101
|
Ferulic acid inhibits H2O2-induced oxidative stress and inflammation in rat vascular smooth muscle cells via inhibition of the NADPH oxidase and NF-κB pathway. Int Immunopharmacol 2015; 28:1018-25. [DOI: 10.1016/j.intimp.2015.07.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 07/16/2015] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
|
102
|
Gupta MK, Lee SH, Crowder SW, Wang X, Hofmeister LH, Nelson CE, Bellan LM, Duvall CL, Sung HJ. Oligoproline-derived nanocarrier for dual stimuli-responsive gene delivery. J Mater Chem B 2015; 3:7271-7280. [PMID: 32262835 PMCID: PMC9088177 DOI: 10.1039/c5tb00988j] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gene therapy is a promising method for the treatment of vascular disease; however, successful strategies depend on the development of safe and effective delivery technologies with specific targeting to a diseased point of vasculature. Reactive oxygen species (ROS) are overproduced by vascular smooth muscle cells (VSMCs) at critical stages of atherosclerosis progression. Therefore, ROS were exploited as a stimulus for vascular targeted gene delivery in this study. A combination of bio-conjugation methods and controlled reverse addition-fragmentation chain-trasfer (RAFT) polymerization was utilized to synthesize a new ROS-cleavable, pH-responsive mPEG113-b-CP5K-b-PDMAEMA42-b-P(DMAEMA22-co-BMA40-co-PAA24) (PPDDBP) polymer as a nanocarrier for plasmid DNA (pDNA) delivery. The ros degradability of PPDDBP polymers was confirmed by SIN-1-mediated cleavage of CP5K peptide linkers through a shift in GPC chromatogram with an appearance of mPEG shoulder peak and an increase in zeta potential (ζ). The polyplex nanocarrier also demonstrated effective PDNA loading, serum stability, and hemocompatibility, indicating its excellent performance under physiological conditions. The polyplexes demonstrated ideal pH responsiveness for endosomal escape and effective ROS responsiveness for improved targeting in an in vitro model of pathogenic VSMCs in terms of both uptake and expression of reporter gene. These data suggest this novel nanocarrier polyplex system is a promising gene delivery tool for preventing or treating areas of high ROS, such as atherosclerotic lesions.
Collapse
Affiliation(s)
- Mukesh K Gupta
- Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Chaplin NL, Nieves-Cintrón M, Fresquez AM, Navedo MF, Amberg GC. Arterial Smooth Muscle Mitochondria Amplify Hydrogen Peroxide Microdomains Functionally Coupled to L-Type Calcium Channels. Circ Res 2015; 117:1013-23. [PMID: 26390880 DOI: 10.1161/circresaha.115.306996] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/18/2015] [Indexed: 01/21/2023]
Abstract
RATIONALE Mitochondria are key integrators of convergent intracellular signaling pathways. Two important second messengers modulated by mitochondria are calcium and reactive oxygen species. To date, coherent mechanisms describing mitochondrial integration of calcium and oxidative signaling in arterial smooth muscle are incomplete. OBJECTIVE To address and add clarity to this issue, we tested the hypothesis that mitochondria regulate subplasmalemmal calcium and hydrogen peroxide microdomain signaling in cerebral arterial smooth muscle. METHODS AND RESULTS Using an image-based approach, we investigated the impact of mitochondrial regulation of L-type calcium channels on subcellular calcium and reactive oxygen species signaling microdomains in isolated arterial smooth muscle cells. Our single-cell observations were then related experimentally to intact arterial segments and to living animals. We found that subplasmalemmal mitochondrial amplification of hydrogen peroxide microdomain signaling stimulates L-type calcium channels, and that this mechanism strongly impacts the functional capacity of the vasoconstrictor angiotensin II. Importantly, we also found that disrupting this mitochondrial amplification mechanism in vivo normalized arterial function and attenuated the hypertensive response to systemic endothelial dysfunction. CONCLUSIONS From these observations, we conclude that mitochondrial amplification of subplasmalemmal calcium and hydrogen peroxide microdomain signaling is a fundamental mechanism regulating arterial smooth muscle function. As the principle components involved are fairly ubiquitous and positioning of mitochondria near the plasma membrane is not restricted to arterial smooth muscle, this mechanism could occur in many cell types and contribute to pathological elevations of intracellular calcium and increased oxidative stress associated with many diseases.
Collapse
Affiliation(s)
- Nathan L Chaplin
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Madeline Nieves-Cintrón
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Adriana M Fresquez
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Manuel F Navedo
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.)
| | - Gregory C Amberg
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins (N.L.C., A.M.F., G.C.A.); and Department of Pharmacology, University of California, Davis (M.N.-C., M.F.N.).
| |
Collapse
|
104
|
Diebold BA, Smith SM, Li Y, Lambeth JD. NOX2 As a Target for Drug Development: Indications, Possible Complications, and Progress. Antioxid Redox Signal 2015; 23:375-405. [PMID: 24512192 PMCID: PMC4545678 DOI: 10.1089/ars.2014.5862] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/08/2014] [Indexed: 12/27/2022]
Abstract
SIGNIFICANCE NOX2 is important for host defense, and yet is implicated in a large number of diseases in which inflammation plays a role in pathogenesis. These include acute and chronic lung inflammatory diseases, stroke, traumatic brain injury, and neurodegenerative diseases, including Alzheimer's and Parkinson's Diseases. RECENT ADVANCES Recent drug development programs have targeted several NOX isoforms that are implicated in a variety of diseases. The focus has been primarily on NOX4 and NOX1 rather than on NOX2, due, in part, to concerns about possible immunosuppressive side effects. Nevertheless, NOX2 clearly contributes to the pathogenesis of many inflammatory diseases, and its inhibition is predicted to provide a novel therapeutic approach. CRITICAL ISSUES Possible side effects that might arise from targeting NOX2 are discussed, including the possibility that such inhibition will contribute to increased infections and/or autoimmune disorders. The state of the field with regard to existing NOX2 inhibitors and targeted development of novel inhibitors is also summarized. FUTURE DIRECTIONS NOX2 inhibitors show particular promise for the treatment of inflammatory diseases, both acute and chronic. Theoretical side effects include pro-inflammatory and autoimmune complications and should be considered in any therapeutic program, but in our opinion, available data do not indicate that they are sufficiently likely to eliminate NOX2 as a drug target, particularly when weighed against the seriousness of many NOX2-related indications. Model studies demonstrating efficacy with minimal side effects are needed to encourage future development of NOX2 inhibitors as therapeutic agents.
Collapse
Affiliation(s)
- Becky A. Diebold
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Susan M.E. Smith
- Department of Biology and Physics, Kennesaw State University, Kennesaw, Georgia
| | - Yang Li
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - J. David Lambeth
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
105
|
Hegedűs P, Korkmaz S, Radovits T, Schmidt H, Li S, Yoshikawa Y, Yasui H, Merkely B, Karck M, Szabó G. Bis (aspirinato) zinc (II) complex successfully inhibits carotid arterial neointima formation after balloon-injury in rats. Cardiovasc Drugs Ther 2015; 28:533-9. [PMID: 25129612 DOI: 10.1007/s10557-014-6549-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Neointima formation following angioplasty is a serious consequence of endothelial damage in arteries. Inflammatory mediators and lack of endothelial regulatory mechanisms lead to migration and proliferation of smooth-muscle cells and thus to restenosis. This study examines the effect of the novel bis (aspirinato) zinc (II) complex on neointima formation in a rat model of carotid balloon-injury. METHODS Rats underwent balloon-injury of the right common carotid artery, then received PEG400 vehicle (untreated-group), acetylsalicylic-acid (ASA-group), zinc-chloride (Zn-group) and bis (aspirinato) zinc (II) complex (Zn(ASA) 2-group) orally for 18 consecutive days. From harvested carotid arteries, histology, immunohistochemistry and mRNA expression analysis were performed. RESULTS Compared to the untreated-group, Zn (ASA) 2-treatment significantly lowered stenosis ratio (54.0 ± 5.8% to 25.5 ± 3.9%) and reduced neointima/media ratio (1.5 ± 0.2 to 0.5 ± 0.1). Significantly higher alpha smooth muscle actin mRNA and protein expression were measured after Zn (ASA)2 and Zn-treatment in comparison with the untreated and ASA-groups while the expression of matrix-metalloproteinase-9 was significantly higher in these groups compared to Zn (ASA)2. The presence of collagen in media was significantly decreased in all treated groups. mRNA expressions of nuclear factor kappa-b, transforming growth-factor-β and proliferating cell nuclear antigen were significantly down-regulated, whereas a20 was up-regulated by Zn (ASA)2 treatment compared to the untreated and ASA-groups. CONCLUSION This study proves the effectivity of the novel bis (aspirinato) zinc complex in reducing neointima formation and restenosis after balloon-injury and supports the hypothesis that inhibition of smooth-muscle transformation/proliferation plays a key role in the prevention of restenosis.
Collapse
Affiliation(s)
- Péter Hegedűs
- Department of Cardiac Surgery, University of Heidelberg, INF 326, Heidelberg, 69120, Germany,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Abstract
Numerous studies have found that NADPH oxidase (NOX) plays a key role in the development of hepatic fibrosis. The activation of NOX in hepatic stellate cells (HSCs) is followed by production of reactive oxygen species and activation of PI3K/Akt and P38MAPK signaling pathways, which in turn causes the excessive expression of type I collagen and its deposition in the liver. Ursolic acid (UA) is a Chinese herbal monomer that widely exists in multiple medicinal plants. Studies have found that UA markedly ameliorates liver fibrosis in experimental animals, and reduces hepatocyte necrosis. In this paper, we review the recent advances in understanding the antifibrotic effect of UA in the liver and the related mechanisms.
Collapse
|
107
|
Choi SH, Park S, Oh CJ, Leem J, Park KG, Lee IK. Dipeptidyl peptidase-4 inhibition by gemigliptin prevents abnormal vascular remodeling via NF-E2-related factor 2 activation. Vascul Pharmacol 2015; 73:11-9. [PMID: 26187356 DOI: 10.1016/j.vph.2015.07.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/10/2015] [Accepted: 07/11/2015] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors exert a potent anti-hyperglycemic effect and reduce cardiovascular risk in type 2 diabetic patients. Several studies have shown that DPP-4 inhibitors including sitagliptin have beneficial effects in atherosclerosis and cardiac infarction involving reactive oxygen species. Here, we show that gemigliptin can directly attenuate the abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) via enhanced NF-E2-related factor 2 (Nrf2) activity. Gemigliptin dramatically prevented ligation injury-induced neointimal hyperplasia in mouse carotid arteries. Likewise, the proliferation of primary VSMCs was significantly attenuated by gemigliptin in a dose-dependent manner consistent with a decrease in phospho-Rb, resulting in G1 cell cycle arrest. We found that gemigliptin enhanced Nrf2 activity not only by mRNA expression, but also by increasing Keap1 proteosomal degradation by p62, leading to the induction of Nrf2 target genes such as HO-1 and NQO1. The anti-proliferative role of gemigliptin disappeared with DPP-4 siRNA knockdown, indicating that the endogenous DPP-4 in VSMCs contributed to the effect of gemigliptin. In addition, gemigliptin diminished TNF-α-mediated cell adhesion molecules such as MCP-1 and VCAM-1 and reduced MMP2 activity in VSMCs. Taken together, our data indicate that gemigliptin exerts a preventative effect on the proliferation and migration of VSMCs via Nrf2.
Collapse
Affiliation(s)
- Seung Hee Choi
- Department of Biomedical Science, Graduate School, Kyungpook National University, Daegu, Republic of Korea
| | - Sungmi Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University, Daegu, Republic of Korea
| | - Chang Joo Oh
- Research Institutes of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Jaechan Leem
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University, Daegu, Republic of Korea
| | - Keun-Gyu Park
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Leading-Edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University, Daegu, Republic of Korea; Research Institutes of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea; Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
108
|
Yang S, Gao Y, Liu G, Li J, Shi K, Du B, Si D, Yang P. The human ATF1 rs11169571 polymorphism increases essential hypertension risk through modifying miRNA binding. FEBS Lett 2015; 589:2087-93. [PMID: 26149214 DOI: 10.1016/j.febslet.2015.06.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/21/2015] [Accepted: 06/22/2015] [Indexed: 11/19/2022]
Abstract
Activating transcription factor 1 (ATF1) may be involved in essential hypertension (EH) by induction of NADPH oxidase 1 (NOX1) and radical oxygen species (ROSs) production. Abnormal expression of ATF1 was found in EH in previous microarray analysis. Here we tested whether a single nucleotide polymorphism (SNP) located in the 3'-untranslated region (3'UTR) of ATF1 was associated with EH susceptibility by affecting microRNA (miRNA) binding. In silico analysis indicated that rs11169571 (T>C) was a candidate SNP to modulate miRNA: ATF1 mRNA complex, with the greatest changed energy for hsa-miR-1283, and the luciferase reporter analysis showed that miR-1283 inhibited the activity of the reporter vector carrying -T allele, but not the -C allele. In addition, inhibition of miR-1283 in HA-VSMCs enhanced the expression of ATF1 mRNA as well as the ROS levels. Further case-control study showed that rs11169571 was significantly associated with increased risk of EH. Finally, we observed an increased ATF1 protein level in peripheral blood of EH patients with CC carriers compared to TT carriers of rs11169571, with an intermediate ATF1 level in TC carriers. These results suggested that rs11169571 of ATF1 gene may be associated with EH, and the SNP-modified posttranscriptional gene regulation by miRNAs could be a potentially pathogenetic mechanism of EH.
Collapse
Affiliation(s)
- Sibao Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yongjian Gao
- Department of Gastrointestinal Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Guohui Liu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Jiayu Li
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Kaiyao Shi
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Beibei Du
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Daoyuan Si
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, Changchun, China.
| |
Collapse
|
109
|
Dan J, Sun X, Li W, Zhang Y, Li X, Xu H, Li Z, Tian Z, Guo S, Yao J, Gao W, Tian Y. 5-Aminolevulinic Acid-Mediated Sonodynamic Therapy Promotes Phenotypic Switching from Dedifferentiated to Differentiated Phenotype via Reactive Oxygen Species and p38 Mitogen-Activated Protein Kinase in Vascular Smooth Muscle Cells. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:1681-1689. [PMID: 25796412 DOI: 10.1016/j.ultrasmedbio.2014.12.664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 12/11/2014] [Accepted: 12/22/2014] [Indexed: 06/04/2023]
Abstract
Sonodynamic therapy (SDT) has been found to inhibit in-stent restenosis in animal models. However, the mechanism is not fully elucidated. Here, we investigated the effects of 5-aminolevulinic acid (ALA)-mediated SDT (ALA-SDT) on vascular smooth muscle cells (VSMCs), a cause of restenosis, with a focus on SDT-induced phenotypic switching. Serum-induced dedifferentiated VSMCs were cultured with ALA (1 mm, 24 h) and exposed to ultrasound (0.8 W/cm(2)) for 5 min. Results indicated that ALA-SDT inhibited the migration and proliferation of VSMCs and enhanced the expression of differentiated phenotypic markers in VSMCs. Additionally, ALA-SDT increased intracellular reactive oxygen species accumulation and phosphorylated p38 mitogen-activated protein kinase in VSMCs. Inhibition of reactive oxygen species elevation or p38 mitogen-activated protein kinase activity abolished the expression of smooth muscle 22α (SM22α) in VSMCs induced by ALA-SDT. Taken together, these results suggest that ALA-SDT promotes transformation of the VSMC phenotype from the dedifferentiated to differentiated status via reactive oxygen species and activated p38 mitogen-activated protein kinase.
Collapse
Affiliation(s)
- Juhua Dan
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China
| | - Xin Sun
- Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wanlu Li
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China
| | - Yun Zhang
- Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xuesong Li
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China
| | - Haobo Xu
- Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhitao Li
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China
| | - Zhen Tian
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China
| | - Shuyuan Guo
- Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jianting Yao
- Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Weidong Gao
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China; Department of Anesthesiology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ye Tian
- Department of Pathophysiology, State Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Harbin Medical University, Harbin, China; Division of Cardiology, First Affiliated Hospital, Harbin Medical University, Harbin, China.
| |
Collapse
|
110
|
Wu XL, Zheng B, Jin LS, Zhang RN, He M, Yang Z, Wen JK. Chinese medicine Tongxinluo reduces atherosclerotic lesion by attenuating oxidative stress and inflammation in microvascular endothelial cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:6323-6333. [PMID: 26261508 PMCID: PMC4525842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/26/2015] [Indexed: 06/04/2023]
Abstract
Oxidative stress and inflammation are the important pathological basis of atherogenesis. So, attenuating oxidative stress and inflammation has a very important significance in the prevention and treatment of atherosclerosis. The aim of present study was to investigate whether anti-atherosclerotic effect of Tongxinluo (TXL), a compound traditional Chinese medicine, is related to its anti-oxidation and anti-inflammation in human cardiac microvascular endothelial cells (HCMEC). We found that TXL treatment significantly reduced serum lipid levels and atherosclerotic plaque formation of apoE-deficient mice, and improved endothelial cell function as evidenced by increased expression of CD31 and eNOS. TXL pretreatment could abrogate the up-regulation of ROS and MDA induced by C16. Further experiments showed that the anti-oxidative effect of TXL may be related to inhibiting the expression of p22(phox), p47(phox) and HO-1 in HCMECs. We also found that TXL could inhibit the release of IL-1β and TNFα induced by C16, which is mediated by inhibiting the expression and activation of NF-κB. In conclusion, TXL decreases atherosclerotic plaque formation and improves endothelial cell function by inhibiting oxidative stress and inflammation in HCMECs. This finding provides a new molecular mechanism for the anti-atherosclerotic effect of TXL.
Collapse
Affiliation(s)
- Xiao-Li Wu
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
- The Second Hospital of Hebei Medical UniversityShijiazhuang 050000, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| | - Li-Shuang Jin
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| | - Ruo-Nan Zhang
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| | - Ming He
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| | - Zhan Yang
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Hebei Medical UniversityZhongshan East Road, Shijiazhuang 050017, China
| |
Collapse
|
111
|
Chinese Herbal Compounds for the Prevention and Treatment of Atherosclerosis: Experimental Evidence and Mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:752610. [PMID: 26089946 PMCID: PMC4451781 DOI: 10.1155/2015/752610] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/15/2014] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is a leading cause of disability and death worldwide. Research into the disease has led to many compelling hypotheses regarding the pathophysiology of atherosclerotic lesion formation and the resulting complications such as myocardial infarction and stroke. Herbal medicine has been widely used in China as well as other Asian countries for the treatment of cardiovascular diseases for hundreds of years; however, the mechanisms of action of Chinese herbal medicine in the prevention and treatment of atherosclerosis have not been well studied. In this review, we briefly describe the mechanisms of atherogenesis and then summarize the research that has been performed in recent years regarding the effectiveness and mechanisms of antiatherogenic Chinese herbal compounds in an attempt to build a bridge between traditional Chinese medicine and cellular and molecular cardiovascular medicine.
Collapse
|
112
|
Abstract
Despite the wealth of pre-clinical support for a role for reactive oxygen and nitrogen species (ROS/RNS) in the aetiology of diabetic complications, enthusiasm for antioxidant therapeutic approaches has been dampened by less favourable outcomes in large clinical trials. This has necessitated a re-evaluation of pre-clinical evidence and a more rational approach to antioxidant therapy. The present review considers current evidence, from both pre-clinical and clinical studies, to address the benefits of antioxidant therapy. The main focus of the present review is on the effects of direct targeting of ROS-producing enzymes, the bolstering of antioxidant defences and mechanisms to improve nitric oxide availability. Current evidence suggests that a more nuanced approach to antioxidant therapy is more likely to yield positive reductions in end-organ injury, with considerations required for the types of ROS/RNS involved, the timing and dosage of antioxidant therapy, and the selective targeting of cell populations. This is likely to influence future strategies to lessen the burden of diabetic complications such as diabetes-associated atherosclerosis, diabetic nephropathy and diabetic retinopathy.
Collapse
|
113
|
Lee KP, Sudjarwo GW, Jung SH, Lee D, Lee DY, Lee GB, Baek S, Kim DY, Lee HM, Kim B, Kwon SC, Won KJ. Carvacrol inhibits atherosclerotic neointima formation by downregulating reactive oxygen species production in vascular smooth muscle cells. Atherosclerosis 2015; 240:367-73. [PMID: 25875388 DOI: 10.1016/j.atherosclerosis.2015.03.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 03/01/2015] [Accepted: 03/21/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Carvacrol (2-methyl-5-(1-methylethyl) phenol), a cyclic monoterpene, exerts protective activities in a variety of pathological states including tumor growth, inflammation, and oxidative stress. However, it is unknown whether carvacrol affects events in vascular cells during the development of atherosclerotic neointima. We investigated the effects of carvacrol on the migration and proliferation of rat aortic smooth muscle cells (RASMCs) and on vascular neointima formation. METHODS AND RESULTS Carvacrol significantly inhibited platelet-derived growth factor (PDGF)-BB-stimulated RASMC migration and proliferation in a concentration-dependent manner. Cell viability was not affected by treatment with carvacrol. Carvacrol attenuated the expression of NADPH oxidase (NOX) 1 and the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase 1/2 in response to PDGF-BB. Moreover, carvacrol suppressed the PDGF-BB-stimulated generation of H2O2 and inhibited the activity of NOX in RASMCs. Treatment with carvacrol inhibited PDGF-BB-induced aortic sprout outgrowth, balloon injury-evoked vascular neointima formation, and expression of proliferating cell nuclear antigen in the neointima. CONCLUSION These findings indicate that carvacrol inhibits migration and proliferation of RASMCs by suppressing the reactive oxygen species-mediated MAPK signaling pathway in these cells, thereby attenuating vascular neointimal formation. Carvacrol may be a promising agent for preventing vascular restenosis or atherosclerosis.
Collapse
Affiliation(s)
- Kang Pa Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Giftania W Sudjarwo
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Seung Hyo Jung
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Donghyen Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Dong-Youb Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Gyoung Beom Lee
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Suji Baek
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Do-Yoon Kim
- Department of Cosmetic Science, College of Natural Science, Hoseo University, Asan 336-795, South Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Natural Science, Hoseo University, Asan 336-795, South Korea
| | - Bokyung Kim
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea
| | - Seong-Chun Kwon
- Department of Physiology, Catholic Kwandong University College of Medicine, Kangneung 201-701, South Korea.
| | - Kyung Jong Won
- Department of Physiology, School of Medicine, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, South Korea.
| |
Collapse
|
114
|
Vascular Smooth Muscle Cells. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
115
|
The role of oxidative stress and autophagy in atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:130315. [PMID: 25866599 PMCID: PMC4381688 DOI: 10.1155/2015/130315] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 03/01/2015] [Accepted: 03/02/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a multifactorial, multistep disorder of large- and medium-sized arteries involving, in addition to age, gender and menopausal status, a complex interplay between lifestyle and genetic risk factors. Atherosclerosis usually begins with the diffusion and retention of atherogenic lipoproteins into the subendothelial space of the artery wall where they become oxidized by local enzymes and accumulate, leading to the formation of a cushion called atheroma or atheromatous or fibrofatty plaque, composed of a mixture of macrophages, lymphocytes, smooth muscle cells (SMCs), cholesterol cleft, necrotic debris, and lipid-laden foam cells. The pathogenesis of atherosclerosis still remains incompletely understood but emerging evidence suggests that it may involve multiple cellular events, including endothelial cell (EC) dysfunction, inflammation, proliferation of vascular SMCs, matrix (ECM) alteration, and neovascularization. Actually, a growing body of evidence indicates that autophagy along with the chronic and acute overproduction of reactive oxygen species (ROS) is integral to the development and progression of the disease and may represent fruitful avenues for biological investigation and for the identification of new therapeutic targets. In this review, we give an overview of ROS and autophagy in atherosclerosis as background to understand their potential role in this vascular disease.
Collapse
|
116
|
Wang L, Yu T, Lee H, O'Brien DK, Sesaki H, Yoon Y. Decreasing mitochondrial fission diminishes vascular smooth muscle cell migration and ameliorates intimal hyperplasia. Cardiovasc Res 2015; 106:272-83. [PMID: 25587046 DOI: 10.1093/cvr/cvv005] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 12/31/2014] [Indexed: 12/13/2022] Open
Abstract
AIMS Vascular smooth muscle cell (VSMC) migration in response to arterial wall injury is a critical process in the development of intimal hyperplasia. Cell migration is an energy-demanding process that is predicted to require mitochondrial function. Mitochondria are morphologically dynamic, undergoing continuous shape change through fission and fusion. However, the role of mitochondrial morphology in VSMC migration is not well understood. The aim of the study is to understand how mitochondrial fission contributes to VSMC migration and provides its in vivo relevance in the mouse model of intimal hyperplasia. METHODS AND RESULTS In primary mouse VSMCs, the chemoattractant PDGF induced mitochondrial shortening through the mitochondrial fission protein dynamin-like protein 1 (DLP1)/Drp1. Perturbation of mitochondrial fission by expressing the dominant-negative mutant DLP1-K38A or by DLP1 silencing greatly decreased PDGF-induced lamellipodia formation and VSMC migration, indicating that mitochondrial fission is an important process in VSMC migration. PDGF induced an augmentation of mitochondrial energetics as well as ROS production, both of which were found to be necessary for VSMC migration. Mechanistically, the inhibition of mitochondrial fission induced an increase of mitochondrial inner membrane proton leak in VSMCs, abrogating the PDGF-induced energetic enhancement and an ROS increase. In an in vivo model of intimal hyperplasia, transgenic mice expressing DLP1-K38A displayed markedly reduced ROS levels and neointima formation in response to femoral artery wire injury. CONCLUSIONS Mitochondrial fission is an integral process in cell migration, and controlling mitochondrial fission can limit VSMC migration and the pathological intimal hyperplasia by altering mitochondrial energetics and ROS levels.
Collapse
Affiliation(s)
- Li Wang
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Tianzheng Yu
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Dawn K O'Brien
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Georgia Regents University, 1120 Fifteenth Street, Augusta, GA 30912-3000, USA
| |
Collapse
|
117
|
Enhanced vascular PI3K/Akt-NOX signaling underlies the peripheral NMDAR-mediated pressor response in conscious rats. J Cardiovasc Pharmacol 2014; 63:395-405. [PMID: 24336015 DOI: 10.1097/fjc.0000000000000059] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The molecular mechanisms for peripheral N-methyl-D-aspartate receptor (NMDAR)-mediated vascular oxidative stress and pressor response are not known. We conducted integrative (in vivo) and ex vivo biochemical studies to test the hypothesis that reactive oxygen species (ROS)-dependent calcium influx, triggered by the activation of vascular kinases, underlies the NMDAR-mediated pressor response. Pharmacological inhibition of phosphoinositide 3-kinase (PI3K)/Akt (wortmannin, 15 μg/kg), protein kinase C (chelerythrine: 5 mg/kg, intravenous), Ca²⁺ influx (nifedipine, 0.35 or 0.75 mg/kg), or NADPH oxidase (NOX: apocynin, 5 mg/kg) attenuated the peripheral NMDAR-mediated pressor response in conscious male Sprague-Dawley rats. NMDAR activation enhanced the phosphorylation of Akt, ERK1, JNK and p38 (Western blot), and NOX activity in vascular tissues collected during the pressor response caused by NMDA infusion (180 μg·kg⁻¹·min⁻¹, 30 minutes). Furthermore, ex vivo studies showed that wortmannin, chelerythrine, or apocynin abrogated the NMDAR-mediated vascular nitric oxide (NO) and ROS generation and NOX activation in the vasculature. These findings implicate vascular PI3K/Akt-protein kinase C signaling in the peripheral NMDAR-mediated increases in vascular NO and NOX activation (ROS), which ultimately lead to calcium influx and pressor response in conscious rats.
Collapse
|
118
|
Liu CF, Zhang J, Shen K, Gao PJ, Wang HY, Jin X, Meng C, Fang NY. Adventitial gene transfer of catalase attenuates angiotensin II-induced vascular remodeling. Mol Med Rep 2014; 11:2608-14. [PMID: 25503998 PMCID: PMC4337488 DOI: 10.3892/mmr.2014.3069] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023] Open
Abstract
Vascular adventitia and adventitia-derived reactive oxygen species (ROS) contribute to vascular remodeling following vascular injury. A previous ex vivo study in adventitial fibroblasts showed that catalase, one of most important anti-oxide enzymes, was downregulated by angiotensin II (AngII). The aim of the present study was to investigate whether adventitial gene transfer of catalase affects AngII-induced vascular remodeling in vivo. Adenoviruses co-expressing catalase and enhanced green fluorescent protein (eGFP) or expressing eGFP only were applied to the adventitial surface of common carotid arteries of Sprague-Dawley rats. Alzet minipumps administering AngII (0.75 mg/kg/day) were then implanted subcutaneously for 14 days. Systolic blood pressure and biological parameters of vascular remodeling were measured in each group. Adventitial fibroblasts were cultured and p38 mitogen-activated protein kinase (MAPK) phosphorylation was measured using western blot analysis. The results showed that adventitial gene transfer of catalase had no effect on AngII-induced systolic blood pressure elevation. However, catalase adenovirus transfection significantly inhibited AngII-induced media hypertrophy compared with that of the control virus (P<0.05). In addition, catalase transfection significantly attenuated AngII-induced ROS generation, macrophage infiltration, collagen deposition and adventitial α-smooth muscle actin expression. Furthermore, catalase transfection significantly inhibited the AngII-induced increase in p38MAPK phosphorylation. In conclusion, the results of the present study demonstrated that adventitial gene transfer of catalase significantly attenuated AngII-induced vascular remodeling in rats via inhibition of adventitial p38MAPK phosphorylation.
Collapse
Affiliation(s)
- Cun-Fei Liu
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Jia Zhang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Kai Shen
- Division of Cardiology, Zhoushan People's Hospital, Zhoushan, Zhejiang 316000, P.R. China
| | - Ping-Jin Gao
- Shanghai Key Laboratory of Vascular Biology at Ruijin Hospital and Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Hai-Ya Wang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Xin Jin
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Chao Meng
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| | - Ning-Yuan Fang
- Department of Geriatrics, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, P.R. China
| |
Collapse
|
119
|
Abdesselem M, Schoeffel M, Maurin I, Ramodiharilafy R, Autret G, Clément O, Tharaux PL, Boilot JP, Gacoin T, Bouzigues C, Alexandrou A. Multifunctional rare-Earth vanadate nanoparticles: luminescent labels, oxidant sensors, and MRI contrast agents. ACS NANO 2014; 8:11126-11137. [PMID: 25290552 DOI: 10.1021/nn504170x] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Collecting information on multiple pathophysiological parameters is essential for understanding complex pathologies, especially given the large interindividual variability. We report here multifunctional nanoparticles which are luminescent probes, oxidant sensors, and contrast agents in magnetic resonance imaging (MRI). Eu(3+) ions in an yttrium vanadate matrix have been demonstrated to emit strong, nonblinking, and stable luminescence. Time- and space-resolved optical oxidant detection is feasible after reversible photoreduction of Eu(3+) to Eu(2+) and reoxidation by oxidants, such as H2O2, leading to a modulation of the luminescence emission. The incorporation of paramagnetic Gd(3+) confers in addition proton relaxation enhancing properties to the system. We synthesized and characterized nanoparticles of either 5 or 30 nm diameter with compositions of GdVO4 and Gd0.6Eu0.4VO4. These particles retain the luminescence and oxidant detection properties of YVO4:Eu. Moreover, the proton relaxivity of GdVO4 and Gd0.6Eu0.4VO4 nanoparticles of 5 nm diameter is higher than that of the commercial Gd(3+) chelate compound Dotarem at 20 MHz. Nuclear magnetic resonance dispersion spectroscopy showed a relaxivity increase above 10 MHz. Complexometric titration indicated that rare-earth leaching is negligible. The 5 nm nanoparticles injected in mice were observed with MRI to concentrate in the liver and the bladder after 30 min. Thus, these multifunctional rare-earth vanadate nanoparticles pave the way for simultaneous optical and magnetic resonance detection, in particular, for in vivo localization evolution and reactive oxygen species detection in a broad range of physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Mouna Abdesselem
- Laboratoire d'Optique et Biosciences, Ecole Polytechnique , CNRS UMR 7645-INSERM U696, 91128 Palaiseau Cedex, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Sakai T, Sato B, Hara K, Hara Y, Naritomi Y, Koyanagi S, Hara H, Nagao T, Ishibashi T. Consumption of water containing over 3.5 mg of dissolved hydrogen could improve vascular endothelial function. Vasc Health Risk Manag 2014; 10:591-7. [PMID: 25378931 PMCID: PMC4207582 DOI: 10.2147/vhrm.s68844] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The redox imbalance between nitric oxide and superoxide generated in the endothelium is thought to play a pivotal role in the development of endothelial dysfunction. A third reactive oxygen species (ROS), H2O2, is known to have both beneficial and detrimental effects on the vasculature. Nonetheless, the influence of the hydroxyl radical, a byproduct of H2O2 decay, is unclear, and there is no direct evidence that the hydroxyl radical impairs endothelial function in conduit arteries. Molecular hydrogen (H2) neutralizes detrimental ROS, especially the hydroxyl radical. OBJECTIVES To assess the influence of the hydroxyl radical on the endothelium and to confirm that a gaseous antioxidant, H2, can be a useful modulator of blood vessel function. METHODS The efficacy of water containing a high concentration of H2 was tested by measuring flow-mediated dilation (FMD) of the brachial artery (BA). The subjects were randomly divided into two groups: the high-H2 group, who drank high-H2 water containing 7 ppm H2 (3.5 mg H2 in 500 mL water); and the placebo group. Endothelial function was evaluated by measuring the FMD of the BA. After measurement of diameter of the BA and FMD at baseline, volunteers drank the high-H2 water or placebo water immediately and with a 30-minute interval; FMD was compared to baseline. RESULTS FMD increased in the high-H2 group (eight males; eight females) from 6.80%±1.96% to 7.64%±1.68% (mean ± standard deviation) and decreased from 8.07%±2.41% to 6.87%±2.94% in the placebo group (ten males; eight females). The ratio to the baseline in the changes of FMD showed significant improvement (P<0.05) in the high-H2 group compared to the placebo group. CONCLUSION H2 may protect the vasculature from shear stress-derived detrimental ROS, such as the hydroxyl radical, by maintaining the nitric oxide-mediated vasomotor response.
Collapse
Affiliation(s)
- Takaaki Sakai
- Department of Cardiology, Haradoi Hospital, Fukuoka, Japan
| | - Bunpei Sato
- MiZ Company Limited, Fujisawa, Kanagawa, Japan
| | - Koji Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Yuichi Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Yuji Naritomi
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | - Samon Koyanagi
- Department of Cardiology, Haradoi Hospital, Fukuoka, Japan
| | - Hiroshi Hara
- Department of Internal Medicine, Haradoi Hospital, Fukuoka, Japan
| | | | - Toru Ishibashi
- Department of Rheumatology and Orthopedic Surgery, Haradoi Hospital, Fukuoka, Japan
| |
Collapse
|
121
|
Kwak SY, Lee HJ, Yang JK, Lee EJ, Seo M, Lee YS. Antioxidant activity of caffeoyl-prolyl-histidine amide and its effects on PDGF-induced proliferation of vascular smooth muscle cells. Amino Acids 2014; 46:2777-85. [PMID: 25218135 DOI: 10.1007/s00726-014-1834-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/26/2014] [Indexed: 11/30/2022]
Abstract
Caffeic acid (CA) is one of the antioxidants found in plants, which protects vascular cells against vascular injuries from oxidative stress. In our previous study, caffeoyl-prolyl-histidine amide (CA-L-Pro-L-His-NH2; CA-PH; a CA derivative) was synthesized, which exhibited a strong antioxidant activity with sufficient stability. In this study, we investigated the role of CA-PH in vascular smooth muscle cells (VSMCs) and confirmed the enhanced antioxidant activity of CA-PH compared with that of CA. In in vitro tube assays, CA-PH showed a higher free-radical-scavenging activity and lipid-peroxidation-inhibition activity than those of CA. In VSMCs, CA-PH significantly reduced hydrogen peroxide-induced ROS generation and increased the expression of heme oxygenase-1. Moreover, CA-PH effectively inhibited the platelet-derived growth factor-induced cellular proliferation of VSMCs, which was confirmed by a decrease in the expression of the proliferating cell nuclear antigen and the phosphorylation of Akt.
Collapse
Affiliation(s)
- Seon-Yeong Kwak
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 151-744, Republic of Korea
| | | | | | | | | | | |
Collapse
|
122
|
Ali Khan H, Mutus B. Protein disulfide isomerase a multifunctional protein with multiple physiological roles. Front Chem 2014; 2:70. [PMID: 25207270 PMCID: PMC4144422 DOI: 10.3389/fchem.2014.00070] [Citation(s) in RCA: 133] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 08/07/2014] [Indexed: 11/19/2022] Open
Abstract
Protein disulfide isomerase (PDI), is a member of the thioredoxin superfamily of redox proteins. PDI has three catalytic activities including, thiol-disulfide oxireductase, disulfide isomerase and redox-dependent chaperone. Originally, PDI was identified in the lumen of the endoplasmic reticulum and subsequently detected at additional locations, such as cell surfaces and the cytosol. This review will provide an overview of the recent advances in relating the structural features of PDI to its multiple catalytic roles as well as its physiological and pathophysiological functions related to redox regulation and protein folding.
Collapse
Affiliation(s)
- Hyder Ali Khan
- Chemistry and Biochemistry Department, University of Windsor Windsor, ON, Canada
| | - Bulent Mutus
- Chemistry and Biochemistry Department, University of Windsor Windsor, ON, Canada
| |
Collapse
|
123
|
Gole HKA, Tharp DL, Bowles DK. Upregulation of intermediate-conductance Ca2+-activated K+ channels (KCNN4) in porcine coronary smooth muscle requires NADPH oxidase 5 (NOX5). PLoS One 2014; 9:e105337. [PMID: 25144362 PMCID: PMC4140784 DOI: 10.1371/journal.pone.0105337] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 07/23/2014] [Indexed: 02/07/2023] Open
Abstract
Aims NADPH oxidase (NOX) is the primary source of reactive oxygen species (ROS) in vascular smooth muscle cells (SMC) and is proposed to play a key role in redox signaling involved in the pathogenesis of cardiovascular disease. Growth factors and cytokines stimulate coronary SMC (CSMC) phenotypic modulation, proliferation, and migration during atherosclerotic plaque development and restenosis. We previously demonstrated that increased expression and activity of intermediate-conductance Ca2+-activated K+ channels (KCNN4) is necessary for CSMC phenotypic modulation and progression of stenotic lesions. Therefore, the purpose of this study was to determine whether NOX is required for KCNN4 upregulation induced by mitogenic growth factors. Methods and Results Dihydroethidium micro-fluorography in porcine CSMCs demonstrated that basic fibroblast growth factor (bFGF) increased superoxide production, which was blocked by the NOX inhibitor apocynin (Apo). Apo also blocked bFGF-induced increases in KCNN4 mRNA levels in both right coronary artery sections and CSMCs. Similarly, immunohistochemistry and whole cell voltage clamp showed bFGF-induced increases in CSMC KCNN4 protein expression and channel activity were abolished by Apo. Treatment with Apo also inhibited bFGF-induced increases in activator protein-1 promoter activity, as measured by luciferase activity assay. qRT-PCR demonstrated porcine coronary smooth muscle expression of NOX1, NOX2, NOX4, and NOX5 isoforms. Knockdown of NOX5 alone prevented both bFGF-induced upregulation of KCNN4 mRNA and CSMC migration. Conclusions Our findings provide novel evidence that NOX5-derived ROS increase functional expression of KCNN4 through activator protein-1, providing another potential link between NOX, CSMC phenotypic modulation, and atherosclerosis.
Collapse
Affiliation(s)
- Hope K. A. Gole
- Department of Biomedical Sciences, University of Missouri Columbia, Columbia, Missouri, United States of America
| | - Darla L. Tharp
- Department of Biomedical Sciences, University of Missouri Columbia, Columbia, Missouri, United States of America
| | - Douglas K. Bowles
- Department of Biomedical Sciences, University of Missouri Columbia, Columbia, Missouri, United States of America
- Dalton Cardiovascular Research Center, University of Missouri Columbia, Columbia, Missouri, United States of America
- Medical Pharmacology and Physiology, University of Missouri Columbia, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
124
|
Angiotensin II induces Fat1 expression/activation and vascular smooth muscle cell migration via Nox1-dependent reactive oxygen species generation. J Mol Cell Cardiol 2014; 66:18-26. [PMID: 24445059 DOI: 10.1016/j.yjmcc.2013.10.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 01/06/2023]
Abstract
Fat1 is an atypical cadherin that controls vascular smooth muscle cell (VSMC) proliferation and migration. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 1 (Nox1) is an important source of reactive oxygen species (ROS) in VSMCs. Angiotensin II (Ang II) induces the expression and/or activation of both Fat1 and Nox1 proteins. This study tested the hypothesis that Ang II-induced Fat1 activation and VSMC migration are mediated by Nox1-dependent ROS generation and redox signaling. Studies were performed in cultured VSMCs from Sprague–Dawley rats. Cells were treated with Ang II (1 μmol/L) for short (5 to 30 min) or long term stimulations (3 to 12 h) in the absence or presence of the antioxidant apocynin (10 μmol/L), extracellular-signal-regulated kinases 1/2 (Erk1/2) inhibitor PD98059 (1 μmol/L), or Ang II type 1 receptor (AT1R) valsartan (1 μmol/L). siRNA was used to knockdown Nox1 or Fat1. Cell migration was determined by Boyden chamber assay. Ang II increased Fat1 mRNA and protein levels and promoted Fat1 translocation to the cell membrane, responses that were inhibited by AT1R antagonist and antioxidant treatment. Downregulation of Nox1 inhibited the effects of Ang II on Fat1 protein expression. Nox1 protein induction, ROS generation, and p44/p42 MAPK phosphorylation in response to Ang II were prevented by valsartan and apocynin, and Nox1 siRNA inhibited Ang II-induced ROS generation. Knockdown of Fat1 did not affect Ang II-mediated increases in Nox1 expression or ROS. Inhibition of p44/p42 MAPK phosphorylation by PD98059 abrogated the Ang II-induced increase in Fat1 expression and membrane translocation. Knockdown of Fat1 inhibited Ang II-induced VSMC migration, which was also prevented by valsartan, apocynin, PD98059, and Nox1 siRNA. Our findings indicate that Ang II regulates Fat1 expression and activity and induces Fat1-dependent VSMC migration via activation of AT1R, ERK1/2, and Nox1-derived ROS, suggesting a role for Fat1 downstream of Ang II signaling that leads to vascular remodeling.
Collapse
|
125
|
Siani A, Tirelli N. Myofibroblast differentiation: main features, biomedical relevance, and the role of reactive oxygen species. Antioxid Redox Signal 2014; 21:768-85. [PMID: 24279926 DOI: 10.1089/ars.2013.5724] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Myofibroblasts are prototypical fibrotic cells, which are involved in a number of more or less pathological conditions, from foreign body reactions to scarring, from liver, kidney, or lung fibrosis to neoplastic phenomena. The differentiation of precursor cells (not only of fibroblastic nature) is characterized by a complex interplay between soluble factors (growth factors such as transforming growth factor β1, reactive oxygen species [ROS]) and material properties (matrix stiffness). RECENT ADVANCES The last 15 years have seen very significant advances in the identification of appropriate differentiation markers, in the understanding of the differentiation mechanism, and above all, the involvement of ROS as causative and persistence factors. CRITICAL ISSUES The specific mechanisms of action of ROS remain largely unknown, although evidence suggests that both intracellular and extracellular phenomena play a role. FUTURE DIRECTIONS Approaches based on antioxidant (ROS-scavenging) principles and on the potentiation of nitric oxide signaling hold much promise in view of a pharmacological therapy of fibrotic phenomena. However, how to make the active principles available at the target sites is yet a largely neglected issue.
Collapse
Affiliation(s)
- Alessandro Siani
- 1 School of Pharmacy and Pharmaceutical Sciences, University of Manchester , Manchester, United Kingdom
| | | |
Collapse
|
126
|
Hsu JH, Liou SF, Yang SN, Wu BN, Dai ZK, Chen IJ, Yeh JL, Wu JR. B-type natriuretic peptide inhibits angiotensin II-induced proliferation and migration of pulmonary arterial smooth muscle cells. Pediatr Pulmonol 2014; 49:734-44. [PMID: 24167111 DOI: 10.1002/ppul.22904] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 06/21/2013] [Accepted: 08/13/2013] [Indexed: 01/10/2023]
Abstract
Pulmonary vascular remodeling, characterized by disordered proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), is a pathognomonic feature of pulmonary arterial hypertension. Thus, pharmacologic strategy targeting on anti-proliferation and anti-migration of PASMCs may have therapeutic implications for PAH. Here we investigated the effects and underlying mechanisms of B-type natriuretic peptide (BNP) on angiotensin II (Ang II)-induced proliferation and migration of PASMCs. Proliferation and migration of PASMCs cultured from Wistar rats were induced by Ang II, with or without BNP treatment. In addition, potential underlying mechanisms including cell cycle progression, Ca(2+) overload, reactive oxygen species (ROS) production, signal transduction of MAPK and Akt, and the cGMP/PKG pathway were examined. We found that BNP inhibited Ang II-induced PASMCs proliferation and migration dose dependently. BNP could also arrest the cell cycle progression in the G0/G1-phase. In addition, BNP attenuated intracellular calcium overload caused by Ang II. Moreover, Ang II-induced ROS production was mitigated by BNP, with associated down-regulation of NAD(P)H oxidase 1 (Nox1) and reduced mitochondrial ROS production. Finally, Ang II-activated MAPKs and Akt were also counteracted by BNP. Of note, all these effects of BNP were abolished by a PKG inhibitor (Rp-8-Br-PET-cGMPS). In conclusion, BNP inhibits Ang II-induced PASMCs proliferation and migration. These effects are potentially mediated by decreased calcium influx, reduced ROS production by Nox1 and mitochondria, and down-regulation of MAPK and Akt signal transduction, through the cGMP/PKG pathway. Therefore, this study implicates that BNP may have a therapeutic role in pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Jong-Hau Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; Department of Pediatrics, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Song Z, Zhu X, Jin R, Wang C, Yan J, Zheng Q, Nanda A, Granger DN, Li G. Roles of the kinase TAK1 in CD40-mediated effects on vascular oxidative stress and neointima formation after vascular injury. PLoS One 2014; 9:e101671. [PMID: 25050617 PMCID: PMC4106789 DOI: 10.1371/journal.pone.0101671] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 05/29/2014] [Indexed: 12/18/2022] Open
Abstract
Although TAK1 has been implicated in inflammation and oxidative stress, its roles in vascular smooth muscle cells (VSMCs) and in response to vascular injury have not been investigated. The present study aimed to investigate the role of TAK1 in modulating oxidative stress in VSMCs and its involvement in neointima formation after vascular injury. Double immunostaining reveals that vascular injury induces a robust phosphorylation of TAK1 (Thr187) in the medial VSMCs of injured arteries in wildtype mice, but this effect is blocked in CD40-deficient mice. Upregulation of TAK1 in VSMCs is functionally important, as it is critically involved in pro-oxidative and pro-inflammatory effects on VSMCs and eventual neointima formation. In vivo, pharmacological inhibition of TAK1 with 5Z-7-oxozeaenol blocked the injury-induced phosphorylation of both TAK1 (Thr187) and NF-kB/p65 (Ser536), associated with marked inhibition of superoxide production, 3-nitrotyrosine, and MCP-1 in the injured arteries. Cell culture experiments demonstrated that either siRNA knockdown or 5Z-7-oxozeaenol inhibition of TAK1 significantly attenuated NADPH oxidase activation and superoxide production induced by CD40L/CD40 stimulation. Co-immunoprecipitation experiments indicate that blockade of TAK1 disrupted the CD40L-induced complex formation of p22phox with p47phox, p67phox, or Nox4. Blockade of TAK1 also inhibited CD40L-induced NF-kB activation by modulating IKKα/β and NF-kB p65 phosphorylation and this was related to reduced expression of proinflammatory genes (IL-6, MCP-1 and ICAM-1) in VSMCs. Lastly, treatment with 5Z-7-oxozeaenol attenuated neointimal formation in wire-injured femoral arteries. Our findings demonstrate previously uncharacterized roles of TAK1 in vascular oxidative stress and the contribution to neointima formation after vascular injury.
Collapse
Affiliation(s)
- Zifang Song
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaolei Zhu
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
| | - Rong Jin
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
| | - Cuiping Wang
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
- Department of Cardiology, The Affiliated Hospital of Jiangsu University, Jiangsu, Zhenjiang, China
| | - Jinchuan Yan
- Department of Cardiology, The Affiliated Hospital of Jiangsu University, Jiangsu, Zhenjiang, China
| | - Qichang Zheng
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anil Nanda
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
| | - D. Neil Granger
- Department of Physiology, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
| | - Guohong Li
- Vascular Biology and Stroke Research Laboratory, Department of Neurosurgery, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
- Department of Physiology, LSU Health Science Center in Shreveport, Shreveport, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
128
|
Chen R, Zhang F, Song L, Shu Y, Lin Y, Dong L, Nie X, Zhang D, Chen P, Han M. Transcriptome profiling reveals that the SM22α-regulated molecular pathways contribute to vascular pathology. J Mol Cell Cardiol 2014; 72:263-72. [DOI: 10.1016/j.yjmcc.2014.04.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 03/20/2014] [Accepted: 04/04/2014] [Indexed: 01/11/2023]
|
129
|
Bouzigues C, Nguyên TL, Ramodiharilafy R, Claeson A, Tharaux PL, Alexandrou A. Regulation of the ROS Response Dynamics and Organization to PDGF Motile Stimuli Revealed by Single Nanoparticle Imaging. ACTA ACUST UNITED AC 2014; 21:647-56. [DOI: 10.1016/j.chembiol.2014.02.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 02/07/2023]
|
130
|
Silpanisong J, Pearce WJ. Vasotrophic regulation of age-dependent hypoxic cerebrovascular remodeling. Curr Vasc Pharmacol 2014; 11:544-63. [PMID: 24063376 DOI: 10.2174/1570161111311050002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 06/08/2012] [Accepted: 07/12/2012] [Indexed: 02/07/2023]
Abstract
Hypoxia can induce functional and structural vascular remodeling by changing the expression of trophic factors to promote homeostasis. While most experimental approaches have been focused on functional remodeling, structural remodeling can reflect changes in the abundance and organization of vascular proteins that determine functional remodeling. Better understanding of age-dependent hypoxic macrovascular remodeling processes of the cerebral vasculature and its clinical implications require knowledge of the vasotrophic factors that influence arterial structure and function. Hypoxia can affect the expression of transcription factors, classical receptor tyrosine kinase factors, non-classical G-protein coupled factors, catecholamines, and purines. Hypoxia's remodeling effects can be mediated by Hypoxia Inducible Factor (HIF) upregulation in most vascular beds, but alterations in the expression of growth factors can also be independent of HIF. PPARγ is another transcription factor involved in hypoxic remodeling. Expression of classical receptor tyrosine kinase ligands, including vascular endothelial growth factor, platelet derived growth factor, fibroblast growth factor and angiopoietins, can be altered by hypoxia which can act simultaneously to affect remodeling. Tyrosine kinase-independent factors, such as transforming growth factor, nitric oxide, endothelin, angiotensin II, catecholamines, and purines also participate in the remodeling process. This adaptation to hypoxic stress can fundamentally change with age, resulting in different responses between fetuses and adults. Overall, these mechanisms integrate to assure that blood flow and metabolic demand are closely matched in all vascular beds and emphasize the view that the vascular wall is a highly dynamic and heterogeneous tissue with multiple cell types undergoing regular phenotypic transformation.
Collapse
Affiliation(s)
- Jinjutha Silpanisong
- Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | | |
Collapse
|
131
|
Toona sinensis inhibits LPS-induced inflammation and migration in vascular smooth muscle cells via suppression of reactive oxygen species and NF-κB signaling pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:901315. [PMID: 24723997 PMCID: PMC3960752 DOI: 10.1155/2014/901315] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/15/2014] [Accepted: 01/23/2014] [Indexed: 12/22/2022]
Abstract
Toona sinensis is one of the most popular vegetarian cuisines in Taiwan and it has been shown to possess antioxidant, antiangiogenic, and anticancer properties. In this study, we investigated the antiatherosclerotic potential of aqueous leaf extracts from Toona sinensis (TS; 25–100 μg/mL) and its major bioactive compound, gallic acid (GA; 5 μg/mL), in LPS-treated rat aortic smooth muscle (A7r5) cells. We found that pretreatment with noncytotoxic concentrations of TS and GA significantly inhibited inflammatory NO and PGE2 production by downregulating their precursors, iNOS and COX-2, respectively, in LPS-treated A7r5 cells. Furthermore, TS and GA inhibited LPS-induced intracellular ROS and their corresponding mediator, p47phox. Notably, TS and GA pretreatment significantly inhibited LPS-induced migration in transwell assays. Gelatin zymography and western blotting demonstrated that treatment with TS and GA suppressed the activity or expression of MMP-9, MMP-2, and t-PA. Additionally, TS and GA significantly inhibited LPS-induced VEGF, PDGF, and VCAM-1 expression. Further investigation revealed that the inhibition of iNOS/COX-2, MMPs, growth factors, and adhesion molecules was associated with the suppression of NF-κB activation and MAPK (ERK1/2, JNK1/2, and p38) phosphorylation. Thus, Toona sinensis may be useful for the prevention of atherosclerosis.
Collapse
|
132
|
Chaterji S, Lam CH, Ho DS, Proske DC, Baker AB. Syndecan-1 regulates vascular smooth muscle cell phenotype. PLoS One 2014; 9:e89824. [PMID: 24587062 PMCID: PMC3934950 DOI: 10.1371/journal.pone.0089824] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 01/24/2014] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE We examined the role of syndecan-1 in modulating the phenotype of vascular smooth muscle cells in the context of endogenous inflammatory factors and altered microenvironments that occur in disease or injury-induced vascular remodeling. METHODS AND RESULTS Vascular smooth muscle cells (vSMCs) display a continuum of phenotypes that can be altered during vascular remodeling. While the syndecans have emerged as powerful and complex regulators of cell function, their role in controlling vSMC phenotype is unknown. Here, we isolated vSMCs from wild type (WT) and syndecan-1 knockout (S1KO) mice. Gene expression and western blotting studies indicated decreased levels of α-smooth muscle actin (α-SMA), calponin, and other vSMC-specific differentiation markers in S1KO relative to WT cells. The spread area of the S1KO cells was found to be greater than WT cells, with a corresponding increase in focal adhesion formation, Src phosphorylation, and alterations in actin cytoskeletal arrangement. In addition, S1KO led to increased S6RP phosphorylation and decreased AKT and PKC-α phosphorylation. To examine whether these changes were present in vivo, isolated aortae from aged WT and S1KO mice were stained for calponin. Consistent with our in-vitro findings, the WT mice aortae stained higher for calponin relative to S1KO. When exposed to the inflammatory cytokine TNF-α, WT vSMCs had an 80% reduction in syndecan-1 expression. Further, with TNF-α, S1KO vSMCs produced increased pro-inflammatory cytokines relative to WT. Finally, inhibition of interactions between syndecan-1 and integrins αvβ3 and αvβ5 using the inhibitory peptide synstatin appeared to have similar effects on vSMCs as knocking out syndecan-1, with decreased expression of vSMC differentiation markers and increased expression of inflammatory cytokines, receptors, and osteopontin. CONCLUSIONS Taken together, our results support that syndecan-1 promotes vSMC differentiation and quiescence. Thus, the presence of syndecan-1 would have a protective effect against vSMC dedifferentiation and this activity is linked to interactions with integrins αvβ3 and αvβ5.
Collapse
Affiliation(s)
- Somali Chaterji
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Christoffer H. Lam
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Derek S. Ho
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Daniel C. Proske
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| | - Aaron B. Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
133
|
Li F, Wu JH, Wang QH, Shu YL, Wan CW, Chan CO, Kam-Wah Mok D, Chan SW. Gui-ling-gao, a traditional Chinese functional food, prevents oxidative stress-induced apoptosis in H9c2 cardiomyocytes. Food Funct 2014; 4:745-53. [PMID: 23467630 DOI: 10.1039/c3fo30182f] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Functional foods have become an increasingly popular alternative to prevent diseases and maintain body health status. Gui-ling-gao (GLG, also known as turtle jelly) is a well-known traditional functional food popular in Southern China and Hong Kong. This study aimed to investigate the antioxidative and anti-apoptotic effects of GLG, a traditional Chinese functional food, on preventing oxidative stress-induced injury in H9c2 cardiomyocytes. In this study, the antioxidative capacities of GLG were measured by using both a cell-free assay [2,2-diphenyl-1-(2,4,6-trinitrophenyl)hydrazyl assay] and biological methods [2,2'-azobis(2-amidinopropane)-induced haemolysis assay and H(2)O(2)-induced cell damage on H9c2 cardiomyocytes]. Additionally, the total phenolic content was measured using the Folin-Ciocalteu method. Furthermore, the anti-apoptotic effect of GLG was evaluated by nuclear staining and a DNA fragmentation assay. GLG was found to have good antioxidant activities and high total phenolic content. In H(2)O(2)-induced cell damage on H9c2 cells, GLG was demonstrated to ameliorate the apoptotic effects, such as nuclear condensations, increased intracellular caspase-3 activity and inter-nucleosomal DNA cleavage, induced by H(2)O(2). The present study demonstrated for the first time that GLG possesses anti-apoptotic potential in vitro and this effect may be mediated, in part, by its antioxidative function. Additionally, the antioxidative capacities of GLG were proved both chemically and biologically. This study provides scientific evidence to prove the anecdotal health-beneficial claim that the consumption of GLG could help the body to handle endogenous toxicants such as free radicals.
Collapse
Affiliation(s)
- Fan Li
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Shenzhen, PR China
| | | | | | | | | | | | | | | |
Collapse
|
134
|
Veit F, Pak O, Egemnazarov B, Roth M, Kosanovic D, Seimetz M, Sommer N, Ghofrani HA, Seeger W, Grimminger F, Brandes RP, Schermuly RT, Weissmann N. Function of NADPH oxidase 1 in pulmonary arterial smooth muscle cells after monocrotaline-induced pulmonary vascular remodeling. Antioxid Redox Signal 2013; 19:2213-31. [PMID: 23706097 DOI: 10.1089/ars.2012.4904] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AIMS Chronic hypoxia induces pulmonary hypertension (PH) that is concomitant with pulmonary vascular remodeling. Reactive oxygen species (ROS) are thought to play a major role in this. Recent findings suggest that ROS production by NADPH oxidase 4 (Nox4) is important in this remodeling. We investigated whether ROS production by Nox is also important in an inflammatory model of monocrotaline (MCT)-induced PH. We examined ROS production, their possible sources, and their impact on the function of pulmonary arterial smooth muscle cells (PASMC) isolated from MCT-treated and healthy rats. RESULTS MCT-PASMC showed increased intracellular superoxide production, migration, and proliferation compared with healthy controls due to increased Nox1 expression. A comparison of PASMC from MCT- and nontreated rats revealed an up-regulation of Sod2, Nrf2, cyclin D1, and matrix metalloproteinase-9 (MMP-9) as well as an increased phosphorylation of cofilin and extracellular signal-regulated kinases (Erk). Expression of Sod2, Nrf2, and cyclin D1 and phosphorylation of cofilin and Erk were Nox1 dependent. INNOVATION The role of ROS in PH is not fully understood. Mitochondria and Nox have been suggested as sources of altered ROS generation in PH, yet it remains unclear whether increased or decreased ROS contributes to the development of PH. Our studies provide evidence that for different triggers of PH, different Nox isoforms regulate proliferation and migration of PASMC. CONCLUSION In contrast to hypoxia-induced PH, Nox1 but not Nox4 is responsible for pathophysiological proliferation and migration of PASMC in an inflammatory model of MCT-induced PH via increased superoxide production. Thus, different Nox isoforms may be targeted in different forms of PH.
Collapse
Affiliation(s)
- Florian Veit
- 1 Excellencecluster Cardio-Pulmonary System (ECCPS), German Lung Center (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), Justus-Liebig-University Giessen , Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Liu CC, Karimi Galougahi K, Weisbrod RM, Hansen T, Ravaie R, Nunez A, Liu YB, Fry N, Garcia A, Hamilton EJ, Sweadner KJ, Cohen RA, Figtree GA. Oxidative inhibition of the vascular Na+-K+ pump via NADPH oxidase-dependent β1-subunit glutathionylation: implications for angiotensin II-induced vascular dysfunction. Free Radic Biol Med 2013; 65:563-572. [PMID: 23816524 PMCID: PMC4474148 DOI: 10.1016/j.freeradbiomed.2013.06.040] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 02/07/2023]
Abstract
Glutathionylation of the Na(+)-K(+) pump's β1-subunit is a key molecular mechanism of physiological and pathophysiological pump inhibition in cardiac myocytes. Its contribution to Na(+)-K(+) pump regulation in other tissues is unknown, and cannot be assumed given the dependence on specific β-subunit isoform expression and receptor-coupled pathways. As Na(+)-K(+) pump activity is an important determinant of vascular tone through effects on [Ca(2+)]i, we have examined the role of oxidative regulation of the Na(+)-K(+) pump in mediating angiotensin II (Ang II)-induced increases in vascular reactivity. β1-subunit glutathione adducts were present at baseline and increased by exposure to Ang II in rabbit aortic rings, primary rabbit aortic vascular smooth muscle cells (VSMCs), and human arterial segments. In VSMCs, Ang II-induced glutathionylation was associated with marked reduction in Na(+)-K(+)ATPase activity, an effect that was abolished by the NADPH oxidase inhibitory peptide, tat-gp91ds. In aortic segments, Ang II-induced glutathionylation was associated with decreased K(+)-induced vasorelaxation, a validated index of pump activity. Ang II-induced oxidative inhibition of Na(+)-K(+) ATPase and decrease in K(+)-induced relaxation were reversed by preincubation of VSMCs and rings with recombinant FXYD3 protein that is known to facilitate deglutathionylation of β1-subunit. Knock-out of FXYD1 dramatically decreased K(+)-induced relaxation in a mouse model. Attenuation of Ang II signaling in vivo by captopril (8 mg/kg/day for 7 days) decreased superoxide-sensitive DHE levels in the media of rabbit aorta, decreased β1-subunit glutathionylation, and enhanced K(+)-induced vasorelaxation. Ang II inhibits the Na(+)-K(+) pump in VSMCs via NADPH oxidase-dependent glutathionylation of the pump's β1-subunit, and this newly identified signaling pathway may contribute to altered vascular tone. FXYD proteins reduce oxidative inhibition of the Na(+)-K(+) pump and may have an important protective role in the vasculature under conditions of oxidative stress.
Collapse
Affiliation(s)
- Chia-Chi Liu
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Keyvan Karimi Galougahi
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia
| | - Robert M Weisbrod
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Thomas Hansen
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Ramtin Ravaie
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Andrea Nunez
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Yi B Liu
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Natasha Fry
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Alvaro Garcia
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Elisha J Hamilton
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia
| | - Kathleen J Sweadner
- Laboratory Membrane Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Richard A Cohen
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute of Medical Research, University of Sydney, Australia; Department of Cardiology, Royal North Shore Hospital, Sydney, Australia.
| |
Collapse
|
136
|
Soe NN, Sowden M, Baskaran P, Kim Y, Nigro P, Smolock EM, Berk BC. Acetylation of cyclophilin A is required for its secretion and vascular cell activation. Cardiovasc Res 2013; 101:444-53. [PMID: 24293519 DOI: 10.1093/cvr/cvt268] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Cyclophilin A (CyPA) is a pro-inflammatory mediator involved in oxidative stress-related cardiovascular diseases. It is secreted from vascular smooth muscle cell (VSMC) in response to reactive oxygen species (ROS) in a highly regulated manner. Extracellular CyPA activates VSMCs and endothelial cells (ECs) promoting inflammation, cell growth, and cell death. Recently, it was shown that acetylated CyPA (AcK-CyPA) affects its function. We investigated the role of acetylation of CyPA for its secretion and signalling in vascular cells. METHODS AND RESULTS We used angiotensin II (Ang II) to create sustained ROS and found significantly increased AcK-CyPA in VSMC. Site-directed mutagenesis showed that lysines K82 and K125 were the predominant CyPA residues acetylated in response to Ang II. Importantly, acetylation of K82 and K125 were required for Ang II-mediated CyPA secretion. ROS inhibitors, Tiron, and N-acetylcysteine inhibited Ang II-induced intracellular CyPA acetylation and also AcK-CyPA secretion. Using secreted CyPA from wild type and K82/125R mutants expressed in transduced VSMC or in vitro acetylated recombinant CyPA, we showed that extracellular AcK-CyPA significantly increased pERK1/2, matrix metalloproteinase-2 activation, and ROS production in VSMC compared with non-acetylated CyPA. Moreover, extracellular AcK-CyPA increased adhesion molecule expression (VCAM-1 and ICAM-1) in EC, which promoted monocyte adhesion. CONCLUSIONS ROS-dependent acetylation of CyPA is required for the generation of extracellular CyPA. Acetylated extracellular CyPA regulates VSMC and EC activation, suggesting that inhibition of acetylation of CyPA may prevent the pathogenesis of oxidative stress-related cardiovascular diseases.
Collapse
Affiliation(s)
- Nwe Nwe Soe
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Box CVRI, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | |
Collapse
|
137
|
Wang Y, Ji L, Jiang R, Zheng L, Liu D. Oxidized high-density lipoprotein induces the proliferation and migration of vascular smooth muscle cells by promoting the production of ROS. J Atheroscler Thromb 2013; 21:204-16. [PMID: 24225481 DOI: 10.5551/jat.19448] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIM As the major atheroprotective particle in plasma, high-density lipoprotein(HDL) is oxidized during atherosclerotic processes. Oxidized HDL(ox-HDL) may lose its cardioprotective properties and develop a proinflammatory and proatherogenic phenotype. The proliferation and migration of vascular smooth muscle cells(VSMCs) play a crucial role in atherogenesis. However, the influence of ox-HDL on VSMC proliferation and migration remains poorly understood. METHODS VSMCs were treated with native HDL(N-HDL) or ox-HDL at varying concentrations for different time intervals and used in several analyses. The degree of cell proliferation was assayed using CCK-8 kits. The level of cell migration was determined using a Transwell chamber and scratch-wound assay. The presence of intracellular reactive oxygen species(ROS) was detected based on ROS-mediated 2',7'-dichlorofluorescein fluorescence. The activation of NADPH oxidase was measured in terms of the Rac1 activity and NADP(+)/NADPH ratio. RESULTS Compared to N-HDL, ox-HDL significantly promoted VSMC proliferation and migration in a dose-dependent manner. In addition, ox-HDL remarkably activated NADPH oxidase and enhanced ROS generation in the VSMCs. Diphenyleneiodonium chloride, an inhibitor of NADPH oxidase, and N-acetylcysteine, a ROS scavenger, efficiently inhibited the ROS production triggered by ox-HDL and subsequently blocked the proliferating and migrating effects of ox-HDL in the VSMCs. CONCLUSIONS Ox-HDL significantly induces VSMC proliferation and migration by promoting NADPH oxidase activation and ROS production. Furthermore, the inhibition of NADPH oxidase and ROS generation blocks the proliferation and migration of VSMCs induced by ox-HDL. These proliferating and migrating effects of ox-HDL are closely related to its proinflammatory and proatherogenic roles.
Collapse
Affiliation(s)
- Yan Wang
- Division of Cardiology, the Affiliated Zhongshan Hospital of Xiamen University, Xiamen Heart Center
| | | | | | | | | |
Collapse
|
138
|
A reactive oxygen species-mediated, self-perpetuating loop persistently activates platelet-derived growth factor receptor α. Mol Cell Biol 2013; 34:110-22. [PMID: 24190966 DOI: 10.1128/mcb.00839-13] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The platelet-derived growth factor (PDGF) receptors (PDGFRs) are central to a spectrum of human diseases. When PDGFRs are activated by PDGF, reactive oxygen species (ROS) and Src family kinases (SFKs) act downstream of PDGFRs to enhance PDGF-mediated tyrosine phosphorylation of various signaling intermediates. In contrast to these firmly established principles of signal transduction, much less is known regarding the recently appreciated ability of ROS and SFKs to indirectly and chronically activate monomeric PDGF receptor α (PDGFRα) in the setting of a blinding condition called proliferative vitreoretinopathy (PVR). In this context, we made a series of discoveries that substantially expands our appreciation of epigenetic-based mechanisms to chronically activate PDGFRα. Vitreous, which contains growth factors outside the PDGF family but little or no PDGFs, promoted formation of a unique SFK-PDGFRα complex that was dependent on SFK-mediated phosphorylation of PDGFRα and activated the receptor's kinase activity. While vitreous engaged a total of five receptor tyrosine kinases, PDGFRα was the only one that was activated persistently (at least 16 h). Prolonged activation of PDGFRα involved mTOR-mediated inhibition of autophagy and accumulation of mitochondrial ROS. These findings reveal that growth factor-containing biological fluids, such as vitreous, are able to tirelessly activate PDGFRα by engaging a ROS-mediated, self-perpetuating loop.
Collapse
|
139
|
Blazevic T, Schwaiberger AV, Schreiner CE, Schachner D, Schaible AM, Grojer CS, Atanasov AG, Werz O, Dirsch VM, Heiss EH. 12/15-lipoxygenase contributes to platelet-derived growth factor-induced activation of signal transducer and activator of transcription 3. J Biol Chem 2013; 288:35592-603. [PMID: 24165129 PMCID: PMC3853304 DOI: 10.1074/jbc.m113.489013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We showed previously that the small molecule indirubin-3′-monoxime (I3MO) prevents vascular smooth muscle cell (VSMC) proliferation by selectively inhibiting signal transducer and activator of transcription 3 (STAT3). Looking for the underlying upstream molecular mechanism, we here reveal the important role of reactive oxygen species (ROS) for PDGF-induced STAT3 activation in VSMC. We show that neither NADPH-dependent oxidases (Noxes) nor mitochondria, but rather 12/15-lipoxygenase (12/15-LO) are pivotal ROS sources involved in the redox-regulated signal transduction from PDGFR to STAT3. Accordingly, pharmacological and genetic interference with 12/15-LO activity selectively inhibited PDGF-induced Src activation and STAT3 phosphorylation. I3MO is able to blunt PDGF-induced ROS and 15(S)-hydroxyeicosatetraenoic acid (15(S)-HETE) production, indicating an inhibitory action of I3MO on 12/15-LO and consequently on STAT3. We identify 12/15-LO as a hitherto unrecognized signaling hub in PDGF-triggered STAT3 activation and show for the first time a negative impact of I3MO on 12/15-LO.
Collapse
Affiliation(s)
- Tina Blazevic
- From the Department for Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria and
| | | | | | | | | | | | | | | | | | | |
Collapse
|
140
|
Kato M, Castro NE, Natarajan R. MicroRNAs: potential mediators and biomarkers of diabetic complications. Free Radic Biol Med 2013; 64:85-94. [PMID: 23770198 PMCID: PMC3762900 DOI: 10.1016/j.freeradbiomed.2013.06.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 06/04/2013] [Accepted: 06/04/2013] [Indexed: 02/07/2023]
Abstract
The incidence of diabetes is escalating worldwide and, consequently, this has become a major health care problem. Moreover, both type 1 and type 2 diabetes are associated with significantly accelerated rates of microvascular complications, including retinopathy, nephropathy, and neuropathy, as well as macrovascular complications such as atherosclerotic cardiovascular and hypertensive diseases. Key factors have been implicated in leading to these complications, including hyperglycemia, insulin resistance, dyslipidemia, advanced glycation end products, growth factors, inflammatory cytokines/chemokines, and related increases in cellular oxidant stress (including mitochondrial) and endoplasmic reticulum stress. However, the molecular mechanisms underlying the high incidence of diabetic complications, which often progress despite glycemic control, are still not fully understood. MicroRNAs (miRNAs) are short noncoding RNAs that have elicited immense interest in recent years. They repress target gene expression via posttranscriptional mechanisms and have diverse cellular and biological functions. Herein, we discuss the role of miRNAs in the pathobiology of various diabetic complications, their involvement in oxidant stress, and also the potential use of differentially expressed miRNAs as novel diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Mitsuo Kato
- Department of Diabetes, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | | | | |
Collapse
|
141
|
Oxidative stress and microRNAs in vascular diseases. Int J Mol Sci 2013; 14:17319-46. [PMID: 23975169 PMCID: PMC3794730 DOI: 10.3390/ijms140917319] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress has been demonstrated to play a causal role in different vascular diseases, such as hypertension, diabetic vasculopathy, hypercholesterolemia and atherosclerosis. Indeed, increased reactive oxygen species (ROS) production is known to impair endothelial and vascular smooth muscle cell functions, contributing to the development of cardiovascular diseases. MicroRNAs (miRNAs) are non-coding RNA molecules that modulate the stability and/or the translational efficiency of target messenger RNAs. They have been shown to be modulated in most biological processes, including in cellular responses to redox imbalance. In particular, miR-200 family members play a crucial role in oxidative-stress dependent endothelial dysfunction, as well as in cardiovascular complications of diabetes and obesity. In addition, different miRNAs, such as miR-210, have been demonstrated to play a key role in mitochondrial metabolism, therefore modulating ROS production and sensitivity. In this review, we will discuss miRNAs modulated by ROS or involved in ROS production, and implicated in vascular diseases in which redox imbalance has a pathogenetic role.
Collapse
|
142
|
Norton CE, Broughton BRS, Jernigan NL, Walker BR, Resta TC. Enhanced depolarization-induced pulmonary vasoconstriction following chronic hypoxia requires EGFR-dependent activation of NAD(P)H oxidase 2. Antioxid Redox Signal 2013; 18:1777-88. [PMID: 22966991 PMCID: PMC3619151 DOI: 10.1089/ars.2012.4836] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
AIMS Chronic hypoxia (CH) enhances depolarization-induced myofilament Ca(2+) sensitization and resultant pulmonary arterial constriction through superoxide (O(2)(-))-dependent stimulation of RhoA. Because NAD(P)H oxidase (NOX) has been implicated in the development of pulmonary hypertension, we hypothesized that vascular smooth muscle (VSM) depolarization increases NOX-derived O(2)(-) production leading to myofilament Ca(2+) sensitization and augmented vasoconstrictor reactivity following CH. As epidermal growth factor receptor (EGFR) mediates Rac1-dependent NOX activation in renal mesangial cells, we further sought to examine the role EGFR plays in this response. RESULTS Vasoconstrictor responses to depolarizing concentrations of KCl were greater in lungs isolated from CH (4 wk, 0.5 atm) rats compared to normoxic controls, and this effect of CH was abolished by the general NOX inhibitor, apocynin. CH similarly augmented KCl-induced vasoconstriction and O(2)(-) generation (assessed using the fluorescent indicator, dihydroethidium) in Ca(2+)-permeabilized, pressurized small pulmonary arteries. These latter responses to CH were prevented by general inhibition of NOX isoforms (apocynin, diphenylene iodonium), and by selective inhibition of NOX 2 (gp91ds-tat), Rac1 (NSC 23766), and EGFR (AG 1478). Consistent with these observations, CH increased KCl-induced EGFR phosphorylation, and augmented depolarization-induced Rac1 activation in an EGFR-dependent manner. INNOVATION This study establishes a novel signaling axis in VSM linking membrane depolarization to contraction that is independent of Ca(2+) influx, and which mediates myofilament Ca(2+) sensitization in the hypertensive pulmonary circulation. CONCLUSION CH augments membrane depolarization-induced pulmonary VSM Ca(2+) sensitization and vasoconstriction through EGFR-dependent stimulation of Rac1 and NOX 2.
Collapse
Affiliation(s)
- Charles E Norton
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | | | | | | | | |
Collapse
|
143
|
Alcantara EH, Shin MY, Feldmann J, Nixon GF, Beattie JH, Kwun IS. Long-term zinc deprivation accelerates rat vascular smooth muscle cell proliferation involving the down-regulation of JNK1/2 expression in MAPK signaling. Atherosclerosis 2013; 228:46-52. [PMID: 23466072 DOI: 10.1016/j.atherosclerosis.2013.01.030] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 12/30/2012] [Accepted: 01/25/2013] [Indexed: 11/25/2022]
Abstract
BACKGROUND The accelerated proliferation of vascular smooth muscle cells (VSMCs) is a contributor for atherosclerosis by thickening the vascular wall. Since zinc modulation of VSMC proliferation has not been clarified, this study investigated whether zinc affects VSMC proliferation. METHODS AND RESULTS Both a rat aorta origin vascular smooth muscle cell line (A7r5 VSMCs) and primary VSMCs which were collected from rat aorta (pVSMCs) were cultured with zinc (0-50 μM Zn) for short- (≤12 d) and long-term (28 d) periods under normal non-calcifying (0 or 1 mM P) or calcifying (>2 mM P) P conditions. Mouse vascular endothelial cells (MS I cells) were also cultured (under 0-50 μM Zn and 10 mM P for 20 d) to compare with VSMC cultures. While during short-term culture of VSMCs, zinc deprivation decreased cell proliferation in a zinc-concentration manner both under non-calcifying and calcifying conditions in A7r5 and pVSMCs (P < 0.05), during long-term cultures (28 d), A7r5 VSMC proliferation was inversely related to medium zinc concentration under normal physiological P conditions (regression coefficient r(2) = -0.563, P = 0.012). The anti-cell proliferative effect of zinc supplementation (>50 μM) was VSMC-specific. Long-term (35 d), low zinc treatment down-regulated JNK expression and activation, while not affecting ERK1/2 MAPK signaling in A7r5 VSMCs. CONCLUSION The results showed that chronic zinc deprivation accelerated VSMC proliferation, perhaps due to down-regulation of MAPK-JNK signaling, and that the anti-cell proliferative role of zinc is VSMC-specific. The findings suggested that zinc may have anti-VSMC proliferative properties in atherosclerosis.
Collapse
Affiliation(s)
- Ethel H Alcantara
- Department of Food Science and Nutrition, Andong National University, 388 Songchundong, Andong, Kyungbook 760-749, South Korea.
| | | | | | | | | | | |
Collapse
|
144
|
Transforming growth factor-β1 induces matrix metalloproteinase-9 expression in rat vascular smooth muscle cells via ROS-dependent ERK-NF-κB pathways. Mol Cell Biochem 2012; 375:11-21. [PMID: 23275087 DOI: 10.1007/s11010-012-1512-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 11/07/2012] [Indexed: 02/07/2023]
Abstract
Both matrix metalloproteinase-9 (MMP9) and transforming growth factors-β1 (TGF-β1) are the important factors in the pathogenesis of the aortic aneurysm (AA) and aortic dissection (AD). Recent studies have shown that inhibition of reactive oxygen species (ROS) production, extracellular signal-regulated kinase 1/2(ERK1/2) or NF-κB pathways is able to suppress aneurysm formation. The median layers of arterial walls are mainly the vascular smooth muscle cells (VSMCs), while the pathogenesis of AA and AD is closely related to the changes in the median layer structure. Thus, we investigated the molecular mechanisms underlying TGF-β1-induced MMP-9 expression in VSMC, the involvement of intracellular ROS and signaling molecules, including ERK1/2 and NF-κB. Rat vascular smooth muscle cells (A7r5) were used. MMP-9 expression was analyzed by gelatin zymography, western blot and RT-PCR. The involvement of intracellular ROS and signaling molecules including ERK1/2 and NF-κB in the responses was investigated using reactive oxygen scavenger N-acetylcysteine (NAC) and pharmacological inhibitors (U0126 and BAY11-7082), determined by ROS testing and western blot testing for their corresponding proteins. TGF-β1 induces MMP-9 expression via ROS-dependent signaling pathway. ROS production leads to activation of ERK1/2 and then activation of the NF-κB transcription factor. Activated NF-κB turns on transcription of the MMP-9 gene. The process in which TGF-β1 induces MMP9 expression involves the ROS-dependent ERK-NF-κB signal pathways in VSMC. This discovery raises a new regulation pathway in the VSMC, and it shows the potential to help to find a new solution to treating aortic aneurysm and aortic dissection.
Collapse
|
145
|
Yu M, Gong D, Lim M, Arutyunyan A, Groffen J, Heisterkamp N. Lack of bcr and abr promotes hypoxia-induced pulmonary hypertension in mice. PLoS One 2012; 7:e49756. [PMID: 23152932 PMCID: PMC3495860 DOI: 10.1371/journal.pone.0049756] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 10/16/2012] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Bcr and Abr are GTPase activating proteins that specifically downregulate activity of the small GTPase Rac in restricted cell types in vivo. Rac1 is expressed in smooth muscle cells, a critical cell type involved in the pathogenesis of pulmonary hypertension. The molecular mechanisms that underlie hypoxia-associated pulmonary hypertension are not well-defined. METHODOLOGY/PRINCIPAL FINDINGS Bcr and abr null mutant mice were compared to wild type controls for the development of pulmonary hypertension after exposure to hypoxia. Also, pulmonary arterial smooth muscle cells from those mice were cultured in hypoxia and examined for proliferation, p38 activation and IL-6 production. Mice lacking Bcr or Abr exposed to hypoxia developed increased right ventricular pressure, hypertrophy and pulmonary vascular remodeling. Perivascular leukocyte infiltration in the lungs was increased, and under hypoxia bcr-/- and abr-/- macrophages generated more reactive oxygen species. Consistent with a contribution of inflammation and oxidative stress in pulmonary hypertension-associated vascular damage, Bcr and Abr-deficient animals showed elevated endothelial leakage after hypoxia exposure. Hypoxia-treated pulmonary arterial smooth muscle cells from Bcr- or Abr-deficient mice also proliferated faster than those of wild type mice. Moreover, activated Rac1, phosphorylated p38 and interleukin 6 were increased in these cells in the absence of Bcr or Abr. Inhibition of Rac1 activation with Z62954982, a novel Rac inhibitor, decreased proliferation, p38 phosphorylation and IL-6 levels in pulmonary arterial smooth muscle cells exposed to hypoxia. CONCLUSIONS Bcr and Abr play a critical role in down-regulating hypoxia-induced pulmonary hypertension by deactivating Rac1 and, through this, reducing both oxidative stress generated by leukocytes as well as p38 phosphorylation, IL-6 production and proliferation of pulmonary arterial smooth muscle cells.
Collapse
Affiliation(s)
- Min Yu
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - Dapeng Gong
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - Min Lim
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Anna Arutyunyan
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
| | - John Groffen
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Nora Heisterkamp
- Section of Molecular Carcinogenesis, Division of Hematology/Oncology, and The Saban Research Institute of Children’s Hospital, Los Angeles, California, United States of America
- Departments of Pediatrics and Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
146
|
Lee KM, Kang HA, Park M, Lee HY, Song MJ, Ko K, Oh JW, Kang HS. Interleukin-24 Suppresses the Growth of Vascular Smooth Muscle Cells by Inhibiting H 2O 2-Induced Reactive Oxygen Species Production. Pharmacology 2012; 90:332-41. [DOI: 10.1159/000343242] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/04/2012] [Indexed: 11/19/2022]
|
147
|
Lv P, Miao SB, Shu YN, Dong LH, Liu G, Xie XL, Gao M, Wang YC, Yin YJ, Wang XJ, Han M. Phosphorylation of Smooth Muscle 22α Facilitates Angiotensin II–Induced ROS Production Via Activation of the PKCδ-P47
phox
Axis Through Release of PKCδ and Actin Dynamics and Is Associated With Hypertrophy and Hyperplasia of Vascular Smooth Muscle Cells In Vitro and In Vivo. Circ Res 2012; 111:697-707. [DOI: 10.1161/circresaha.112.272013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
We have demonstrated that smooth muscle (SM) 22α inhibits cell proliferation via blocking Ras-ERK1/2 signaling in vascular smooth muscle cells (VSMCs) and in injured arteries. The recent study indicates that SM22α disruption can independently promote arterial inflammation through activation of reactive oxygen species (ROS)-mediated NF-κB pathways. However, the mechanisms by which SM22α controls ROS production have not been characterized.
Objective:
To investigate how SM22α disruption promotes ROS production and to characterize the underlying mechanisms.
Methods and Results:
ROS level was measured by dihydroethidium staining for superoxide and TBA assay for malondialdehyde, respectively. We showed that downregulation and phosphorylation of SM22α were associated with angiotensin (Ang) II–induced increase in ROS production in VSMCs of rats and human. Ang II induced the phosphorylation of SM22α at Serine 181 in an Ang II type 1 receptor–PKCδ pathway–dependent manner. Phosphorylated SM22α activated the protein kinase C (PKC)δ-p47
phox
axis via 2 distinct pathways: (1) disassociation of PKCδ from SM22α, and in turn binding to p47
phox
, in the early stage of Ang II stimulation; and (2) acceleration of SM22α degradation through ubiquitin-proteasome, enhancing PKCδ membrane translocation via induction of actin cytoskeletal dynamics in later oxidative stress. Inhibition of SM22α phosphorylation abolished the Ang II–activated PKCδ-p47
phox
axis and inhibited the hypertrophy and hyperplasia of VSMCs in vitro and in vivo, accompanied with reduction of ROS generation.
Conclusions:
These findings indicate that the disruption of SM22α plays pivotal roles in vascular oxidative stress. PKCδ-mediated SM22α phosphorylation is a novel link between actin cytoskeletal remodeling and oxidative stress and may be a potential target for the development of new therapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Pin Lv
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Sui-Bing Miao
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Ya-Nan Shu
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Li-Hua Dong
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - George Liu
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Xiao-Li Xie
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Min Gao
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Yu-Can Wang
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Ya-Juan Yin
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Xiao-Juan Wang
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| | - Mei Han
- From the Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang, China (P.L., S.-B.M., Y.-N.S., L.-H.D., X.-L.X., M.G., Y.-C.W., Y.-J.Y., X.-J.W., M.H.); and The Institute of Cardiovascular Sciences, Peking University and Key Laboratory of Cardiovascular Sciences, China Administration of Education, Peking University, Beijing, China (G.L.)
| |
Collapse
|
148
|
Ramachandran S, Kartha C. Cyclophilin-A: a potential screening marker for vascular disease in type-2 diabetes. Can J Physiol Pharmacol 2012; 90:1005-15. [DOI: 10.1139/y2012-038] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathophysiology of vascular disease in diabetes involves abnormalities in endothelial cells, vascular smooth muscle cells, and monocytes. The metabolic abnormalities that characterize diabetes, such as hyperglycemia, increased free fatty acids, and insulin resistance, each provoke molecular mechanisms that contribute to vascular dysfunction. Several molecules have been identified as risk markers, and have been studied to prevent progression of disease and long-term complications. Markers such as C-reactive protein and monocyte chemoattractant protein-1 are used to assess risk for adverse cardiac events, but elevated levels are possible due to the presence of other risk factors as part of the natural physiological defense mechanism. In this review we discuss potential of cyclophilin-A, a secreted oxidative-stress-induced immunophilin with diverse functions. We present evidence for a significant role of cyclophilin-A in the pathogenesis of atherosclerosis in diabetes, and its potential as a marker for vascular disease in type-2 diabetes.
Collapse
Affiliation(s)
- Surya Ramachandran
- Cardiovascular Disease Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695 014, India
| | - C.C. Kartha
- Cardiovascular Disease Biology Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695 014, India
| |
Collapse
|
149
|
Huh JY, Son DJ, Lee Y, Lee J, Kim B, Lee HM, Jo H, Choi S, Ha H, Chung MH. 8-Hydroxy-2-deoxyguanosine prevents plaque formation and inhibits vascular smooth muscle cell activation through Rac1 inactivation. Free Radic Biol Med 2012; 53:109-21. [PMID: 22580124 PMCID: PMC5489255 DOI: 10.1016/j.freeradbiomed.2012.03.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 02/16/2012] [Accepted: 03/12/2012] [Indexed: 12/23/2022]
Abstract
8-Hydroxy-2-deoxyguanosine (8-OHdG), a marker of oxidative stress, has been recently rediscovered to inhibit Rac1 in neutrophils and macrophages, thereby inhibiting Rac1-linked functions of these cells, including reactive oxygen species production through NADPH oxidase activation, phagocytosis, chemotaxis, and cytokine release. In vascular smooth muscle cells (VSMCs), reactive oxygen species also induce abnormal proliferation and migration leading to progression of atherosclerosis. Based upon the involvement of reactive oxygen species in phagocytic cells and VSMCs during the atherosclerotic process, we hypothesized that 8-OHdG could have antiatherosclerotic action and tested this hypothesis in an experimentally induced atherosclerosis in mice. Partially ligated ApoE knockout mice, a more physiologically relevant model of low and oscillatory flow, developed an advanced lesion in 2 weeks, and orally administered 8-OHdG significantly reduced plaque formation along with reduced superoxide formation, monocyte/macrophage infiltration, and extracellular matrix (ECM) accumulation. The effects of 8-OHdG observed in primary VSMCs were consistent with the in vivo effects of 8-OHdG and were inhibitory to angiotensin II or platelet-derived growth factor-induced production of reactive oxygen species, proliferation, migration, and ECM production. Also, angiotensin II-induced Rac1 activity in VSMCs was significantly inhibited by 8-OHdG, and transfection of constitutively active Rac1 reversed the inhibitory effect of 8-OHdG on VSMC activation. Molecular docking study showed that 8-OHdG stabilizes Rac1-GEF complex, indicating the physical contact of 8-OHdG with Rac1. These findings highly suggest that the antiatherosclerotic effect of 8-OHdG is mediated by inhibition of Rac1 activity. In conclusion, our results show a novel action of orally active 8-OHdG in suppressing atherosclerotic plaque formation in vivo and VSMC activation in vitro through inhibition of Rac1, which emphasizes a new therapeutic avenue to benefit atherosclerosis.
Collapse
MESH Headings
- 8-Hydroxy-2'-Deoxyguanosine
- Angiotensin II/pharmacology
- Animals
- Apolipoproteins E/physiology
- Blotting, Western
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Chemotaxis/drug effects
- Deoxyguanosine/analogs & derivatives
- Deoxyguanosine/pharmacology
- Immunoenzyme Techniques
- Male
- Mice
- Mice, Knockout
- Models, Molecular
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Oxidative Stress/drug effects
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/prevention & control
- Reactive Oxygen Species/metabolism
- Superoxides/metabolism
- Vasoconstrictor Agents/pharmacology
- rac1 GTP-Binding Protein/antagonists & inhibitors
- rac1 GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Joo Young Huh
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Dong Ju Son
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Yoonji Lee
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Junghyun Lee
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Boyeon Kim
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
| | - Hwan Myung Lee
- Department of Cosmetic Science, College of Natural Sciences, Hoseo University, Asan, Korea
| | - Hanjoong Jo
- Department of Bioinspired Science, Ewha Womans University, Seoul 120-750, Korea
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Sun Choi
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
- Corresponding author. (S. Choi), (H. Ha)
| | - Hunjoo Ha
- Division of Life & Pharmaceutical Sciences and Center for Cell Signaling & Drug Discovery Research, College of Pharmacy, Ewha Womans University, Seoul 120-750, Korea
- Department of Bioinspired Science, Ewha Womans University, Seoul 120-750, Korea
- Corresponding author. Fax: +82 2 3277 2851
| | - Myung-Hee Chung
- Samsung Advanced Institute for Health Sciences & Technology, Sung Kyun Kwan University, Seoul, Korea
| |
Collapse
|
150
|
Dou D, Zheng X, Liu J, Xu X, Ye L, Gao Y. Hydrogen peroxide enhances vasodilatation by increasing dimerization of cGMP-dependent protein kinase type Iα. Circ J 2012; 76:1792-8. [PMID: 22498562 DOI: 10.1253/circj.cj-11-1368] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND cGMP-dependent protein kinase type I (PKG I) plays a key role in vasodilatation caused by cGMP-elevating agents. It is a homodimer in mammalian cells, existing as 2 isoforms, Iα and Iβ. The aim of the present study was both to determine whether PKG I dimerization and activity are modulated by hydrogen peroxide (H(2)O(2)) and its influence on vasodilatation. METHODS AND RESULTS The dimers and monomers of total PKG I and PKG Iβ were analyzed by Western blotting. PKG I activity was assayed by measuring the incorporation of (32)P into BPDEtide. Changes in vessels tension were determined by organ chamber technique. In isolated porcine coronary arteries, H(2)O(2) increased the dimers of total PKG I in a concentration-dependent manner, but had no effect on dimerization of PKG Iβ. The dimerization of PKG I caused by H(2)O(2) was prevented by catalase but not by deferoxamine and tiron. H(2)O(2) promoted the translocation of PKG I from cytoplasm to membrane. H(2)O(2) enhanced the activity of PKG I and relaxations of porcine coronary arteries to the nitric oxide donor and 8-Br-cGMP. Inhibition of catalase under in vivo conditions significantly decreased rat mean arterial pressure, which was associated with increased dimerization of PKG I. CONCLUSIONS The present study suggests that H(2)O(2) may enhance the activity of PKG Iα-and PKG I-dependent vasodilatation via increased dimerization of the enzyme.
Collapse
Affiliation(s)
- Dou Dou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | |
Collapse
|