine/pharmacology , Animals , Cardiotonic Agents/pharmacology , Cardiotoxicity/pathology , Cell Death/drug effects , Cystathionine gamma-Lyase/metabolism , Cytoprotection/drug effects , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Time Factors
14.
Mol Med Rep ; 13(2): 1689-94, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26709007

ABSTRACT

Resveratrol is a polyphenolic compound found in wine, which is mainly produced by the grapevine and exerts chemopreventive effects against hepatocellular carcinoma. However, the underlying molecular mechanisms have remained to be fully elucidated. The present study assessed whether resveratrol-induced apoptosis was mediated via the activation of the forkhead box O3a (FoxO3a) transcription factor. It was demonstrated that resveratrol treatment induced apoptosis in HepG2 cells, and that this pro-apoptotic effect was accompanied with increases in the expression of apoptotic protein Bim. Following resveratrol treatment, Akt-mediated phosphorylation of FoxO3a was observed to be diminished in HepG2 cells. Furthermore, resveratrol enhanced the nuclear levels of FoxO3a and mediated neuronal death via Bim. The present study demonstrated that resveratrol induced apoptosis in HepG2 cells through activation of the transcription factor FoxO3a and increasing the expression of Bim protein.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Forkhead Transcription Factors/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Bcl-2-Like Protein 11 , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Forkhead Box Protein O3 , Hep G2 Cells , Humans , Phosphorylation/drug effects , Protein Transport/drug effects , Resveratrol
15.
Exp Ther Med ; 10(4): 1515-1520, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26622517

ABSTRACT

Doxorubicin (DOX) is an efficient drug used in cancer therapy; however, it produces reactive oxygen species (ROS) that induce severe cytotoxicity, limiting its clinical application. The aim of the present study was to investigate the role of peroxiredoxin III (Prx III) in DOX-induced H9c2 cell injuries. Following DOX treatment, the expression of phosphorylated-FoxO3a (p-FoxO3a) was decreased and Prx III expression was increased in H9c2 cells. In order to detect whether oxidative stress was involved in the induction of Prx III expression by FoxO3a, exogenous H2O2 was used to induce oxidative stress in the H9c2 cells. Apoptosis of H9c2 cardiomyocytes was assessed using methyl thiazolyl tetrazolium assay and Hoechst staining. The levels of Prx III and p-FoxO3a were evaluated using western blot analysis. As expected, H2O2 was found to mimic the effect of DOX, decreasing the expression of p-FoxO3a and increasing the expression of Prx III. In addition, the study evaluated whether the transcription factor FoxO3a was essential for the expression of Prx III. Pretreatment of H9c2 cells with N-acetyl-L-cysteine (NAC), a scavenger of ROS, prior to exposure to DOX dramatically increased the phosphorylation of FoxO3a and led to a marked reduction in Prx III expression in the H9c2 cells. In conclusion, the results of the current study suggest that FoxO3a mediates the expression of Prx III in DOX-induced injuries.

16.
Mol Med Rep ; 12(5): 6841-8, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26299281

ABSTRACT

Doxorubicin (DOX) is a potent and available antitumor therapeutic agent; however, its clinical application is limited due to its cardiotoxicity. Preliminary evidence suggests that hydrogen sulfide (H2S) may exert protective effects on DOX­induced cardiotoxicity. Therefore, the aim of the present study was to investigate whether the extracellular signal­regulated kinase (ERK) 1/2 signaling pathway is involved in the cardioprotection of H2S against DOX­induced cardiotoxicity. The present study demonstrated that pretreatment with sodium hydrosulfide (NaHS; a donor of H2S) prior to DOX exposure attenuated the decreased cell viability, the increased apoptosis rate and the intracellular accumulation of reactive oxygen species (ROS) in H9c2 cardiac myocytes. Exposure of H9c2 cardiac myocytes to DOX upregulated the expression levels of phosphorylated ERK1/2, which had been reduced by pretreatment with NaHS or N­acetyl­L­cysteine, a ROS scavenger. In addition, H2S upregulated the anti­apoptotic protein, Bcl­2 and downregulated the pro­apoptotic protein, Bax. Notably, U0126, a selective inhibitor of ERK1/2, was observed to mimic the above­mentioned cytoprotective activity of H2S. In conclusion, these findings indicate that H2S attenuates DOX­induced cardiotoxicity by inhibiting ROS-mediated activation of ERK1/2 in H9c2 cardiac myocytes.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cardiotonic Agents/pharmacology , Doxorubicin/toxicity , Hydrogen Sulfide/pharmacology , Myocytes, Cardiac/metabolism , Acetylcysteine/pharmacology , Animals , Cell Line , Cystathionine gamma-Lyase/metabolism , Drug Evaluation, Preclinical , Enzyme Activation , Free Radical Scavengers/pharmacology , Heart Diseases/chemically induced , Heart Diseases/prevention & control , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/drug effects , Oxidative Stress , Phosphorylation , Protein Processing, Post-Translational , Rats , Reactive Oxygen Species/metabolism
18.
Mol Med Rep ; 12(4): 5197-202, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26134131

ABSTRACT

Doxorubicin (DOX) is a potent and currently available antitumor therapeutic agent; however, its clinical application is limited by the occurrence of cardiotoxicity. Preliminary evidence indicates that hydrogen sulfide (H2S) may exert protective effects against DOX cardiotoxicity. Therefore, the aim of the present study was to investigate whether calreticulin (CRT) is involved in the cardioprotection of H2S against DOX­induced cardiotoxicity. DOX was observed to markedly induce injuries, including cytotoxicity and apoptosis, and also enhance the expression level of CRT. Notably, pretreatment of H9c2 cells with sodium hydrosulfide (a donor of H2S) significantly attenuated the decreased cell viability, the increased apoptosis rate and the increased expression level of CRT in H9c2 cells. In addition, pretreatment of H9c2 cells with N­acetyl­L­cysteine, a scavenger of reactive oxygen species (ROS) prior to exposure to DOX, markedly decreased the expression of CRT. These results indicate that exogenous H2S attenuates DOX­induced cardiotoxicity by inhibiting CRT expression in H9c2 cardiac cells.


Subject(s)
Antineoplastic Agents/pharmacology , Calreticulin/genetics , Doxorubicin/pharmacology , Gene Expression Regulation/drug effects , Hydrogen Sulfide/pharmacology , Antineoplastic Agents/adverse effects , Apoptosis/drug effects , Apoptosis/genetics , Cardiotoxicity , Cell Line , Doxorubicin/adverse effects , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism
19.
DNA Cell Biol ; 34(9): 561-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26102194

ABSTRACT

Curcumin, a traditional Chinese derivative from the rhizomes of Curcuma longa, is beneficial to health by modulating lipid metabolism and suppressing atherogenesis. A key part of atherosclerosis is the failure of macrophages to restore their cellular cholesterol homeostasis and the formation of foam cells. In this study, results showed that curcumin dramatically increased the expression of ATP-binding cassette transporter 1 (ABCA1), promoted cholesterol efflux from THP-1 macrophage-derived foam cells, and reduced cellular cholesterol levels. Curcumin activated AMP-activated protein kinase (AMPK) and SIRT1, and then activated LXRα in THP-1 macrophage-derived foam cells. Inhibiting AMPK/SIRT1 activity by its specific inhibitor or by small interfering RNA could inhibit LXRα activation and abolish curcumin-induced ABCA1 expression and cholesterol efflux. Thus, curcumin enhanced cholesterol efflux by upregulating ABCA1 expression through activating AMPK-SIRT1-LXRα signaling in THP-1 macrophage-derived foam cells. This study describes a possible mechanism for understanding the antiatherogenic effects of curcumin on attenuating the progression of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Curcumin/pharmacology , Foam Cells/metabolism , Hypolipidemic Agents/pharmacology , ATP Binding Cassette Transporter 1/genetics , Adenylate Kinase/metabolism , Cell Differentiation , Cell Line , Cell Survival/drug effects , Foam Cells/drug effects , Humans , Liver X Receptors , Orphan Nuclear Receptors/metabolism , Signal Transduction , Sirtuin 1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...