1
|
Tao Y, Li G, Yang Y, Wang Z, Wang S, Li X, Yu T, Fu X. Epigenomics in aortic dissection: From mechanism to therapeutics. Life Sci 2023; 335:122249. [PMID: 37940070 DOI: 10.1016/j.lfs.2023.122249] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Aortic dissection (AD) has an unfavorable prognosis. It requires early diagnosis, appropriate treatment strategies, and suspicion to recognize symptoms; thus, it is commonly described as an acute aortic emergency. The clinical manifestations of painless AD are complex and variable. However, there is no effective treatment to prevent the progression of AD. Therefore, study of the molecular targets and mechanisms of AD to enable prevention or early intervention is particularly important. Although multiple gene mutations have been proposed as linked to AD development, evidence that multiple epigenetic elements are strongly associated is steadily increasing. These epigenetic processes include DNA methylation, N6-methyladenosine, histone modification, non-histone posttranslational modification, and non-coding RNAs (ncRNAs). Among these processes, resveratrol targeting Sirtuin 1 (SIRT1), 5-azacytidine (5azaC) targeting DNA methyltransferase (DNMT), and vitamin C targeting ten-eleven translocation 2 (Tet2) showed unique advantages in improving AD and vascular dysfunction. Finally, we explored potential epigenetic drugs and diagnostic methods for AD, which might provide options for the future.
Collapse
Affiliation(s)
- Yan Tao
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Gang Li
- Department of Vascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, Shandong 250021, China; Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, Basic Medicine School, Qingdao University, No. 308 Ningxia Road, Qingdao 266071, People's Republic of China
| | - Zhibin Wang
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Shizhong Wang
- The department of Cardiology surgery, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China
| | - Tao Yu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao 266021, People's Republic of China.
| | - Xiuxiu Fu
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao 266000, People's Republic of China.
| |
Collapse
|
2
|
Brownfoot F, Binder N, Hastie R, Harper A, Beard S, Tuohey L, Keenan E, Tong S, Hannan N. Nicotinamide and its effects on endothelial dysfunction and secretion of antiangiogenic factors by primary human placental cells and tissues. Placenta 2021; 109:28-31. [PMID: 33957335 DOI: 10.1016/j.placenta.2021.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 01/08/2023]
Abstract
Preeclampsia is a serious pregnancy complication associated with elevated antiangiogenic markers and endothelial dysfunction. Recently nicotinamide (vitamin B3) was shown to reduce high blood pressure and proteinuria in mice models of the disease. Using primary human pregnancy tissue we show nicotinamide did not change antiangiogenic factor secretion including soluble fms-like tyrosine kinase 1 or soluble endoglin from primary cytotrophoblasts and placental explants. Furthermore, it did not reverse markers of endothelial dysfunction. Therefore, we did not demonstrate an effect of nicotinamide on reducing markers of preeclampsia from primary human placental tissues and vascular cells.
Collapse
Affiliation(s)
- Fiona Brownfoot
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia.
| | - Natalie Binder
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Roxanne Hastie
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Alesia Harper
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Sally Beard
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Laura Tuohey
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Emerson Keenan
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Stephen Tong
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| | - Natalie Hannan
- Mercy Perinatal, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia; Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, University of Melbourne. Mercy Perinatal, Mercy Hospital for Women 163 Studley Rd, Heidelberg, 3084, Victoria, Australia
| |
Collapse
|
3
|
Liu S, Yu J, Fu M, Wang X, Chang X. Regulatory effects of hawthorn polyphenols on hyperglycemic, inflammatory, insulin resistance responses, and alleviation of aortic injury in type 2 diabetic rats. Food Res Int 2021; 142:110239. [PMID: 33773689 DOI: 10.1016/j.foodres.2021.110239] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/26/2021] [Accepted: 02/11/2021] [Indexed: 12/24/2022]
Abstract
Hawthorn polyphenol extract (HPE) is beneficial for patients with type 2 diabetes (T2D). However, the mechanism underlying its beneficial effects remains unclear. We investigated the inhibitory effects and mechanisms of HPE on insulin resistance, inflammation, and aortic injury in T2D rats, using metformin (MF) as a positive control. High-performance liquid chromatography-electrospray ionization-tandem mass spectrometry (HPLC-ESI-MS/MS) was used to determine the primary polyphenols in HPE. Hematoxylin & Eosin (H&E) staining was used to evaluate pathological conditions of the skeletal muscle, liver, and aorta vessels in each group. The levels of serum and intestinal tissue oxidative stress, tumor necrosis factor α (TNF-α), and inflammatory interleukin-6 (IL-6) were also assessed. Western blotting was used to evaluate protein expression levels in the associated molecular pathway. Volatile organic compounds (VOCs) from colon contents were determined using headspace-gas chromatography-ion mobility chromatography. Our results showed that supplementation with 300 mg HPE/kg body weight over four weeks significantly improved total cholesterol (TC), total triglyceride (TG), insulin, and lipopolysaccharide (LPS) levels in diabetic rats (p < 0.01). The lesions of skeletal muscle, liver, and aorta in diabetic rats were significantly improved. HPE supplementation also significantly downregulated the inflammatory factors (IL-6, TNF-α, and MCP-1) in the liver of diabetic rats via the SIRT1/AMPK/NF-κB signaling pathway. Furthermore, HPE significantly reduced insulin resistance in T2D rats by upregulating the phosphorylation of glucose absorption protein (GLUT4) and insulin resistance-associated proteins, p-IRS1, p-AKT, and p-PI3K, in the rat liver (p < 0.01). The findings show that HPE could also alleviate aortic injury by activating SIRT1 and regulating the NF-κB and Wnt2/β-catenin signaling pathways. Overall, the results of this study suggest that both HPE and MF have similar inhibitory effects on T2D in rats and that HPE could be used as a functional food component in the adjuvant treatment of T2D.
Collapse
Affiliation(s)
- Suwen Liu
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China.
| | - Jincheng Yu
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China
| | - Mengfan Fu
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China
| | - Xinfang Wang
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China
| | - Xuedong Chang
- College of Food Science & Technology, Hebei Normal University of Science and Technology, Qinhuangdao, Hebei 066004, China; Hebei (Chengde) Hawthorn Industrial Technology Research Institute, Chengde, Hebei 067000, China
| |
Collapse
|
4
|
He Z, Wang Y, He Q, Chen M. microRNA-491-5p protects against atherosclerosis by targeting matrix metallopeptidase-9. Open Med (Wars) 2020; 15:492-500. [PMID: 33313408 PMCID: PMC7706122 DOI: 10.1515/med-2020-0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/30/2020] [Accepted: 02/04/2020] [Indexed: 11/15/2022] Open
Abstract
Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are critical processes that are involved in atherosclerosis. The aim of this study was to explore the role of microRNA-491-5p (miR-491-5p) in the progression of atherosclerosis by regulating the growth and migration of VSMCs. In this study, we showed that the expression of miR-491-5p was downregulated in the atherosclerotic plaque tissues and plasma samples of the patients with atherosclerosis. The bioinformatic analysis and dual-luciferase reporter assay identified that matrix metallopeptidase-9 (MMP-9) was a target gene of miR-491-5p. The results showed a significant upregulation of MMP-9 in the atherosclerotic plaque tissues and plasma samples. Subsequently, the results also showed that downregulation of miR-491-5p significantly promoted the proliferation and migration of VSMCs and inhibited the apoptosis in VSMCs. Furthermore, we detected the effects of miR-491-5p mimic on the growth and migration of VSMCs, and the results illustrated that miR-491-5p mimic could inhibit the proliferation and migration of VSMCs and promote the apoptosis of VSMCs. Notably, MMP-9 plasmid could reverse all the effects of miR-491-5p mimic on VSMCs. Collectively, our study provides the first evidence that miR-491-5p inhibited the growth and migration of VSMCs by targeting MMP-9, which might provide new biomarkers and potential therapeutic targets for atherosclerosis treatment.
Collapse
Affiliation(s)
- Zhonghan He
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yayun Wang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qin He
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Manhua Chen
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang'an District, Wuhan 430030, Hubei Province, China
| |
Collapse
|
5
|
Wang F, Chen HZ. Histone Deacetylase SIRT1, Smooth Muscle Cell Function, and Vascular Diseases. Front Pharmacol 2020; 11:537519. [PMID: 33117155 PMCID: PMC7573826 DOI: 10.3389/fphar.2020.537519] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 09/18/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs), located in the media of artery, play key roles in maintaining the normal vascular physiological functions. Abnormality in VSMCs is implicated in vascular diseases (VDs), including atherosclerosis, abdominal aortic aneurysm (AAA), aortic dissection, and hypertension by regulating the process of inflammation, phenotypic switching, and extracellular matrix degradation. Sirtuins (SIRTs), a family of proteins containing seven members (from SIRT1 to SIRT7) in mammals, function as NAD+-dependent histone deacetylases and ADP-ribosyltransferases. In recent decades, great attention has been paid to the cardiovascular protective effects of SIRTs, especially SIRT1, suggesting a new therapeutic target for the treatment of VDs. In this review, we introduce the basic functions of SIRT1 against VSMC senescence, and summarize the contribution of SIRT1 derived from VSMCs in VDs. Finally, the potential new strategies based on SIRT1 activation have also been discussed with an emphasis on SIRT1 activators and calorie restriction to improve the prognosis of VDs.
Collapse
Affiliation(s)
- Fang Wang
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Hou-Zao Chen
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Liu G, Baird AW, Parsons MJ, Fan K, Skerrett-Byrne DA, Nair PM, Makanyengo S, Chen J, Neal R, Goggins BJ, Tay H, Mathe A, Soh WS, Minahan K, Hansbro PM, Nixon B, McCaughan GW, Holtmann G, Colgan SP, Keely S. Platelet activating factor receptor acts to limit colitis-induced liver inflammation. FASEB J 2020; 34:7718-7732. [PMID: 32293760 DOI: 10.1096/fj.201901779r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/31/2022]
Abstract
Liver inflammation is a common extraintestinal manifestation in inflammatory bowel disease (IBD), yet, the mechanisms driving gut-liver axis inflammation remain poorly understood. IBD leads to a breakdown in the integrity of the intestinal barrier causing an increase in portal and systemic gut-derived antigens, which challenge the liver. Here, we examined the role of platelet activating factor receptor (PAFR) in colitis-associated liver damage using dextran sulfate sodium (DSS) and anti-CD40-induced colitis models. Both DSS and anti-CD40 models exhibited liver inflammation associated with colitis. Colitis reduced global PAFR protein expression in mouse livers causing an exclusive re-localization of PAFR to the portal triad. The global decrease in liver PAFR was associated with increased sirtuin 1 while relocalized PAFR expression was limited to Kupffer cells (KCs) and co-localized with toll-like receptor 4. DSS activated the NLRP3-inflammasome and increased interleukin (IL)-1β in the liver. Antagonism of PAFR amplified the inflammasome response by increasing NLRP3, caspase-1, and IL-1β protein levels in the liver. LPS also increased NLRP3 response in human hepatocytes, however, overexpression of PAFR restored the levels of NLPR3 and caspase-1 proteins. Interestingly, KCs depletion also increased IL-1β protein in mouse liver after DSS challenge. These data suggest a protective role for PAFR-expressing KCs during colitis and that regulation of PAFR is important for gut-liver axis homeostasis.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Alan W Baird
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Marie J Parsons
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kening Fan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - David A Skerrett-Byrne
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Prema M Nair
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Samwel Makanyengo
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Rachel Neal
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Bridie J Goggins
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Hock Tay
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Andrea Mathe
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Wai S Soh
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kyra Minahan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Phil M Hansbro
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Brett Nixon
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Geoffrey W McCaughan
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Gerald Holtmann
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Sean P Colgan
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Keely
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
7
|
Xu S, Kamato D, Little PJ, Nakagawa S, Pelisek J, Jin ZG. Targeting epigenetics and non-coding RNAs in atherosclerosis: from mechanisms to therapeutics. Pharmacol Ther 2019; 196:15-43. [PMID: 30439455 PMCID: PMC6450782 DOI: 10.1016/j.pharmthera.2018.11.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, the principal cause of cardiovascular death worldwide, is a pathological disease characterized by fibro-proliferation, chronic inflammation, lipid accumulation, and immune disorder in the vessel wall. As the atheromatous plaques develop into advanced stage, the vulnerable plaques are prone to rupture, which causes acute cardiovascular events, including ischemic stroke and myocardial infarction. Emerging evidence has suggested that atherosclerosis is also an epigenetic disease with the interplay of multiple epigenetic mechanisms. The epigenetic basis of atherosclerosis has transformed our knowledge of epigenetics from an important biological phenomenon to a burgeoning field in cardiovascular research. Here, we provide a systematic and up-to-date overview of the current knowledge of three distinct but interrelated epigenetic processes (including DNA methylation, histone methylation/acetylation, and non-coding RNAs), in atherosclerotic plaque development and instability. Mechanistic and conceptual advances in understanding the biological roles of various epigenetic modifiers in regulating gene expression and functions of endothelial cells (vascular homeostasis, leukocyte adhesion, endothelial-mesenchymal transition, angiogenesis, and mechanotransduction), smooth muscle cells (proliferation, migration, inflammation, hypertrophy, and phenotypic switch), and macrophages (differentiation, inflammation, foam cell formation, and polarization) are discussed. The inherently dynamic nature and reversibility of epigenetic regulation, enables the possibility of epigenetic therapy by targeting epigenetic "writers", "readers", and "erasers". Several Food Drug Administration-approved small-molecule epigenetic drugs show promise in pre-clinical studies for the treatment of atherosclerosis. Finally, we discuss potential therapeutic implications and challenges for future research involving cardiovascular epigenetics, with an aim to provide a translational perspective for identifying novel biomarkers of atherosclerosis, and transforming precision cardiovascular research and disease therapy in modern era of epigenetics.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Wooloongabba, QLD 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou 510520, China
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita 12-jo Nishi 6-chome, Kita-ku, Sapporo 060-0812, Japan
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar der Technischen Universitaet Muenchen, Germany
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| |
Collapse
|
8
|
Zhang S, Yang Y, Huang S, Deng C, Zhou S, Yang J, Cao Y, Xu L, Yuan Y, Yang J, Chen G, Zhou L, Lv Y, Wang L, Zou X. SIRT1 inhibits gastric cancer proliferation and metastasis via STAT3/MMP‐13 signaling. J Cell Physiol 2019; 234:15395-15406. [PMID: 30710340 DOI: 10.1002/jcp.28186] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Shu Zhang
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Yang Yang
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Shuling Huang
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Chao Deng
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Siqi Zhou
- Department of Gastroenterology Nanjing Medical University Affiliated Drum Tower Clinical Medical College Nanjing China
| | - Jie Yang
- Department of Gastroenterology Nanjing Medical University Affiliated Drum Tower Clinical Medical College Nanjing China
| | - Yu Cao
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Lei Xu
- Department of Gastroenterology Nanjing Medical University Affiliated Drum Tower Clinical Medical College Nanjing China
| | - Yue Yuan
- Department of Gastroenterology Nanjing Medical University Affiliated Drum Tower Clinical Medical College Nanjing China
| | - Jun Yang
- Department of Pathology Drum Tower Hospital Affiliated Medical School of Nanjing University Nanjing China
| | - Guangxia Chen
- Department of Gastroenterology First People's Hospital of Xuzhou Xuzhou China
| | - Lin Zhou
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Ying Lv
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Lei Wang
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| | - Xiaoping Zou
- Department of Gastroenterology Nanjing University Medical School Affiliated Drum Tower Hospital Nanjing China
- Jiangsu Clinical Medical Center of Digestive Disease Nanjing China
| |
Collapse
|
9
|
Kim JS, Pak K, Goh TS, Jeong DC, Han ME, Kim J, Oh SO, Kim CD, Kim YH. Prognostic Value of MicroRNAs in Coronary Artery Diseases: A Meta-Analysis. Yonsei Med J 2018; 59:495-500. [PMID: 29749132 PMCID: PMC5949291 DOI: 10.3349/ymj.2018.59.4.495] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/12/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Coronary artery diseases (CADs) are the leading causes of death in the world. Recent studies have reported that differentially expressed microRNAs (miRNAs) are associated with prognosis or major adverse cardiac events (MACEs) in CAD patients. In a previous meta-analysis, the authors made serious mistakes that we aimed to correct through an updated systematic review and meta-analysis of the prognostic value of altered miRNAs in patients with CADs. MATERIALS AND METHODS We performed a systematic search of MEDLINE (from inception to May 2017) and EMBASE (from inception to May 2017) for English-language publications. Studies of CADs with results on miRNAs that reported survival data or MACEs were included. Data were extracted from each publication independently by two reviewers. RESULTS After reviewing 515 articles, a total eight studies were included in this study. We measured pooled hazard ratios (HRs) and 95% confidence intervals (CIs) of miRNA 133a with a fixed-effect model (pooled HR, 2.35; 95% CI, 1.56-3.55). High expression of miRNA 133a, 208b, 126, 197, 223, and 122-5p were associated with high mortality. Additionally, high levels of miRNA 208b, 499-5p, 134, 328, and 34a were related with MACEs. CONCLUSION The present study confirmed that miRNA 133a, which was associated with high mortality in CAD patients, holds prognostic value in CAD. More importantly, this study corrected issues raised against a prior meta-analysis and provides accurate information.
Collapse
Affiliation(s)
- Ji Suk Kim
- BEER, Busan Society of Evidence-based Mdicine and Research, Busan, Korea
- Department of Family Medicine, BHS Han Seo Hospital, Busan, Korea
| | - Kyoungjune Pak
- BEER, Busan Society of Evidence-based Mdicine and Research, Busan, Korea
- Department of Nuclear Medicine, Pusan National University Hospital, Busan, Korea
| | - Tae Sik Goh
- BEER, Busan Society of Evidence-based Mdicine and Research, Busan, Korea
- Department of Orthopaedic Surgery, Pusan National University Hospital, Busan, Korea
| | | | - Myoung Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - Jihyun Kim
- Department of Family Medicine, BHS Han Seo Hospital, Busan, Korea
| | - Sae Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yun Hak Kim
- BEER, Busan Society of Evidence-based Mdicine and Research, Busan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea.
| |
Collapse
|
10
|
Desimine VL, McCrink KA, Parker BM, Wertz SL, Maning J, Lymperopoulos A. Biased Agonism/Antagonism of Cardiovascular GPCRs for Heart Failure Therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:41-61. [PMID: 29776604 DOI: 10.1016/bs.ircmb.2018.02.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
G protein-coupled receptors (GPCRs) are among the most important drug targets currently used in clinic, including drugs for cardiovascular indications. We now know that, in addition to activating heterotrimeric G protein-dependent signaling pathways, GPCRs can also activate G protein-independent signaling, mainly via the βarrestins. The major role of βarrestin1 and -2, also known as arrestin2 or -3, respectively, is to desensitize GPCRs, i.e., uncoupled them from G proteins, and to subsequently internalize the receptor. As the βarrestin-bound GPCR recycles inside the cell, it serves as a signalosome transducing signals in the cytoplasm. Since both G proteins and βarrestins can transduce signals from the same receptor independently of each other, any given GPCR agonist might selectively activate either pathway, which would make it a biased agonist for that receptor. Although this selectivity is always relative (never absolute), in cases where the G protein- and βarrestin-dependent signals emanating from the same GPCR result in different cellular effects, pharmacological exploitation of GPCR-biased agonism might have therapeutic potential. In this chapter, we summarize the GPCR signaling pathways and their biased agonism/antagonism examples discovered so far that can be exploited for heart failure treatment. We also highlight important issues that need to be clarified along the journey of these ligands from bench to the clinic.
Collapse
Affiliation(s)
- Victoria L Desimine
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Katie A McCrink
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Barbara M Parker
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Shelby L Wertz
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Jennifer Maning
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States
| | - Anastasios Lymperopoulos
- From the Laboratory for the Study of Neurohormonal Control of the Circulation, Nova Southeastern University College of Pharmacy, Fort Lauderdale, FL, United States.
| |
Collapse
|
11
|
Hwang ES, Song SB. Nicotinamide is an inhibitor of SIRT1 in vitro, but can be a stimulator in cells. Cell Mol Life Sci 2017; 74:3347-3362. [PMID: 28417163 PMCID: PMC11107671 DOI: 10.1007/s00018-017-2527-8] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 03/24/2017] [Accepted: 04/12/2017] [Indexed: 01/15/2023]
Abstract
Nicotinamide (NAM), a form of vitamin B3, plays essential roles in cell physiology through facilitating NAD+ redox homeostasis and providing NAD+ as a substrate to a class of enzymes that catalyze non-redox reactions. These non-redox enzymes include the sirtuin family proteins which deacetylate target proteins while cleaving NAD+ to yield NAM. Since the finding that NAM exerts feedback inhibition to the sirtuin reactions, NAM has been widely used as an inhibitor in the studies where SIRT1, a key member of sirtuins, may have a role in certain cell physiology. However, once administered to cells, NAM is rapidly converted to NAD+ and, therefore, the cellular concentration of NAM decreases rapidly while that of NAD+ increases. The result would be an inhibition of SIRT1 for a limited duration, followed by an increase in the activity. This possibility raises a concern on the validity of the interpretation of the results in the studies that use NAM as a SIRT1 inhibitor. To understand better the effects of cellular administration of NAM, we reviewed published literature in which treatment with NAM was used to inhibit SIRT1 and found that the expected inhibitory effect of NAM was either unreliable or muted in many cases. In addition, studies demonstrated NAM administration stimulates SIRT1 activity and improves the functions of cells and organs. To determine if NAM administration can generate conditions in cells and tissues that are stimulatory to SIRT1, the changes in the cellular levels of NAM and NAD+ reported in the literature were examined and the factors that are involved in the availability of NAD+ to SIRT1 were evaluated. We conclude that NAM treatment can hypothetically be stimulatory to SIRT1.
Collapse
Affiliation(s)
- Eun Seong Hwang
- Department of Life Science, University of Seoul, Dongdaemungu, 163 Seoulsiripdaero, Seoul, 02504, Republic of Korea.
| | - Seon Beom Song
- Department of Life Science, University of Seoul, Dongdaemungu, 163 Seoulsiripdaero, Seoul, 02504, Republic of Korea
| |
Collapse
|
12
|
Yang X, Wei J, He Y, Jing T, Li Y, Xiao Y, Wang B, Wang W, Zhang J, Lin R. SIRT1 inhibition promotes atherosclerosis through impaired autophagy. Oncotarget 2017; 8:51447-51461. [PMID: 28881659 PMCID: PMC5584260 DOI: 10.18632/oncotarget.17691] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/24/2017] [Indexed: 11/25/2022] Open
Abstract
SIRT1, a highly conserved NAD+-dependent protein deacetylase, plays a pivotal role in the pathogenesis and therapy of atherosclerosis (AS). The aim of this study is to investigate the potential effects of SIRT1 on AS in ApoE-/- mice and the underlying mechanisms of autophagy in an ox-LDL-stimulated human monocyte cell line, THP-1. In vivo, the accelerated atherosclerotic progression of mice was established by carotid collar placement; then, mice were treated for 4 weeks with a SIRT1-specific inhibitor, EX-527. The atherosclerotic lesion size of EX-527-treated mice was greatly increased compared to that of the mice in the control group. Immunostaining protocols confirmed that the inhibition of SIRT1 during plaque initiation and progression enhanced the extent of intraplaque macrophage infiltration and impaired the autophagy process. In vitro cultured THP-1 macrophages exposed to ox-LDL were utilized to study the link between the SIRT1 function, autophagy flux, pro-inflammatory cytokine secretion, and foam cell formation using different methods. Our data showed that ox-LDL markedly suppressed SIRT1 protein expression and the autophagy level, while it elevated the MCP-1 production and lipid uptake. Additionally, the application of the SIRT1 inhibitor EX-527 or SIRT1 siRNA further attenuated ox-LDL-induced autophagy inhibition. In conclusion, our results show that the inhibition of SIRT1 promoted atherosclerotic plaque development in ApoE-/- mice by increasing the MCP-1 expression and macrophage accumulation. In particular, we demonstrate that blocking SIRT1 can exacerbate the acetylation of key autophagy machinery, the Atg5 protein, which further regulates the THP-1 macrophage-derived foam cell formation that is triggered by ox-LDL.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| | - Jingyuan Wei
- Liaoning Province Academy of Analytic Science, Shenyang 110015, Liaoning, P. R. China
| | - Yanhao He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| | - Ting Jing
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| | - Yanxiang Li
- Taizhou Polytechnic College, Taizhou 225300, Jiangsu, P. R. China
| | - Yunfang Xiao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| | - Bo Wang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P. R. China
| | - Jiye Zhang
- School of Pharmacology, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P. R. China
| | - Rong Lin
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, P. R. China
| |
Collapse
|
13
|
Kim YH, Bae JU, Kim IS, Chang CL, Oh SO, Kim CD. SIRT1 prevents pulmonary thrombus formation induced by arachidonic acid via downregulation of PAF receptor expression in platelets. Platelets 2016; 27:735-742. [DOI: 10.1080/09537104.2016.1190005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yun Hak Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jin Ung Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - In Suk Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine and Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, Republic of Korea
| | - Chulhun L. Chang
- Department of Laboratory Medicine, Pusan National University School of Medicine and Pusan National University Yangsan Hospital, Yangsan, Gyeongnam, Republic of Korea
| | - Sae Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| |
Collapse
|
14
|
|
15
|
Zhang MJ, Zhou Y, Chen L, Wang X, Long CY, Pi Y, Gao CY, Li JC, Zhang LL. SIRT1 improves VSMC functions in atherosclerosis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 121:11-5. [PMID: 27080738 DOI: 10.1016/j.pbiomolbio.2016.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/17/2016] [Indexed: 12/19/2022]
Abstract
Despite advancements in diagnosis and treatment of cardiovascular diseases (CVDs), the morbidity and mortality of CVDs are still rising. Atherosclerosis is a chronic inflammatory disease contributing to multiple CVDs. Considering the complexity and severity of atherosclerosis, it is apparent that exploring the mechanisms of atherosclerotic formation and seeking new therapies for patients with atherosclerosis are required to overcome the heavy burden of CVDs on the quality and length of life of the global population. Vascular smooth muscle cells (VSMCs) play a dominant role in functional and structural changes of the arterial walls in response to atherogenic factors. Therefore, improvement of VSMC functions will slow down the development of atherosclerosis to a large extent. Given its protective performances on regulation of cholesterol metabolism and inflammatory responses, SIRT1 has long been known as an anti-atherosclerosis factor. In this review, we focus on the effects of SIRT1 on VSMC functions and thereby the development of atherosclerosis.
Collapse
Affiliation(s)
- Ming-Jie Zhang
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Yi Zhou
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Lei Chen
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Xu Wang
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Chun-Yan Long
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Yan Pi
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Chang-Yue Gao
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Jing-Cheng Li
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China
| | - Li-Li Zhang
- Department of Neurology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, 10 Changjiang Branch Road, Yuzhong District, Chongqing 400042, PR China.
| |
Collapse
|