1
|
Sun Y, Zhu D, Kong L, Du W, Qu L, Yang Y, Rao G, Huang F, Tong X. Vasicine attenuates atherosclerosis via lipid regulation, inflammation inhibition, and autophagy activation in ApoE -/- mice. Int Immunopharmacol 2024; 142:112996. [PMID: 39243558 DOI: 10.1016/j.intimp.2024.112996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/18/2024] [Accepted: 08/19/2024] [Indexed: 09/09/2024]
Abstract
Atherosclerosis is marked with the accumulation of low-density lipoproteins and chronic inflammation. The anti-inflammatory therapies exert protective effects on atherosclerosis. Vasicine is a bioactive alkaloid with anti-inflammatory activity from a medicinal plant in Ayurveda and Unani. In this study, the effects of vasicine were evaluated on atherosclerosis in vivo and in vitro. The results showed that vasicine alleviated atherosclerotic lesions and regulated the lipid synthesis by reducing the levels of TC, TG, LDL-C and inhibiting the expresses of scavenger receptors (SR-A, CD36 and LOX-1) to inhibit foam cell formations. And vasicine decreased the levels of IL-1β, IL-6, MCP-1, and TNF-α to modulate inflammatory response. Besides, vasicine downregulated MAPK and PI3K/AKT/mTOR pathway to activated autophagy, which inhibited the procession of atherosclerosis.
Collapse
Affiliation(s)
- Yun Sun
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Defen Zhu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Lingqi Kong
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Wenxia Du
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Lu Qu
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Yingfei Yang
- Zhaotong Hospital of Chinese Traditional Medicine, Zhaotong 657000, People's Republic of China
| | - Gaoxiong Rao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China
| | - Feng Huang
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China; School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China.
| | - Xiaoyun Tong
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming 650500, People's Republic of China; The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650021, People's Republic of China.
| |
Collapse
|
2
|
Mu J, Lin Q, Chen Y, Wang J, Yu X, Huang F, Liu X, Fang Y, Li Y, Zhu B, Liang Y. Rice bran active peptide (RBAP) inhibited macrophage differentiation to foam cell and atherosclerosis in mice via regulating cholesterol efflux. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155864. [PMID: 39032281 DOI: 10.1016/j.phymed.2024.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Atherosclerosis is a long-lasting inflammatory condition affecting the walls of arteries, marked by the buildup of fats, plaque formation, and vascular remodeling. Recent findings highlight the significance of cholesterol removal pathways in influencing atherosclerosis, yet the connection between cholesterol removal and regulation of macrophage inflammation remains poorly understood. RBAP could serve as an anti-inflammatory agent; however, its role in atherosclerosis and the mechanism behind it are still not well understood. PURPOSE The objective of this research is to explore how RBAP impacts cholesterol efflux, which is a considerable element in the advancement of atherosclerosis. METHODS An atherosclerosis mouse model was established by using an ApoE KO strain mouse on a high-fat diet (HFD) to assess the effects of RBAP, conducted either orally or through injection. Additionally, in vitro experiments were conducted where the induction of THP-1 cells was conducted for the differentiation towards macrophages, and along with mouse RAW264.7 cells, were challenged with ox-LDL to evaluate the impact of RBAP. RESULTS In this study, RBAP was found to reduce the production and downregulate TNF-α, IL-1β, and IL-6 levels and inhibited the activation of the TLR4/MyD88/NF-κB signaling in atherosclerosis model mice, as well as in ox-LDL-challenged THP-1 cells and mouse RAW264.7 macrophages. RBAP's effectiveness also improved the enhancement of reverse cholesterol transport (RCT) and cholesterol removal to HDL and apoA1 by increasing the activity of genes related to cholesterol removal PPARγ/LXRα/ABCA1/ABCG1, both in ApoE-/- mice and in THP-1 cells and mouse RAW264.7 macrophages. Notably, RBAP exerted similar effects on atherosclerosis model mice and macrophages to those of TAK-242, an inhibitor of the TLR4 signaling. When RBAP and TAK-242 were applied simultaneously, the improvement was not enhanced compared with either RBAP or TAK-242 treatment alone. CONCLUSION These findings suggest that RBAP, as a TLR4 inhibitor, has anti-atherosclerotic effects by improving inflammation and promoting cholesterol effection, indicating its therapeutic potential in intervening atherosclerosis.
Collapse
Affiliation(s)
- Jianfei Mu
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Qinlu Lin
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Yajuan Chen
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Jianqiang Wang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Xudong Yu
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Fang Huang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Xinxin Liu
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China
| | - Yong Fang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, Jiangsu 210023, PR China
| | - Yusheng Li
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Beiwei Zhu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, PR China.
| | - Ying Liang
- Molecular Nutrition Branch, National Engineering Research Center of Rice and By-product Deep Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha 410004, Hunan, PR China.
| |
Collapse
|
3
|
Grazide MH, Ruidavets JB, Martinet W, Elbaz M, Vindis C. Association of Circulating Autophagy Proteins ATG5 and Beclin 1 with Acute Myocardial Infarction in a Case-Control Study. Cardiology 2024; 149:217-224. [PMID: 38432214 PMCID: PMC11152019 DOI: 10.1159/000537816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/09/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) is a main contributor of sudden cardiac death worldwide. The discovery of new biomarkers that can improve AMI risk prediction meets a major clinical need for the identification of high-risk patients and the tailoring of medical treatment. Previously, we reported that autophagy a highly conserved catabolic mechanism for intracellular degradation of cellular components is involved in atherosclerotic plaque phenotype and cardiac pathological remodeling. The crucial role of autophagy in the normal and diseased heart has been well described, and its activation functions as a pro-survival process in response to myocardial ischemia. However, autophagy is dysregulated in ischemia/reperfusion injury, thus promoting necrotic or apoptotic cardiac cell death. Very few studies have focused on the plasma levels of autophagy markers in cardiovascular disease patients, even though they could be companion biomarkers of AMI injury. The aims of the present study were to evaluate (1) whether variations in plasma levels of two key autophagy regulators autophagy-related gene 5 (ATG5) and Beclin 1 (the mammalian yeast ortholog Atg6/Vps30) are associated with AMI and (2) their potential for predicting AMI risk. METHODS The case-control study population included AMI patients (n = 100) and control subjects (n = 99) at high cardiovascular risk but without known coronary disease. Plasma levels of ATG5 and Beclin 1 were measured in the whole population study by enzyme-linked immunosorbent assay. RESULTS Multivariate analyses adjusted on common cardiovascular factors and medical treatments, and receiver operating characteristic curves demonstrated that ATG5 and Beclin 1 levels were inversely associated with AMI and provided original biomarkers for AMI risk prediction. CONCLUSION Plasma levels of autophagy regulators ATG5 and Beclin 1 represent relevant candidate biomarkers associated with AMI.
Collapse
Affiliation(s)
- Marie-Hélène Grazide
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Meyer Elbaz
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
| | - Cécile Vindis
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
| |
Collapse
|
4
|
Marzoog BA. Autophagy Behavior in Endothelial Cell Regeneration. Curr Aging Sci 2024; 17:58-67. [PMID: 37861048 DOI: 10.2174/0118746098260689231002044435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023]
Abstract
Autophagy plays a crucial role in maintaining endothelial cell homeostasis through the turnover of intracellular components during stress conditions in a lysosomal-dependent manner. The regeneration strategy involves several aspects, including autophagy. Autophagy is a catabolic degenerative lysosomal-dependent degradation of intracellular components. Autophagy modifies cellular and subcellular endothelial cell functions, including mitochondria stress, lysosomal stress, and endoplasmic reticulum unfolded protein response. Activation of common signaling pathways of autophagy and regeneration and enhancement of intracellular endothelial cell metabolism serve as the bases for the induction of endothelial regeneration. Endothelial progenitor cells include induced pluripotent stem cells (iPSC), embryonic stem cells, and somatic cells, such as fibroblasts. Future strategies of endothelial cell regeneration involve the induction of autophagy to minimize the metabolic degeneration of the endothelial cells and optimize the regeneration outcomes.
Collapse
Affiliation(s)
- Basheer Abdullah Marzoog
- World-Class Research Center «Digital Biodesign and Personalized Healthcare», I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| |
Collapse
|
5
|
Wang S, Yuan R, Liu M, Zhang Y, Jia B, Ruan J, Shen J, Zhang Y, Liu M, Wang T. Targeting autophagy in atherosclerosis: Advances and therapeutic potential of natural bioactive compounds from herbal medicines and natural products. Biomed Pharmacother 2022; 155:113712. [PMID: 36130420 DOI: 10.1016/j.biopha.2022.113712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis (AS) is the most common causes of cardiovascular disease characterized by the formation of atherosclerotic plaques in the arterial wall, and it has become a dominant public health problem that seriously threaten people worldwide. Autophagy is a cellular self-catabolism process, which is critical to protect cellular homeostasis against harmful conditions. Emerging evidence suggest that dysregulated autophagy is involved in the development of AS. Therefore, pharmacological interventions have been developed to inhibit the AS via autophagy induction. Among various AS treating methods, herbal medicines and natural products have been applied as effective complementary and alternative medicines to ameliorate AS and its associated cardiovascular disease. Recently, mounting evidence revealed that natural bioactive compounds from herbs and natural products could induce autophagy to suppress the occurrence and development of AS, by promoting cholesterol efflux, reducing plaque inflammation, and inhibiting apoptosis or senescence. In the present review, we highlight recent findings regarding possible effects and molecular mechanism of natural compounds in autophagy-targeted mitigation of atherosclerosis, aiming to provide new potential therapeutic strategies for the atherosclerosis treatment preclinically and clinically.
Collapse
Affiliation(s)
- Sijian Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruolan Yuan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Miao Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yiwen Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bona Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jingya Ruan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiayan Shen
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Mengyang Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
6
|
Lorusso B, Cerasoli G, Falco A, Frati C, Graiani G, Madeddu D, Nogara A, Corradini E, Roti G, Cerretani E, Gherli A, Caputi M, Gnetti L, Pilato FP, Quaini F, Lagrasta C. Β-blockers activate autophagy on infantile hemangioma-derived endothelial cells in vitro. Vascul Pharmacol 2022; 146:107110. [PMID: 36103993 DOI: 10.1016/j.vph.2022.107110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 01/18/2023]
Abstract
The mechanisms underlying the success of propranolol in the treatment of infantile hemangioma (IH) remain elusive and do not fully explain the rapid regression of hemangiomatous lesions following drug administration. As autophagy is critically implicated in vascular homeostasis, we determined whether β-blockers trigger the autophagic flux on infantile hemangioma-derived endothelial cells (Hem-ECs) in vitro. MATERIAL AND METHODS Fresh tissue specimens, surgically removed for therapeutic purpose to seven children affected by proliferative IH, were subjected to enzymatic digestion. Cells were sorted with anti-human CD31 immunolabeled magnetic microbeads. Following phenotypic characterization, expanded Hem-ECs, at P2 to P6, were exposed to different concentrations (50 μM to 150 μM) of propranolol, atenolol or metoprolol alone and in combination with the autophagy inhibitor Bafilomycin A1. Rapamycin, a potent inducer of autophagy, was also used as control. Autophagy was assessed by Lysotracker Red staining, western blot analysis of LC3BII/LC3BI and p62, and morphologically by transmission electron microscopy. RESULTS Hem-ECs treated with either propranolol, atenolol or metoprolol displayed positive LysoTracker Red staining. Increased LC3BII/LC3BI ratio, as well as p62 modulation, were documented in β-blockers treated Hem-ECs. Abundant autophagic vacuoles and multilamellar bodies characterized the cytoplasmic ultrastructural features of autophagy in cultured Hem-ECs exposed in vitro to β-blocking agents. Importantly, similar biochemical and morphologic evidence of autophagy were observed following rapamycin while Bafilomycin A1 significantly prevented the autophagic flux promoted by β-blockers in Hem-ECs. CONCLUSION Our data suggest that autophagy may be ascribed among the mechanisms of action of β-blockers suggesting new mechanistic insights on the potential therapeutic application of this class of drugs in pathologic conditions involving uncontrolled angiogenesis.
Collapse
Affiliation(s)
- Bruno Lorusso
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Cerasoli
- Pediatric Surgery, Ospedale dei Bambini of Parma, University Hospital of Parma, Parma, Italy
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gallia Graiani
- Dental School, University of Parma Medical School, Parma, Italy
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonella Nogara
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elisa Cerretani
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Andrea Gherli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Letizia Gnetti
- Pathology Section, University Hospital of Parma, Parma, Italy
| | | | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Costanza Lagrasta
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
7
|
Tripathi M, Singh BK, Liehn EA, Lim SY, Tikno K, Castano-Mayan D, Rattanasopa C, Nilcham P, Abdul Ghani SAB, Wu Z, Azhar SH, Zhou J, Hernández-Resèndiz S, Crespo-Avilan GE, Sinha RA, Farah BL, Moe KT, De Silva DA, Angeli V, Singh MK, Singaraja RR, Hausenloy DJ, Yen PM. Caffeine prevents restenosis and inhibits vascular smooth muscle cell proliferation through the induction of autophagy. Autophagy 2022; 18:2150-2160. [PMID: 35012409 PMCID: PMC9466618 DOI: 10.1080/15548627.2021.2021494] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Caffeine is among the most highly consumed substances worldwide, and it has been associated with decreased cardiovascular risk. Although caffeine has been shown to inhibit the proliferation of vascular smooth muscle cells (VSMCs), the mechanism underlying this effect is unknown. Here, we demonstrated that caffeine decreased VSMC proliferation and induced macroautophagy/autophagy in an in vivo vascular injury model of restenosis. Furthermore, we studied the effects of caffeine in primary human and mouse aortic VSMCs and immortalized mouse aortic VSMCs. Caffeine decreased cell proliferation, and induced autophagy flux via inhibition of MTOR signaling in these cells. Genetic deletion of the key autophagy gene Atg5, and the Sqstm1/p62 gene encoding a receptor protein, showed that the anti-proliferative effect by caffeine was dependent upon autophagy. Interestingly, caffeine also decreased WNT-signaling and the expression of two WNT target genes, Axin2 and Ccnd1 (cyclin D1). This effect was mediated by autophagic degradation of a key member of the WNT signaling cascade, DVL2, by caffeine to decrease WNT signaling and cell proliferation. SQSTM1/p62, MAP1LC3B-II and DVL2 were also shown to interact with each other, and the overexpression of DVL2 counteracted the inhibition of cell proliferation by caffeine. Taken together, our in vivo and in vitro findings demonstrated that caffeine reduced VSMC proliferation by inhibiting WNT signaling via stimulation of autophagy, thus reducing the vascular restenosis. Our findings suggest that caffeine and other autophagy-inducing drugs may represent novel cardiovascular therapeutic tools to protect against restenosis after angioplasty and/or stent placement.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,Contact Madhulika Tripathi Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore169857
| | - Brijesh Kumar Singh
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - Elisa A. Liehn
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-,Insitute for Molecular Medicine, University of Southern Denmark, Odense, J.B. Winsløws Vej 25, 5230, Odense, Denmark,Department for Cardiology, Angiology and Intensive Care, Aachen, Germany
| | - Sheau Yng Lim
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Keziah Tikno
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - David Castano-Mayan
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chutima Rattanasopa
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Pakhwan Nilcham
- Department for Cardiology, Angiology and Intensive Care, Aachen, Germany
| | | | - Zihao Wu
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Syaza Hazwany Azhar
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore
| | - Sauri Hernández-Resèndiz
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Gustavo E. Crespo-Avilan
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Rohit Anthony Sinha
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Uttar Pradesh, India
| | - Benjamin Livingston Farah
- Department of Anatomical Pathology, Division of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Kyaw Thu Moe
- Newcastle University Medicine Malaysia, Newcastle University, 79200 Gelang Patah, Johor,Malaysia
| | - Deidre Anne De Silva
- Department of Neurology, National Neuroscience Institute, Department of Neurology, Singapore General Hospital, Outram Road, Singapore, 169608
| | - Veronique Angeli
- Immunology Translational Research Program, Department of Microbiology & Immunology, Immunology Programme, Life Sciences Institute, Singapore- 117456
| | - Manvendra K. Singh
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-
| | - Roshni R. Singaraja
- Translational Laboratories in Genetic Medicine, A*star Institute, and Yong Loo Lin School of Medicine, National University of Singapore, Singapore,Yong Loo Lin School of Medicine, National University, Singapore-117597
| | - Derek J. Hausenloy
- National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore-,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, 7 Chenies Mews, Bloomsbury, London WC1E 6HX, United Kingdom,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, 500 Liufeng Road, Wufeng District, Taichung City, Taiwan,Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Paul Michael Yen
- Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, 169857, Singapore,Endocrinology, Diabetes, and Metabolism Division, Duke University School of Medicine, Durham, NC, USA,Paul M. Yen Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
| |
Collapse
|
8
|
Kocot AM, Wróblewska B. Nutritional strategies for autophagy activation and health consequences of autophagy impairment. Nutrition 2022; 103-104:111686. [PMID: 35843038 DOI: 10.1016/j.nut.2022.111686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/26/2022]
|
9
|
Ma X, Ji C. Remote Ischemic Conditioning: A Potential Treatment for Chronic Cerebral Hypoperfusion. Eur Neurol 2022; 85:253-259. [PMID: 35104816 DOI: 10.1159/000521803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 12/19/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a clinical syndrome, which is characterized by significantly decreased cerebral blood flow (CBF). CCH is a common consequence of cerebrovascular and cardiovascular diseases and the elderly. CCH results in a series of pathological damages, increasing cell death, autophagy dysfunction, amyloid β (Aβ) peptide accumulation, blood-brain barrier (BBB) disruption, and endothelial damage, which are found in CCH models. In addition, CCH is a prominent risk factor of cognitive impairment, such as vascular dementia, and CCH contributes to the occurrence and development of Alzheimer's disease. Therefore, the treatment of patients with CCH is of great value. It has been confirmed that remote ischemic conditioning (RIC) is a safe, promising treatment for acute and chronic cerebrovascular diseases. RIC significantly increases CBF in both CCH models and patients, inhibits neuronal apoptosis, reduces Aβ deposition, protects BBB integrity and endothelial function, alleviates neuroinflammation, improves cognitive impairment, and exerts neuroprotection. SUMMARY With the development of animal models, the pathophysiological mechanisms of CCH and RIC are increasingly revealed. Key Messages: We discuss the mechanisms related to hypoperfusion in the brain and explore the potential treatment of RIC for CCH to promote its transformation and application in humans.
Collapse
Affiliation(s)
- Xiao Ma
- Department of General Practice Medicine, Dalian Municipal Central Hospital, Dalian, China
| | - Chenhua Ji
- Department of General Practice Medicine, Dalian Municipal Central Hospital, Dalian, China
| |
Collapse
|
10
|
WANG S, SHANG S, LV J, HOU D. Effects of quercetin on renal autophagy and interstitial fibrosis in diabetes mellitus. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.122821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
Zhang Y, He N, Zhou X, Wang F, Cai H, Huang SH, Chen X, Hu Z, Jin X. Betulinic acid induces autophagy-dependent apoptosis via Bmi-1/ROS/AMPK-mTOR-ULK1 axis in human bladder cancer cells. Aging (Albany NY) 2021; 13:21251-21267. [PMID: 34510030 PMCID: PMC8457576 DOI: 10.18632/aging.203441] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/09/2021] [Indexed: 12/22/2022]
Abstract
Betulinic acid (BA), a pentacyclic triterpenoid isolated from tree bark, exhibits antitumor effects against solid malignancies and triggers autophagy and/or apoptosis in human cancer cells. Nonetheless, the relationship between autophagy and apoptosis and the potential modulatory actions of BA on autophagy-dependent bladder cancer cell death remain unclear. The present study showed that BA exposure significantly suppressed viability, proliferation, and migration of EJ and T24 human bladder cancer cells. These effects reflected caspase 3-mediated apoptosis and could be attenuated or abolished by inhibiting ROS production with N-acetyl-L-cysteine, inhibiting autophagy with chloroquine, or silencing ATG7 with targeted siRNA. BA-induced autophagy was evidenced by epifluorescence imaging of lentivirus-induced expression of mCherry-GFP-LC3B and increased expression of two autophagy-related proteins, LC3B-II and TEM. Moreover, enhanced AMPK phosphorylation and decreased mTOR and ULK-1 phosphorylation suggested BA activates autophagy via the AMPK/mTOR/ULK1 pathway. Accordingly, exposure to dorsomorphin (Compound C), an AMPK inhibitor, and AICAR, an AMPK activator, respectively inhibited and stimulated BA-induced autophagy in EJ and T24 cells. The effects of Bmi-1 overexpression in vitro and decreased Bmi-1 expression in BA-treated T24 cell xenografts in nude mice suggested that downregulation of Bmi-1 is the underlying mechanism in BA-mediated, autophagy-dependent apoptosis.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Ning He
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Xuejian Zhou
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Feifan Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Hairong Cai
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Shih Han Huang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Xianwu Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Zhenghui Hu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| | - Xiaodong Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, P.R. China
| |
Collapse
|
12
|
Xu Q, Zhao B, Ye Y, Li Y, Zhang Y, Xiong X, Gu L. Relevant mediators involved in and therapies targeting the inflammatory response induced by activation of the NLRP3 inflammasome in ischemic stroke. J Neuroinflammation 2021; 18:123. [PMID: 34059091 PMCID: PMC8166383 DOI: 10.1186/s12974-021-02137-8] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is a member of the NLR family of inherent immune cell sensors. The NLRP3 inflammasome can detect tissue damage and pathogen invasion through innate immune cell sensor components commonly known as pattern recognition receptors (PRRs). PRRs promote activation of nuclear factor kappa B (NF-κB) pathways and the mitogen-activated protein kinase (MAPK) pathway, thus increasing the transcription of genes encoding proteins related to the NLRP3 inflammasome. The NLRP3 inflammasome is a complex with multiple components, including an NAIP, CIITA, HET-E, and TP1 (NACHT) domain; apoptosis-associated speck-like protein containing a CARD (ASC); and a leucine-rich repeat (LRR) domain. After ischemic stroke, the NLRP3 inflammasome can produce numerous proinflammatory cytokines, mediating nerve cell dysfunction and brain edema and ultimately leading to nerve cell death once activated. Ischemic stroke is a disease with high rates of mortality and disability worldwide and is being observed in increasingly younger populations. To date, there are no clearly effective therapeutic strategies for the clinical treatment of ischemic stroke. Understanding the NLRP3 inflammasome may provide novel ideas and approaches because targeting of upstream and downstream molecules in the NLRP3 pathway shows promise for ischemic stroke therapy. In this manuscript, we summarize the existing evidence regarding the composition and activation of the NLRP3 inflammasome, the molecules involved in inflammatory pathways, and corresponding drugs or molecules that exert effects after cerebral ischemia. This evidence may provide possible targets or new strategies for ischemic stroke therapy.
Collapse
Affiliation(s)
- Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yonggang Zhang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
13
|
Stimulation of AMPK Prevents Diabetes-Induced Photoreceptor Cell Degeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5587340. [PMID: 34093959 PMCID: PMC8140850 DOI: 10.1155/2021/5587340] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/19/2021] [Accepted: 04/01/2021] [Indexed: 12/18/2022]
Abstract
Diabetic retinopathy (DR) is a kind of severe retinal neurodegeneration. The advanced glycation end products (AGEs) affect autophagy, and mitochondrial function is involved in DR. Adenosine-activated protein kinase (AMPK) is an important metabolic sensor that can regulate energy homeostasis in cells. However, the effect of AMPK in DR is still not fully understood. In this study, we investigated the effect of AMPK on diabetes-induced photoreceptor cell degeneration. In vivo, a diabetic mouse model was established by streptozotocin (STZ) injection. Haematoxylin-eosin (HE) staining was used to observe retinal morphology and measure the thicknesses of different layers in the retina. Electroretinogram (ERG) was used to evaluate retinal function. In vitro, 661w cells were treated with AGEs with/without an AMPK agonist (metformin) or AMPK inhibitor (compound C). Flow cytometry and CCK-8 assays were used to analyse apoptosis. Mitochondrial membrane potential was analysed by JC-1. Western blotting and qRT-PCR were used to examine the expression of related proteins and genes, respectively. The wave amplitude and the thickness of the outer nuclear layer were decreased in diabetic mice. The expression of rhodopsin and opsin was also decreased in diabetic mice. In vitro, the percentage of apoptotic cells was increased, the expression of the apoptosis-related protein Bax was increased, and Bcl-2 was decreased after AGE treatment in 661w cells. The expression of the autophagy-related protein LC3 was decreased, and p62 was increased. The mitochondrial-related gene expression and membrane potential were decreased, and mitochondrial morphology was abnormal, as observed by TEM. However, AMPK stimulation ameliorated this effect. These results indicate that AMPK stimulation can delay diabetes-induced photoreceptor degeneration by regulating autophagy and mitochondrial function.
Collapse
|
14
|
Yang Y, Gao H, Liu W, Liu X, Jiang X, Li X, Wu Q, Xu Z, Zhao Q. Arctium lappa L. roots ameliorates cerebral ischemia through inhibiting neuronal apoptosis and suppressing AMPK/mTOR-mediated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153526. [PMID: 33691269 DOI: 10.1016/j.phymed.2021.153526] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Arctium lappa L. roots are very popular cultivated vegetables, which possesses various pharmacological activities. Our previous studies have demonstrated that Arctium lappa L. roots exerted protective effects against H2O2, glutamate and N-methyl-D-aspartic acid (NMDA)-induced neuronal injury in vitro. However, whether Arctium lappa L. roots could prevent against cerebral ischemia and the underlying mechanism remain unclear. PURPOSE The objective of the present study was to investigate the neuroprotective effects of ethyl acetate extract of Arctium lappa L. roots (EAL) and the active ingredient 4,5-O-dicaffeoyl-1-O-[4-malic acid methyl ester]-quinic acid (DCMQA) in EAL against cerebral ischemia and explore the underlying mechanism. STUDY DESIGN The neuroprotective effects of EAL and DCMQA were investigated in rats with permanent middle cerebral artery occlusion (MCAO) and in oxygen glucose deprivation/reoxygenation (OGD/R)-stimulated SH-SY5Y cells, respectively. METHODS The infarct volume, brain edema and neurological deficits were measured following MCAO. TUNEL and Nissl staining were performed to detect neuronal loss and apoptosis of neurons in rat brains. Cell survival was measured by MTT and LDH assay. In addition, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) levels were determined by DCFH-DA and JC-1 fluorescent probe, respectively. Hoechst 33342 staining and Annexin V-FITC/PI double staining were performed to evaluate neuronal apoptosis. The expression levels of proteins were evaluated by western blot. RESULTS EAL reduced brain infarct volume, ameliorated brain edema and improved neurological deficits in MCAO rats. In addition, EAL inhibited oxidative stress and inflammatory responses following MCAO. Besides, active compound DCMQA alleviated cytotoxicity as well as inhibited over-production of intracellular ROS and loss of MMP induced by OGD/R in SH-SY5Y cells. Moreover, EAL and DCMQA inhibited apoptosis by decreasing the expressions of pro-apoptotic proteins including bax, cytochrome c and cleaved caspase-3 while promoting the bcl-2 expression in MCAO rats and OGD/R-stimulated neurons, respectively. In addition, DCMQA suppressed the production of autophagosomes and down-regulated expression of Beclin 1 and LC3. Furthermore, inhibiting AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) signaling pathway contributed to DCMQA-mediated suppression of autophagy induced by OGD/R. CONCLUSION Our findings demonstrate that Arctium lappa L. roots protect against cerebral ischemia through inhibiting apoptosis and AMPK/mTOR-mediated autophagy in vitro and in vivo, providing a theoretical basis for the development of CQAs in Arctium lappa L. roots as neuroprotective drugs for the prevention and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yue Yang
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Huan Gao
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Wenwu Liu
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Xin Liu
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Xiaowen Jiang
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Xiang Li
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Qiong Wu
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Zihua Xu
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China
| | - Qingchun Zhao
- Department of Pharmacy, General Hospital of Northern Theater Command, 83 Wenhua Road, Shenyang, Liaoning 110016, China; School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, China.
| |
Collapse
|
15
|
Terwilliger ZS, Ryan TE, Goldberg EJ, Schmidt CA, Yamaguchi DJ, Karnekar R, Brophy P, Green TD, Zeczycki TN, Mac Gabhann F, Annex BH, McClung JM. Racial differences in the limb skeletal muscle transcriptional programs of patients with critical limb ischemia. Vasc Med 2021; 26:247-258. [PMID: 33685287 DOI: 10.1177/1358863x20983918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Critical limb ischemia (CLI) is the most severe manifestation of peripheral artery disease (PAD) and is characterized by high rates of morbidity and mortality. As with most severe cardiovascular disease manifestations, Black individuals disproportionately present with CLI. Accordingly, there remains a clear need to better understand the reasons for this discrepancy and to facilitate personalized therapeutic options specific for this population. Gastrocnemius muscle was obtained from White and Black healthy adult volunteers and patients with CLI for whole transcriptome shotgun sequencing (WTSS) and enrichment analysis was performed to identify alterations in specific Reactome pathways. When compared to their race-matched healthy controls, both White and Black patients with CLI demonstrated similar reductions in nuclear and mitochondrial encoded genes and mitochondrial oxygen consumption across multiple substrates, indicating a common bioenergetic paradigm associated with amputation outcomes regardless of race. Direct comparisons between tissues of White and Black patients with CLI revealed hemostasis, extracellular matrix organization, platelet regulation, and vascular wall interactions to be uniquely altered in limb muscles of Black individuals. Among traditional vascular growth factor signaling targets, WTSS revealed only Tie1 to be significantly altered from White levels in Black limb muscle tissues. Quantitative reverse transcription polymerase chain reaction validation of select identified targets verified WTSS directional changes and supports reductions in MMP9 and increases in NUDT4P1 and GRIK2 as unique to limb muscles of Black patients with CLI. This represents a critical first step in better understanding the transcriptional program similarities and differences between Black and White patients in the setting of amputations related to CLI and provides a promising start for therapeutic development in this population.
Collapse
Affiliation(s)
- Zoe S Terwilliger
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Emma J Goldberg
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Cameron A Schmidt
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Dean J Yamaguchi
- Department of Cardiovascular Sciences, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Division of Surgery, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Reema Karnekar
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Patricia Brophy
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Thomas D Green
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Tonya N Zeczycki
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Biochemistry, East Carolina University, Brody Medical Center, Greenville, NC, USA
| | - Feilim Mac Gabhann
- Department of Biomedical Engineering and Institute for Computational Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Brian H Annex
- Department of Medicine, Medical College of Georgia, Augusta, GA, USA.,Vascular Biology Center, Medical College of Georgia, Augusta, GA, USA
| | - Joseph M McClung
- Diabetes and Obesity Institute, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Physiology, East Carolina University, Brody Medical Center, Greenville, NC, USA.,Department of Cardiovascular Sciences, East Carolina University, Brody Medical Center, Greenville, NC, USA
| |
Collapse
|
16
|
Zheng L, Wang Z, Li Z, Wang M, Wang W, Chang G. MicroRNA-130a inhibits proliferation of vascular smooth muscle cells by suppressing autophagy via ATG2B. J Cell Mol Med 2021; 25:3829-3839. [PMID: 33611856 PMCID: PMC8051697 DOI: 10.1111/jcmm.16305] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/24/2020] [Accepted: 12/14/2020] [Indexed: 01/01/2023] Open
Abstract
Numerous microRNAs participate in regulating the pathological process of atherosclerosis. We have found miR-130a is one of the most significantly down-regulated microRNAs in arteriosclerosis obliterans. Our research explored the function of miR-130a in regulating proliferation by controlling autophagy in arteriosclerosis obliterans development. A Gene Ontology (GO) enrichment analysis of miR-130a target genes indicated a correlation between miR-130a and cell proliferation. Thus, cell cycle, CCK-8 assays and Western blot analysis were performed, and the results indicated that miR-130a overexpression in vascular smooth muscle cells (VSMCs) significantly attenuated cell proliferation, which was validated by an in vivo assay in a rat model. Moreover, autophagy is thought to be involved in the regulation of proliferation. As our results indicated, miR-130a could inhibit autophagy, and ATG2B was predicted to be a target of miR-130a. The autophagy inhibition effect of miR-130a overexpression was consistent with the effect of ATG2B knockdown. The results that ATG2B plasmids and miR-130a mimics were cotransfected in VSMCs further confirmed our conclusion. In addition, by using immunohistochemistry, the positive results of LC3 II/I and ATG2B in the rat model and artery vascular tissues from the patient were in accordance with in vitro data. In conclusion, our data demonstrate that miR-130a inhibits VSMCs proliferation via ATG2B, which indicates that miR-130a could be a potential therapeutic target that regulates autophagy in atherosclerosis obliterans.
Collapse
Affiliation(s)
- Liang Zheng
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhecun Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zilun Li
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mian Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenjian Wang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guangqi Chang
- Laboratory of General Surgery, Division of Vascular Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
17
|
Hydrogen sulfide exacerbated periodontal inflammation and induced autophagy in experimental periodontitis. Int Immunopharmacol 2021; 93:107399. [PMID: 33529908 DOI: 10.1016/j.intimp.2021.107399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/27/2022]
Abstract
Hydrogen sulfide (H2S), the metabolite produced by gram-negative bacteria, is present in deep periodontal pockets of periodontitis patients at high concentrations. The harsh conditions in the diseased periodontium may stimulate a local autophagy response. However, how H2S participates in pathogenesis and whether H2S induces autophagy in periodontitis remain partially unknown. In this article, we determined the role of the slow-releasing H2S donor GYY4137 in experimental periodontitis and its possible regulation in autophagy involved. We found that GYY4137 dose-dependently decreased cell viability and increased the level of proinflammatory cytokines in LPS-stimulated human periodontal ligament cells (HPDLCs). Topically applied GYY4137 also exacerbated periodontal inflammation and alveolar bone loss in ligature-induced rats. Moreover, GYY4137 activated autophagy by upregulating the expression levels of the autophagy-related proteins LC3 and Beclin-1 and downregulating P62 in LPS-treated HPDLCs and inflamed periodontal tissues. Blocking autophagy with 3-methyladenine resulted in further increased expression of proinflammatory cytokines in LPS- and GYY4137-induced HPDLCs. Our results indicate that GYY4137 exerted proinflammatory effects and promoted autophagy in periodontitis, and the induced autophagy may function as a cytoprotective mechanism to prevent excessive inflammation.
Collapse
|
18
|
Zheng H, Feng H, Zhang W, Han Y, Zhao W. Targeting autophagy by natural product Ursolic acid for prevention and treatment of osteoporosis. Toxicol Appl Pharmacol 2020; 409:115271. [PMID: 33065153 DOI: 10.1016/j.taap.2020.115271] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
With the growth of the aging population, osteoporosis is becoming a global health problem. Ursolic acid (UA) is an active ingredient existed in a variety of foods and nature plants and owns plenty of pharmacological effects especially in treating metabolic disease. Our predication from network pharmacology hinted that UA has potential for ameliorating osteoporosis. Firstly through in vivo experiment, we confirmed that UA administration obviously protected against ovariectomy (OVX)-induced osteoporosis in rats by improving microarchitectural deterioration of trabecular bone (P < 0.001), decreasing numbers of TRAP positive osteoclast in vertebra (P < 0.001), as well as decreasing serum osteoclast-specific cytokines release (P < 0.001). Besides, UA ameliorated kidney damage secondary to OVX-induced osteoporosis by ameliorating glomerular atrophy, decreasing BUN and creatinine levels in OVX rats. In vitro, UA noticeably decreased osteoclastic-special marker proteins c-Fos and NFATc1 expressions (P < 0.001) in response to RANKL stimulation in macrophagy. Importantly, autophagy pathway was activated in the process of osteoclast differentiation and blocked by UA pretreatment. Furthermore, autophagy inhibitors suppressed osteoclast differentiation (P < 0.001). Collectively, UA may ameliorate osteoporosis by suppressing osteoclast differentiation mediated by autophagy. Our research provides scientific support for UA treating osteoporosis and offers an optimal dose for daily intake of UA safely to prevent bone diseases.
Collapse
Affiliation(s)
- Haoyi Zheng
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Haitao Feng
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Wenzhong Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yantao Han
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China
| | - Wenwen Zhao
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong 266021, China.
| |
Collapse
|
19
|
Gao JW, He WB, Xie CM, Gao M, Feng LY, Liu ZY, Wang JF, Huang H, Liu PM. Aldosterone enhances high phosphate-induced vascular calcification through inhibition of AMPK-mediated autophagy. J Cell Mol Med 2020; 24:13648-13659. [PMID: 33150736 PMCID: PMC7754028 DOI: 10.1111/jcmm.15813] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/11/2020] [Accepted: 08/10/2020] [Indexed: 12/25/2022] Open
Abstract
It remains unclear whether the necessity of calcified mellitus induced by high inorganic phosphate (Pi) is required and the roles of autophagy plays in aldosterone (Aldo)‐enhanced vascular calcification (VC) and vascular smooth muscle cell (VSMC) osteogenic differentiation. In the present study, we found that Aldo enhanced VC both in vivo and in vitro only in the presence of high Pi, alongside with increased expression of VSMC osteogenic proteins (BMP2, Runx2 and OCN) and decreased expression of VSMC contractile proteins (α‐SMA, SM22α and smoothelin). However, these effects were blocked by mineralocorticoid receptor inhibitor, spironolactone. In addition, the stimulatory effects of Aldo on VSMC calcification were further accelerated by the autophagy inhibitor, 3‐MA, and were counteracted by the autophagy inducer, rapamycin. Moreover, inhibiting adenosine monophosphate‐activated protein kinase (AMPK) by Compound C attenuated Aldo/MR‐enhanced VC. These results suggested that Aldo facilitates high Pi‐induced VSMC osteogenic phenotypic switch and calcification through MR‐mediated signalling pathways that involve AMPK‐dependent autophagy, which provided new insights into Aldo excess‐associated VC in various settings.
Collapse
Affiliation(s)
- Jing-Wei Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wan-Bing He
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chang-Ming Xie
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Ming Gao
- Department of Radiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei-Yu Feng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhao-Yu Liu
- Medical Research Center, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hui Huang
- Cardiovascular Department, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Pin-Ming Liu
- Department of Cardiology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Jeong IH, Bae WY, Choi JS, Jeong JW. Ischemia induces autophagy of endothelial cells and stimulates angiogenic effects in a hindlimb ischemia mouse model. Cell Death Dis 2020; 11:624. [PMID: 32796816 PMCID: PMC7429831 DOI: 10.1038/s41419-020-02849-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 11/09/2022]
Abstract
Although peripheral artery disease (PAD) is a major health problem, there have been limited advances in medical therapies. In PAD patients, angiogenesis is regarded as a promising therapeutic strategy to promote new arterial vessels and improve perfusion of ischemic tissue. Autophagy plays a critical role in catabolic processes for cell survival under normal and stressful conditions and plays fundamental biological roles in various cellular functions. In the present study, we showed that autophagy in endothelial cells is important for the repair and regeneration of damaged tissues. In a hindlimb ischemia mouse model, autophagy was stimulated in endothelial cells of the quadriceps muscle, and adjacent cells proliferated and regenerated. The autophagy pathway was induced under prolonged hypoxia in endothelial cells, and autophagy increased angiogenic activities. Moreover, conditioned media from endothelial cells blocked autophagy and inhibited the proliferation of muscle cells, suggesting that autophagic stimulation in endothelial cells affects the survival of adjacent cells, such as muscle. Collectively, hypoxia/ischemia-induced autophagy angiogenesis, and the damaged tissue surrounded by neo-vessels was regenerated in an ischemia model. Therefore, we strongly suggest that stimulation of autophagy in endothelial cells may be a potent therapeutic strategy in severe vascular diseases, including PAD.
Collapse
Affiliation(s)
- In-Hye Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Woom-Yee Bae
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae-Sun Choi
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea. .,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
21
|
Salidroside-Mediated Autophagic Targeting of Active Src and Caveolin-1 Suppresses Low-Density Lipoprotein Transcytosis across Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9595036. [PMID: 32685103 PMCID: PMC7333065 DOI: 10.1155/2020/9595036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 05/14/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022]
Abstract
Subendothelial retention of apolipoprotein B100-containing lipoprotein, such as low-density lipoprotein (LDL), is the initial step of atherogenesis. Activation of autophagy exhibits beneficial effects for the treatment of atherosclerosis. In our previous study, we demonstrated that hyperglycemia suppressed autophagic degradation of caveolin-1, which in turn resulted in acceleration of caveolae-mediated LDL transcytosis across endothelial cells and lipid retention. Therefore, targeting the crossed pathway in autophagy activation and LDL transcytosis interruption may be a promising antiatherosclerotic strategy. In metabolic diseases, including atherosclerosis, salidroside, a phenylpropanoid glycoside compound (3,5-dimethoxyphenyl) methyl-β-glucopyranoside), is the most important compound responsible for the therapeutic activities of Rhodiola. However, whether salidroside suppresses LDL transcytosis to alleviate atherosclerosis has not yet been elucidated. In the present study, we demonstrated that salidroside significantly decreased LDL transcytosis across endothelial cells. Salidroside-induced effects were dramatically blocked by AMPK (adenosine monophosphate-activated protein kinase) inhibitor (compound c, AMPKα siRNA) and by overexpression of exogenous tyrosine-phosphorylated caveolin-1 using transfected cells with phosphomimicking caveolin-1 on tyrosine 14 mutant plasmids (Y14D). Furthermore, we observed that salidroside promoted autophagosome formation via activating AMPK. Meanwhile, the interaction between caveolin-1 and LC3B-II, as well as the interaction between active Src (indicated by the phosphorylation of Src on tyrosine 416) and LC3B-II, was significantly increased, upon stimulation with salidroside. In addition, both bafilomycin A1 (a lysosome inhibitor) and an AMPK inhibitor (compound c) markedly prevented salidroside-induced autophagic degradation of p-Src and caveolin-1. Moreover, the phosphorylation of caveolin-1 on tyrosine 14 was disrupted due to the downregulation of p-Src and caveolin-1, thereby directly decreasing LDL transcytosis by attenuating the number of caveolae on the cell membrane and by preventing caveolae-mediated LDL endocytosis released from the cell membrane. In ApoE−/− mice, salidroside significantly delayed the formation of atherosclerotic lesions. Meanwhile, a significant increase in LC3B, accompanied by attenuated accumulation of the autophagy substrate SQSTM1, was observed in aortic endothelium of ApoE−/− mice. Taken together, our findings demonstrated that salidroside protected against atherosclerosis by inhibiting LDL transcytosis through enhancing the autophagic degradation of active Src and caveolin-1.
Collapse
|
22
|
Kim D, Hwang HY, Kwon HJ. Targeting Autophagy In Disease: Recent Advances In Drug Discovery. Expert Opin Drug Discov 2020; 15:1045-1064. [DOI: 10.1080/17460441.2020.1773429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Dasol Kim
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hui-Yun Hwang
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Ho Jeong Kwon
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Liu Y, Li J, Han Y, Chen Y, Liu L, Lang J, Yang C, Luo H, Ning J. Advanced glycation end-products suppress autophagy by AMPK/mTOR signaling pathway to promote vascular calcification. Mol Cell Biochem 2020; 471:91-100. [PMID: 32514882 DOI: 10.1007/s11010-020-03769-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 05/31/2020] [Indexed: 12/16/2022]
Abstract
Vascular calcification is closely linked to patients in diabetes mellitus and chronic kidney disease. Advanced glycation end-products (AGEs) are associated with osteogenic differentiation of vascular smooth muscle cell (VSMC), vascular calcification, and autophagy that takes part in the process. However, the underlying mechanism of the effects of AGEs on the phenotypic transition and autophagy of VSMCs is not clearly understood. In this study, we cultured the rat VSMC line (A7R5) and thoracic aorta organ with bovine serum albumin (BSA) or AGEs (AGEs-BSA) and detected proteins expression by Western blotting or immunofluorescence. Autophagosome was observed by transmission electron microscopy (TEM). The mineralization and calcific nodules were identified by Alizarin Red S and Von Kossa staining. AGEs significantly downregulated p-AMPKα expression and upregulated p-mTOR expression and then increased the expression of osteoblastic differentiation, while suppressing autophagy in a time-dependent pattern. Pretreatment with autophagy activator rapamycin and AMPK activator AICAR both upregulated the autophagy level and downregulated the effects of AGEs on osteoblastic differentiation of VSMCs. Moreover, the result from rat thoracic aorta culture also confirmed that AGEs promote vascular calcification in a time-dependent manner. Thus, our study showed that AGEs quicken vascular calcification and suppress autophagy associated with AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yanyan Liu
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Jing Li
- Department of Endocrinology, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Yuting Han
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Yuying Chen
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Lixuan Liu
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Jiangli Lang
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Chuan Yang
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou, 510120, China
| | - Hengcong Luo
- Department of Endocrinology, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China.
| | - Jie Ning
- Department of Endocrinology, Shenzhen Longhua District Central Hospital, Guangdong Medical University Affiliated Longhua Central Hospital, Shenzhen, 518110, Guangdong, China.
| |
Collapse
|
24
|
Li X, Zhou J, Dou Y, Shi Y, Wang Y, Hong J, Zhao J, Zhang J, Yuan Y, Zhou M, Wei X. The protective effects of angelica organic acid against ox-LDL-induced autophagy dysfunction of HUVECs. BMC Complement Med Ther 2020; 20:164. [PMID: 32487223 PMCID: PMC7268640 DOI: 10.1186/s12906-020-02968-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/19/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Angelica root is the dry root of the Umbelliferae plant Angelica sinensis (oliv) Diels. Angelica organic acid (OA) is the main active ingredient in Angelica sinensis, and it exerts potential anti-atherosclerotic effects by preventing Oxidized low-density lipoprotein (Ox-LDL) induced endothelial injury. To study the protective effects of OA on ox-LDL-induced HUVECs autophagic flux dysfunction and inflammatory injury. METHODS OA were isolated by water extraction and alcohol precipitation, and then the content of ferulic acid (FA) in the OA was determined by high performance liquid chromatography. The ox-LDL-induced endothelial injury model was established. The effect of ferulic acid on the survival of Human umbilical vein endothelial cells (HVUECs) was detected by CCK-8 assay. HUVECs were pretreated with different concentrations of OA (20 μmol/L, 40 μmol/L, and 80 μmol/L), and Western Blot was used to detect the expressions of LC3II, p62, MCP-1, VCAM-1 and LOX-1. The autophagosomes in HUVECs were observed by transmission electron microscopy (TEM). RESULTS 20 μmol/L OA could increase the expression of LC3II and decrease the expression of p62, MCP-1, VCAM-1 and LOX-1. The results of TEM showed that angelica organic acids promoted cell organelle degradation in autolysosomes. CONCLUSION OA could reduce inflammation, protect endothelial cells and play an anti-atherosclerotic role by enhancing the autophagy flux of damaged endothelial cells, in which FA the major active ingredient of OA played a major role.
Collapse
Affiliation(s)
- Xuefeng Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jing Zhou
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yinghuan Dou
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yanbin Shi
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Ying Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jianli Hong
- Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Junnan Zhao
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiaying Zhang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yang Yuan
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Mengru Zhou
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Xiangxiang Wei
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
25
|
Hyperoside attenuates pregnancy loss through activating autophagy and suppressing inflammation in a rat model. Life Sci 2020; 254:117735. [PMID: 32360572 DOI: 10.1016/j.lfs.2020.117735] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/24/2020] [Accepted: 04/25/2020] [Indexed: 12/20/2022]
Abstract
AIMS Recurrent pregnancy loss (RPL) is one of the most common obstetrical diseases, which is a manifestation of antiphospholipid syndrome (APS) with no effective therapy methods. Autophagy and inflammatory responses both play an important role in the pathogenesis of RPL and hyperoside has been demonstrated to have multifarious bioactivities including enhancing autophagy and anti-inflammation. This study aims to investigate the effect of hyperoside on anticardiolipin (aCL)-IgG fractions-induced pregnancy loss. MAIN METHODS In the present study, the effect of hyperoside was evaluated in a rat model of pregnancy loss induced by aCL-IgG fractions isolated from serum of APS patients. The fetuses were counted and the placentas were weighted and the protein expressions of inflammation and autophagy were measured by western blot analysis. KEY FINDINGS Treatment with hyperoside (40 mg/kg) improved pregnancy outcome manifest as increasing the weight of fetuses and decreasing the fetal resorption rate. In addition, hyperoside treatment downregulated the expressions of phosphorylated mammalian target of rapamycin (mTOR), phosphorylated p70S6 Kinase (S6K) and inhibited the expressions of Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88) and NF-kB p-p65 in pregnancy loss animal models. SIGNIFICANCE Hyperoside attenuated pregnancy loss through regulating mTOR/S6K and TLR4/MyD88/NF-kB signaling pathways, which may provide a potential drug candidate for recurrent pregnancy loss therapy.
Collapse
|
26
|
Luo Y, Lu S, Gao Y, Yang K, Wu D, Xu X, Sun G, Sun X. Araloside C attenuates atherosclerosis by modulating macrophage polarization via Sirt1-mediated autophagy. Aging (Albany NY) 2020; 12:1704-1724. [PMID: 31986489 PMCID: PMC7053643 DOI: 10.18632/aging.102708] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 01/02/2020] [Indexed: 12/17/2022]
Abstract
Atherosclerosis-related cardiovascular disease is still the predominant cause of death worldwide. Araloside C (AsC), a natural saponin, exerts extensive anti-inflammatory properties. In this study, we explored the protective effects and mechanism of AsC on macrophage polarization in atherosclerosis in vivo and in vitro. Using a high-fat diet (HFD)-fed ApoE-/- mouse model and RAW264.7 macrophages exposed to ox-LDL, AsC was evaluated for its effects on polarization and autophagy. AsC significantly reduced the plaque area in atherosclerotic mice and lipid accumulation in ox-LDL-treated macrophages, promoted M2 phenotype macrophage polarization, increased the number of autophagosomes and modulated the expression of autophagy-related proteins. Moreover, the autophagy inhibitor 3-methyladenine and BECN1 siRNA obviously abolished the antiatherosclerotic and M2 macrophage polarization effects of AsC. Mechanistically, AsC targeted Sirt1and increased its expression, and this increase in expression was associated with increased autophagy and M2 phenotype polarization. In contrast, the effects of AsC were markedly blocked by EX527 and Sirt1 siRNA. Altogether, AsC attenuates foam cell formation and lessens atherosclerosis by modulating macrophage polarization via Sirt1-mediated autophagy.
Collapse
Affiliation(s)
- Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Shan Lu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Ye Gao
- College of Pharmacy, Harbin University of Commerce, Harbin 150076, Heilongjiang, China
| | - Ke Yang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Daoshun Wu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xudong Xu
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing 100193, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing 100193, China
| |
Collapse
|
27
|
Bai XL, Deng XL, Wu GJ, Li WJ, Jin S. Rhodiola and salidroside in the treatment of metabolic disorders. Mini Rev Med Chem 2019; 19:1611-1626. [PMID: 31481002 DOI: 10.2174/1389557519666190903115424] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 07/31/2017] [Accepted: 07/31/2017] [Indexed: 12/14/2022]
Abstract
Over the past three decades, the knowledge gained about the mechanisms that underpin the potential use of Rhodiola in stress- and ageing-associated disorders has increased, and provided a universal framework for studies that focused on the use of Rhodiola in preventing or curing metabolic diseases. Of particular interest is the emerging role of Rhodiola in the maintenance of energy homeostasis. Moreover, over the last two decades, great efforts have been undertaken to unravel the underlying mechanisms of action of Rhodiola in the treatment of metabolic disorders. Extracts of Rhodiola and salidroside, the most abundant active compound in Rhodiola, are suggested to provide a beneficial effect in mental, behavioral, and metabolic disorders. Both in vivo and ex vivo studies, Rhodiola extracts and salidroside ameliorate metabolic disorders when administered acutely or prior to experimental injury. The mechanism involved includes multi-target effects by modulating various synergistic pathways that control oxidative stress, inflammation, mitochondria, autophagy, and cell death, as well as AMPK signaling that is associated with possible beneficial effects on metabolic disorders. However, evidence-based data supporting the effectiveness of Rhodiola or salidroside in treating metabolic disorders is limited. Therefore, a comprehensive review of available trials showing putative treatment strategies of metabolic disorders that include both clinical effective perspectives and fundamental molecular mechanisms is warranted. This review highlights studies that focus on the potential role of Rhodiola extracts and salidroside in type 2 diabetes and atherosclerosis, the two most common metabolic diseases.
Collapse
Affiliation(s)
- Xiang-Li Bai
- Department of Clinical Laboratory, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430077, China
| | - Xiu-Ling Deng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guang-Jie Wu
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan, Hubei 430077, China
| | - Wen-Jing Li
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan, Hubei 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology. Wuhan, Hubei 430077, China
| |
Collapse
|
28
|
Tian J, Popal MS, Zhao Y, Liu Y, Chen K, Liu Y. Interplay between Exosomes and Autophagy in Cardiovascular Diseases: Novel Promising Target for Diagnostic and Therapeutic Application. Aging Dis 2019; 10:1302-1310. [PMID: 31788341 PMCID: PMC6844582 DOI: 10.14336/ad.2018.1020] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/20/2018] [Indexed: 12/12/2022] Open
Abstract
Exosome, is identified as a nature nanocarrier and intercellular messenger that regulates cell to cell communication. Autophagy is critical in maintenance of protein homeostasis by degradation of damaged proteins and organelles. Autophagy and exosomes take pivotal roles in cellular homeostasis and cardiovascular disease. Currently, the coordinated mechanisms for exosomes and autophagy in the maintenance of cellular fitness are now garnering much attention. In the present review, we discussed the interplay of exosomes and autophagy in the context of physiology and pathology of the heart, which might provide novel insights for diagnostic and therapeutic application of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinfan Tian
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mohammad Sharif Popal
- 1Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yingke Zhao
- 3Li Ka Shing Faculty of Medicine, The University of HongKong, Pokfulam, Hong Kong
| | - Yanfei Liu
- 4Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Keji Chen
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yue Liu
- 2Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
29
|
Tang Y, Zhang Y, Liu S, Sun Z, Wang C, Li L, Zhou W, Cheng S. 14-3-3ζ binds to and stabilizes phospho-beclin 1 S295 and induces autophagy in hepatocellular carcinoma cells. J Cell Mol Med 2019; 24:954-964. [PMID: 31709727 PMCID: PMC6933394 DOI: 10.1111/jcmm.14806] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 09/21/2019] [Accepted: 10/20/2019] [Indexed: 12/21/2022] Open
Abstract
Data from The Cancer Genome Atlas (TCGA) indicate that the expression levels of 14‐3‐3ζ and beclin 1 (a key molecule involved in cellular autophagy) are up‐regulated and positively correlated with each other (R = .5, P < .05) in HCC tissues. Chemoresistance developed in hepatoma cancer cells is associated with autophagy initiation. This study aimed to explore 14‐3‐3ζ’s role in regulating autophagy in HCC cells, with a focus on beclin 1. The co‐localization of 14‐3‐3ζ and beclin 1 was detectable in primary HCC tissues. To simulate in vivo tumour microenvironment (hypoxia), CSQT‐2 and HCC‐LM3 cells were exposed to 2% oxygen for 24 hours. The protein levels of 14‐3‐3ζ and phospho‐beclin 1S295 peaked at 12 hours following hypoxia. Meanwhile, the strongest autophagy flux occurred: LC3II was increased, and p62 was decreased significantly. By sequencing the coding area of BECN 1 gene of CSQT‐2 and HCC‐LM3 cells, we found that the predicted translational products of BECN 1 gene contained RLPS295VP (R, arginine; L, leucine; P, proline; S, serine; V, valine), a classic 14‐3‐3ζ binding motif. CO‐IP results confirmed that 14‐3‐3ζ bound to beclin 1, and this connection was markedly weakened when S295 was mutated into A295 (alanine). Further, 14‐3‐3ζ overexpression prevented phospho‐beclin 1S295 from degradation and enhanced its binding to VPS34, whilst its knockdown accelerated the degradation. Additionally, 14‐3‐3ζ enhanced the chemoresistance of HCC cells to cis‐diammined dichloridoplatium by activating autophagy. Our work reveals that 14‐3‐3ζ binds to and stabilizes phospho‐beclin 1S295 and induces autophagy in HCC cells to resist chemotherapy.
Collapse
Affiliation(s)
- Yufu Tang
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Yibing Zhang
- Department of Medical Affairs, The General Hospital of Northern Theater Command, Shenyang, China
| | - Shupeng Liu
- Department of Gynecology and Obstetrics, Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zhongyi Sun
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Chunhui Wang
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Longfei Li
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Wenping Zhou
- Department of Hepatobiliary Surgery, The General Hospital of Northern Theater Command, Shenyang, China
| | - Shuqun Cheng
- Department of Liver Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Schizandrin Protects against OGD/R-Induced Neuronal Injury by Suppressing Autophagy: Involvement of the AMPK/mTOR Pathway. Molecules 2019; 24:molecules24193624. [PMID: 31597329 PMCID: PMC6804185 DOI: 10.3390/molecules24193624] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 09/25/2019] [Accepted: 10/06/2019] [Indexed: 01/11/2023] Open
Abstract
The neuroprotective role of schizandrin (SA) in cerebral ischemia-reperfusion (I/R) was recently highlighted. However, whether SA plays a regulatory role on autophagy in cerebral I/R injury is still unclear. This study aimed to explore whether the neuroprotective mechanisms of SA were linked to its regulation of AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/autophagy pathway in vivo and in vitro. The present study confirmed that SA significantly improved oxygen-glucose deprivation/re-oxygenation (OGD/R)-induced PC12 cells injury. The results of immunoblotting and confocal microscope showed that SA decreased autophagy in OGD/R-injured PC12 cells, which was reflected by the decreased Beclin-1 and LC3-II expression, autophagy flux level, and LC3 puncta formation. In addition, the autophagy inducer rapamycin partially prevented the effects of SA on cell viability and autophagy after OGD/R, whereas the autophagy inhibitor 3-methyladenine (3-MA) exerted the opposite effect. The results of Western blotting showed that SA markedly decreased the phosphorylation of AMPK (p-AMPK), whereas the phosphor-mTOR (p-mTOR) levels increased in the presence of OGD/R insult. Furthermore, pretreatment with the AMPK inducer AICAR partially reversed the protective effects and autophagy inhibition of SA. However, AMPK inhibitor Compound C pretreatment further promoted the inhibition of SA on autophagy induction and cell damage induced by OGD/R. Taken together, these findings demonstrate that SA protects against OGD/R insult by inhibiting autophagy through the regulation of the AMPK-mTOR pathway and that SA may have therapeutic value for protecting neurons from cerebral ischemia.
Collapse
|
31
|
Moulis M, Grousset E, Faccini J, Richetin K, Thomas G, Vindis C. The Multifunctional Sorting Protein PACS-2 Controls Mitophagosome Formation in Human Vascular Smooth Muscle Cells through Mitochondria-ER Contact Sites. Cells 2019; 8:cells8060638. [PMID: 31242668 PMCID: PMC6627983 DOI: 10.3390/cells8060638] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/20/2019] [Accepted: 06/22/2019] [Indexed: 12/19/2022] Open
Abstract
Mitochondria-associated ER membranes (MAMs) are crucial for lipid transport and synthesis, calcium exchange, and mitochondrial functions, and they also act as signaling platforms. These contact sites also play a critical role in the decision between autophagy and apoptosis with far reaching implications for cell fate. Vascular smooth muscle cell (VSMC) apoptosis accelerates atherogenesis and the progression of advanced lesions, leading to atherosclerotic plaque vulnerability and medial degeneration. Though the successful autophagy of damaged mitochondria promotes VSMC survival against pro-apoptotic atherogenic stressors, it is unknown whether MAMs are involved in VSMC mitophagy processes. Here, we investigated the role of the multifunctional MAM protein phosphofurin acidic cluster sorting protein 2 (PACS-2) in regulating VSMC survival following a challenge by atherogenic lipids. Using high-resolution confocal microscopy and proximity ligation assays, we found an increase in MAM contacts as in PACS-2-associated MAMs upon stimulation with atherogenic lipids. Correspondingly, the disruption of MAM contacts by PACS-2 knockdown impaired mitophagosome formation and mitophagy, thus potentiating VSMC apoptosis. In conclusion, our data shed new light on the significance of the MAM modulatory protein PACS-2 in vascular cell physiopathology and suggest MAMs may be a new target to modulate VSMC fate and favor atherosclerotic plaque stability.
Collapse
Affiliation(s)
- Manon Moulis
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases/I2MC, F-31342 Toulouse, France.
- University of Toulouse III, F-31342 Toulouse, France.
| | - Elisa Grousset
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases/I2MC, F-31342 Toulouse, France.
- University of Toulouse III, F-31342 Toulouse, France.
| | - Julien Faccini
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases/I2MC, F-31342 Toulouse, France.
- University of Toulouse III, F-31342 Toulouse, France.
| | - Kevin Richetin
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, 1005 Lausanne, Switzerland.
| | - Gary Thomas
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
- Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Cecile Vindis
- INSERM, UMR 1048, Institute of Metabolic and Cardiovascular Diseases/I2MC, F-31342 Toulouse, France.
- University of Toulouse III, F-31342 Toulouse, France.
| |
Collapse
|
32
|
Abstract
During the pathogenesis of early atherosclerosis, lipid-loaded macrophages are involved in plaque development and progression. As a novel adipokine, C1q/tumor necrosis factor–related protein-9 (CTRP9) has beneficial effects in cardiovascular disease. However, previous reports have not studied whether the formation of macrophage foam cell induced by oxidized low-density lipoprotein (ox-LDL) is affected by CTRP9. According to our study, in ox-LDL–induced THP-1 macrophages, CTRP9 could reduce the quantity of lipid droplets, lower the level of cholesteryl ester (CE), promote cholesterol efflux, as well as increase the expression level of the cholesterol transport receptors ATP-binding membrane cassette transporter A1 (ABCA1) and G1 (ABCG1). In addition, the protein of LC3 II is elevated and that of p62 is decreased in CTRP9-treated foam cells by enhancing autophagy. However, using 3-methyladenine (3-MA) abolished the role of CTRP9 by inhibiting autophagy. Mechanistically, the autophagy-promoting effects of CTRP9 on foam cells was reversed by an AMPK inhibitor, Compound C, which inhibited the signaling pathway of adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR). These results show that CTRP9 protects against atherosclerosis by promoting cholesterol efflux to reduce the formation of foam cell in virtue of inducing autophagy in an AMPK/mTOR signaling pathway–dependent manner.
Collapse
|
33
|
Zhou M, Ren P, Zhang Y, Li S, Li M, Li P, Shang J, Liu W, Liu H. Shen-Yuan-Dan Capsule Attenuates Atherosclerosis and Foam Cell Formation by Enhancing Autophagy and Inhibiting the PI3K/Akt/mTORC1 Signaling Pathway. Front Pharmacol 2019; 10:603. [PMID: 31214032 PMCID: PMC6554665 DOI: 10.3389/fphar.2019.00603] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 05/13/2019] [Indexed: 01/08/2023] Open
Abstract
Background and Aim: The phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin complex 1 (mTORC1) signaling pathway plays a crucial role in autophagy and inflammation. Our previous studies demonstrated that Shen-Yuan-Dan Capsule (SYDC), a Chinese medicine used for treating angina pectoris, has anti-atherosclerotic and anti-inflammatory effects in mice. However, its effects on autophagy and the PI3K/Akt/mTORC1 signaling pathway remain unclear. This study aimed to explore the effects of SYDC on autophagy and PI3K/Akt/mTORC1 signaling in the apolipoprotein E knockout (ApoE−/−) mouse model and in macrophage-derived foam cells to delineate the underlying mechanism. Methods: After 6 weeks of high-fat diet, ApoE–/– mice were randomly grouped into control, Lipitor, low-SYDC (SYDC-L), middle-SYDC (SYDC-M), and high-SYDC (SYDC-H) groups (n = 10). The mice were intragastrically administered the respective treatment for 6 weeks. Murine RAW264.7 cells were stimulated with oxidized low-density lipoprotein (ox-LDL) (80 µg/ml) for 24 h and then pretreated with SYDC freeze-dried powder for another 24 h. Cells treated with SYDC were co-cultured for 24 h with LY294002, tricirbine, and rapamycin to investigate the effects on the PI3K/Akt/mTORC1 signaling pathway. Results: SYDC ameliorated blood lipid levels, reduced the atherosclerotic index and plaque areas in the aortic root in mice, and inhibited total cholesterol (TC) levels and cholinesterase (ChE)/TC ratios in ox-LDL stimulated macrophages. Moreover, SYDC up-regulated Beclin1 and LC3II/I proteins in mice and in the ox-LDL–stimulated macrophages. Moreover, SYDC inhibited AKT phosphorylation at Ser473 and mTOR phosphorylation at Ser2448 in mice and in ox-LDL–stimulated macrophages. Furthermore, SYDC’s inhibitory of ChE/TC ratios in ox-LDL–stimulated macrophages was not changed by selective inhibition of the PI3K/Akt/mTORC1 pathway. Conclusions: Our results highlight that SYDC treatment attenuates foam cell formation by promoting autophagy via inhibiting activation of the PI3K/Akt/mTORC1 signaling pathway. This study provides new insights into the molecular mechanism underlying SYDC’s therapeutic potential for treating atherosclerosis.
Collapse
Affiliation(s)
- Mingxue Zhou
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Pan Ren
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Ying Zhang
- Department of Traditional Chinese Medicine, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Sinai Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Mengjie Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Ping Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Juju Shang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weihong Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing Institute of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
34
|
Nabavi SF, Sureda A, Sanches-Silva A, Pandima Devi K, Ahmed T, Shahid M, Sobarzo-Sánchez E, Dacrema M, Daglia M, Braidy N, Vacca RA, Berindan-Neagoe I, Gulei D, Barreca D, Banach M, Nabavi SM, Dehpour AR, Shirooie S. Novel therapeutic strategies for stroke: The role of autophagy. Crit Rev Clin Lab Sci 2019; 56:182-199. [PMID: 30821184 DOI: 10.1080/10408363.2019.1575333] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Autophagy is an important biological mechanism involved in the regulation of numerous fundamental cellular processes that are mainly associated with cellular growth and differentiation. Autophagic pathways are vital for maintaining cellular homeostasis by enhancing the turnover of nonfunctional proteins and organelles. Neuronal cells, like other eukaryotic cells, are dependent on autophagy for neuroprotection in response to stress, but can also induce cell death in cerebral ischemia. Recent studies have demonstrated that autophagy may induce neuroprotection following acute brain injury, including ischemic stroke. However in some special circumstances, activation of autophagy can induce cell death, playing a deleterious role in the etiology and progression of ischemic stroke. Currently, there are no therapeutic options against stroke that demonstrate efficient neuroprotective abilities. In the present work, we will review the significance of autophagy in the context of ischemic stroke by first outlining its role in ischemic neuronal death. We will also highlight the potential therapeutic applications of pharmacological modulators of autophagy, including some naturally occurring polyphenolic compounds that can target this catabolic process. Our findings provide renewed insight on the mechanism of action of autophagy in stroke together with potential neuroprotective compounds, which may partially exert their function through enhancing mitochondrial function and attenuating damaging autophagic processes.
Collapse
Affiliation(s)
- Seyed Fazel Nabavi
- a Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Antoni Sureda
- b Research Group on Community Nutrition and Oxidative Stress (NUCOX) and CIBEROBN (Physiopathology of Obesity and Nutrition CB12/03/30038), University of Balearic Islands , Palma de Mallorca , Spain
| | - Ana Sanches-Silva
- c National Institute for Agricultural and Veterinary Research (INIAV) , Vila do Conde , Portugal.,d Center for Study in Animal Science (CECA), ICETA, University of Oporto , Oporto , Portugal
| | - Kasi Pandima Devi
- e Department of Biotechnology , Alagappa University , Karaikudi , Tamil Nadu, India
| | - Touqeer Ahmed
- f Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology , Islamabad , Pakistan
| | - Momina Shahid
- f Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology , Islamabad , Pakistan
| | - Eduardo Sobarzo-Sánchez
- g Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Santiago de Compostela , Santiago de Compostela , Spain.,h Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud , Universidad Central de Chile , Chile
| | - Marco Dacrema
- i Department of Drug Sciences , Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia , Pavia , Italy
| | - Maria Daglia
- i Department of Drug Sciences , Medicinal Chemistry and Pharmaceutical Technology Section, University of Pavia , Pavia , Italy
| | - Nady Braidy
- j Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales , New South Wales , Australia
| | - Rosa Anna Vacca
- k Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies National Council of Research , Bari , Italy
| | - Ioana Berindan-Neagoe
- l MEDFUTURE - Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania.,m Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania.,n Department of Functional Genomics and Experimental Pathology , The Oncology Institute "Prof. Dr. Ion Chiricuta" , Cluj-Napoca , Romania
| | - Diana Gulei
- l MEDFUTURE - Research Center for Advanced Medicine, "Iuliu-Hatieganu" University of Medicine and Pharmacy , Cluj-Napoca , Romania
| | - Davide Barreca
- o Department of Chemical, Biological, Pharmaceutical and Environmental Sciences , University of Messina , Messina , Italy
| | - Maciej Banach
- p Department of Hypertension , WAM University Hospital in Lodz, Medical University of Lodz , Lodz , Poland.,q Polish Mother's Memorial Hospital Research Institute (PMMHRI) , Lodz , Poland.,r Cardiovascular Research Centre, University of Zielona Gora , Zielona Gora , Poland
| | - Seyed Mohammad Nabavi
- a Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Ahmad Reza Dehpour
- s Department of Pharmacology, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,t Experimental Medicine Research Center, Tehran University of Medical Sciences , Tehran , Iran
| | - Samira Shirooie
- u Department of Pharmacology, School of Pharmacy , Kermanshah University of Medical Sciences , Kermanshah , Iran
| |
Collapse
|
35
|
Autophagy-associated signal pathways of functional foods for chronic diseases. FOOD SCIENCE AND HUMAN WELLNESS 2019. [DOI: 10.1016/j.fshw.2019.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
36
|
Yao BC, Meng LB, Hao ML, Zhang YM, Gong T, Guo ZG. Chronic stress: a critical risk factor for atherosclerosis. J Int Med Res 2019; 47:1429-1440. [PMID: 30799666 PMCID: PMC6460614 DOI: 10.1177/0300060519826820] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic stress refers to the non-specific systemic reaction that occurs when the body is stimulated by various internal and external negative factors over a long time. The physiological response to chronic stress exposure has long been recognized as a potent modulator in the occurrence of atherosclerosis. Furthermore, research has confirmed the correlation between atherosclerosis and cardiovascular events. Chronic stress is pervasive during negative life events and may lead to the formation of plaque. Several epidemiological studies have shown that chronic stress is an independent risk factor for the development of vascular disease and for increased morbidity and mortality in patients with pre-existing coronary artery disease. One possible mechanism for this process is that chronic stress causes endothelial injury, directly activating macrophages, promoting foam cell formation and generating the formation of atherosclerotic plaque. This mechanism involves numerous variables, including inflammation, signal pathways, lipid metabolism and endothelial function. The mechanism of chronic stress in atherosclerosis should be further investigated to provide a theoretical basis for efforts to eliminate the effect of chronic stress on the cardiocerebral vascular system.
Collapse
Affiliation(s)
- Bo-Chen Yao
- 1 Graduate College, Tianjin Medical University, Tianjin, China.,2 Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, China
| | - Ling-Bing Meng
- 3 Neurology Department, Beijing Hospital, National Center of Gerontology, Dong Dan, Beijing, P. R. China
| | - Meng-Lei Hao
- 4 Department of geriatric medicine, Qinghai University, Xining, Qinghai, China
| | - Yuan-Meng Zhang
- 5 Internal medicine, Jinzhou Medical University, Linghe District, Jinzhou City, Liaoning Province, China
| | - Tao Gong
- 1 Graduate College, Tianjin Medical University, Tianjin, China
| | - Zhi-Gang Guo
- 2 Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin, China
| |
Collapse
|
37
|
Altered mitochondrial quality control in Atg7-deficient VSMCs promotes enhanced apoptosis and is linked to unstable atherosclerotic plaque phenotype. Cell Death Dis 2019; 10:119. [PMID: 30741928 PMCID: PMC6370858 DOI: 10.1038/s41419-019-1400-0] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 01/06/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are one of the main cellular determinants in arterial pathology. A large body of evidence indicates that death of VSMCs is associated with features of high-risk/vulnerable atherosclerotic plaques. Mitochondrial turnover is an essential aspect of the mitochondrial quality control in which dysfunctional mitochondria are selectively eliminated through autophagy and replaced through expansion of preexisting mitochondria. Even though successful autophagy promotes VSMC survival, it is unclear whether reduced autophagic flux affects mitochondrial quality control of VSMCs in atherosclerotic plaques. By using apolipoprotein E-deficient (ApoE−/−) mice carrying a VSMC-specific deletion of the essential autophagy gene Atg7, we show in the present study that impaired VSMC autophagy promotes an unstable plaque phenotype, as well as the accumulation of fragmented mitochondria with reduced bioenergetic efficiency and more oxidative stress. Furthermore, we demonstrate that disrupted autophagic flux is linked to defective mitophagy and biogenesis of mitochondria, which exacerbate VSMC apoptosis and in turn plaque vulnerability. Overall, our data indicate that mitochondrial quality control is a promising therapeutic target to stabilize atherosclerotic plaques.
Collapse
|
38
|
Hou X, Summer R, Chen Z, Tian Y, Ma J, Cui J, Hao X, Guo L, Xu H, Wang H, Liu H. Lipid Uptake by Alveolar Macrophages Drives Fibrotic Responses to Silica Dust. Sci Rep 2019; 9:399. [PMID: 30674959 PMCID: PMC6344530 DOI: 10.1038/s41598-018-36875-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
Silicosis is a common occupational disease and represents a significant contributor to respiratory morbidity and mortality worldwide. Lipid-laden macrophages, or foam cells, are observed in the lungs of patients with silicosis but the mechanisms mediating their formation remain poorly understood. In this study, we sought to elucidate the mechanisms by which silica promotes foam cell formation in the lung, and to determine whether uptake of lipids alone is sufficient to drive TGF-β production by alveolar macrophages. Consistent with previous reports, we found that foam cells were markedly increased in the lungs of patients with silicosis and that these findings associated with both higher levels of intracellular lipid levels (oxidized LDL, ox-LDL) and elevated transcript levels for the lipid scavenger receptor CD36 and the nuclear receptor PPARγ. Employing a rat alveolar macrophage cell line, we found that exposure to silica dust or ox-LDL alone had a modest effect on the induction of foam cell formation and only silica was capable of inducing the production of TGF-β. In contrast, foam cell formation and TGF-β production were both dramatically increased when cells were exposed to a combination of silica dust and ox-LDL. Moreover, we found that these endpoints were markedly attenuated by either blocking CD36 or inhibiting the activity of PPARγ. Altogether, our findings suggest that foam cell formation and TGF-β production are driven by the simultaneous uptake of silica and lipids in alveolar macrophages and that strategies aimed at blocking lipid uptake by alveolar macrophages might be effective in ameliorating fibrotic responses to silica in the lung.
Collapse
Affiliation(s)
- Xiaomin Hou
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Ziying Chen
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Ying Tian
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Jingjing Ma
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Xiaohui Hao
- Medical Research Center, Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Lingli Guo
- Medical Research Center, Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Hong Xu
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.,Medical Research Center, Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China
| | - Hongli Wang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China. .,Medical Research Center, Health, North China University of Science and Technology, Tangshan, Hebei, 063210, China.
| |
Collapse
|
39
|
Jiang X, Niu X, Guo Q, Dong Y, Xu J, Yin N, Qi Q, Jia Y, Gao L, He Q, Lv P. FoxO1-mediated autophagy plays an important role in the neuroprotective effects of hydrogen in a rat model of vascular dementia. Behav Brain Res 2019; 356:98-106. [DOI: 10.1016/j.bbr.2018.05.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/21/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
|
40
|
Autophagy in Metabolic Age-Related Human Diseases. Cells 2018; 7:cells7100149. [PMID: 30249977 PMCID: PMC6210409 DOI: 10.3390/cells7100149] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a highly conserved homeostatic cellular mechanism that mediates the degradation of damaged organelles, protein aggregates, and invading pathogens through a lysosome-dependent pathway. Over the last few years, specific functions of autophagy have been discovered in many tissues and organs; however, abnormal upregulation or downregulation of autophagy has been depicted as an attribute of a variety of pathologic conditions. In this review, we will describe the current knowledge on the role of autophagy, from its regulation to its physiological influence, in metabolic age-related disorders. Finally, we propose to discuss the therapeutic potential of pharmacological and nutritional modulators of autophagy to treat metabolic diseases.
Collapse
|
41
|
Liu M, Li H, Zhou Q, Zhao H, Lv D, Cao J, Jiang J, Tang M, Wu D, Liu J, Wu L, Hu H, He L, Huang S, Chen Z, Li L, Chen L. ROS-Autophagy pathway mediates monocytes-human umbilical vein endothelial cells adhesion induced by apelin-13. J Cell Physiol 2018; 233:6839-6850. [PMID: 29691838 DOI: 10.1002/jcp.26554] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 02/16/2018] [Indexed: 01/03/2023]
Abstract
Apelin is the endogenous ligand of APJ receptor. Both monocytes (MCs) and human umbilical vein endothelial cells (HUVECs) express apelin and APJ, which play important roles in the physiological processes of atherosclerosis. Our previous research indicated that apelin-13 promoted MCs-HUVECs adhesion. Here, we further explore the mechanism responsible for MCs-HUVECs adhesion induced by apelin-13. Apelin-13 promoted reactive oxygen species (ROS) generation and NOX4 expression in HUVECs. Apelin-13 inducedautophagy, increased proteins beclin1 and LC3-II/I expression and induced autophagy flux in HUVECs, which was blocked by NAC, catalase and DPI. Autophagy flux induced by apelin-13 was inhibited by NAC and catalase but not hydroxychloroquine (HCQ). NAC, catalase, and DPI prevented apelin-13 induced ICAM-1 expression in HUVECs. Rapamycin enhanced MCs-HUVECs adhesion that was reversed by NAC, catalase, and DPI. Down-regulation of beclin1 and LC3 by siRNA blocked MCs-HUVECs adhesion. Apelin-13 induced atherosclerotic plaque and increased NOX4, LC3-II/I expression in ApoE-/-(HFD) mouse model. Our results demonstrated that apelin-13 induced MCs-HUVECs adhesion via a ROS-autophagy pathway.
Collapse
Affiliation(s)
- Meiqing Liu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,Department of Pharmacy, The Second People's Hospital of Yunnan province, Kunming, China
| | - Hening Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,Department of Pharmacy, The First People's Hospital of Yueyang, Hunan, China
| | - Qun Zhou
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China.,College of Pharmacy, Hunan University of Medicine, Huaihua, China
| | - Hong Zhao
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Deguan Lv
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Jiangang Cao
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Jinyong Jiang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Mingzhu Tang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Di Wu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Jiaqi Liu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Lele Wu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Haoliang Hu
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Lu He
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Shifang Huang
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Zhe Chen
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Lanfang Li
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Linxi Chen
- Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| |
Collapse
|
42
|
Ou H, Liu C, Feng W, Xiao X, Tang S, Mo Z. Role of AMPK in atherosclerosis via autophagy regulation. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1212-1221. [PMID: 29656339 DOI: 10.1007/s11427-017-9240-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/05/2017] [Indexed: 01/12/2023]
Abstract
Atherosclerosis is characterized by the accumulation of lipids and deposition of fibrous elements in the vascular wall, which is the primary cause of cardiovascular diseases. Adenosine monophosphate-activated protein kinase (AMPK) is a metabolic sensor of energy metabolism that regulates multiple physiological processes, including lipid and glucose metabolism and the normalization of energy imbalances. Overwhelming evidence indicates that AMPK activation markedly attenuates atherosclerosis development. Autophagy inhibits cell apoptosis and inflammation and promotes cholesterol efflux and efferocytosis. Physiological autophagy is essential for maintaining normal cardiovascular function. Increasing evidence demonstrates that autophagy occurs in developing atherosclerotic plaques. Emerging evidence indicates that AMPK regulates autophagy via a downstream signaling pathway. The complex relationship between AMPK and autophagy has attracted the attention of many researchers because of this close relationship to atherosclerosis development. This review demonstrates the role of AMPK and autophagy in atherosclerosis. An improved understanding of this interrelationship will create novel preventive and therapeutic strategies for atherosclerosis.
Collapse
Affiliation(s)
- Hanxiao Ou
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China
| | - Chuhao Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.,2016 Grade Excellent Doctor Class of Medical School, University of South China, Hengyang, 421001, China
| | - Wenjie Feng
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.,2015 Grade Medical Imaging Class of Medical School, University of South China, Hengyang, 421001, China
| | - Xinwen Xiao
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.,2015 Grade Medical Imaging Class of Medical School, University of South China, Hengyang, 421001, China
| | - Shengsong Tang
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China. .,Center for Life Science, Hunan University of Medicine, Huaihua, 418000, China.
| | - Zhongcheng Mo
- Clinical Anatomy & Reproductive Medicine Application Institute, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, 421001, China.
| |
Collapse
|
43
|
Defective Autophagy in Atherosclerosis: To Die or to Senesce? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7687083. [PMID: 29682164 PMCID: PMC5846382 DOI: 10.1155/2018/7687083] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/29/2017] [Accepted: 01/18/2018] [Indexed: 11/17/2022]
Abstract
Autophagy is a subcellular process that plays an important role in the degradation of proteins and damaged organelles such as mitochondria (a process termed “mitophagy”) via lysosomes. It is crucial for regulating protein and mitochondrial quality control and maintaining cellular homeostasis, whereas dysregulation of autophagy has been implicated in a wide range of diseases including atherosclerosis. Recent evidence has shown that the autophagic process becomes dysfunctional during the progression of atherosclerosis, regardless of whether there are many autophagy-stimulating factors (e.g., reactive oxygen species, oxidized lipids, and cytokines) present within the atherosclerotic plaque. This review highlights the recent insights into the causes and consequences of defective autophagy in atherosclerosis, with a special focus on the role of autophagy and mitophagy in plaque macrophages, vascular smooth muscle cells (VSMCs), and endothelial cells (ECs). It has been shown that defective autophagy can promote apoptosis in macrophages but that it accelerates premature senescence in VSMCs. In the ECs, defective autophagy promotes both apoptosis and senescence. We will discuss the discrepancy between these three cell types in their response to autophagy deficiency and underline the cell type-dependent role of autophagy, which may have important implications for the efficacy of autophagy-targeted treatments for atherosclerosis.
Collapse
|
44
|
Swiader A, Nahapetyan H, Faccini J, D'Angelo R, Mucher E, Elbaz M, Boya P, Vindis C. Mitophagy acts as a safeguard mechanism against human vascular smooth muscle cell apoptosis induced by atherogenic lipids. Oncotarget 2018; 7:28821-35. [PMID: 27119505 PMCID: PMC5045359 DOI: 10.18632/oncotarget.8936] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/12/2016] [Indexed: 12/31/2022] Open
Abstract
Mitophagy is a critical cellular process that selectively targets damaged mitochondria for autophagosomal degradation both under baseline conditions and in response to stress preventing oxidative damage and cell death. Recent studies have linked alterations in mitochondria function and reduced autophagy with the development of age-related pathologies. However, the significance of mitochondrial autophagy in vessel wall in response to atherogenic lipid stressors is not known. In the present study, we investigated the role of mitophagy on human vascular smooth muscle cells (VSMC) apoptosis induced by oxidized low-density lipoproteins (LDL). We reported for the first time that the engulfment of defective mitochondria by autophagosomes occurred in human VSMC in response to oxidized LDL. The molecular mechanism mediating mitophagy in human VSMC involved dynamin-related protein 1 (Drp1)-mediated mitochondrial fission, accumulation of PTEN-induced putative kinase 1 (PINK1) and the recruitment of the E3 ubiquitin ligase Parkin to mitochondria. Likewise, we found increased voltage-dependent anion channel 1 (VDAC1) and mitofusin 2 (Mnf2) mitochondrial proteins ubiquitination and LC3 association to mitochondria. Using flow cytometry in the presence of lysosomal inhibitors, we showed that PINK1 and Parkin silencing impaired mitophagy flux and enhanced oxidized LDL-induced VSMC apoptosis. In addition, overexpression of PINK1 and Parkin were protective by limiting cell death. Moreover, reduced Bax levels found in VSMC-overexpressing Parkin indicated cross talk among mitophagy and mitochondrial apoptotic signalling pathways. Altogether these data demonstrate that mitophagy is a safeguard mechanism against human VSMC apoptosis induced by atherogenic stressors and highlight mitophagy as a potential target to stabilize atherosclerotic plaque.
Collapse
Affiliation(s)
- Audrey Swiader
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Hripsime Nahapetyan
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Julien Faccini
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Romina D'Angelo
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Elodie Mucher
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Meyer Elbaz
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Madrid, Spain
| | - Cécile Vindis
- Inserm, UMR-1048, Institute of Metabolic and Cardiovascular Diseases, Toulouse, France.,Toulouse University Paul Sabatier, Toulouse, France
| |
Collapse
|
45
|
Autophagy in health and disease: focus on the cardiovascular system. Essays Biochem 2017; 61:721-732. [PMID: 29233881 DOI: 10.1042/ebc20170022] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/03/2017] [Accepted: 10/16/2017] [Indexed: 12/20/2022]
Abstract
Autophagy is a highly conserved mechanism of lysosome-mediated protein and organelle degradation that plays a crucial role in maintaining cellular homeostasis. In the last few years, specific functions for autophagy have been identified in many tissues and organs. In the cardiovascular system, autophagy appears to be essential to heart and vessel homeostasis and function; however defective or excessive autophagy activity seems to contribute to major cardiovascular disorders including heart failure (HF) or atherosclerosis. Here, we review the current knowledge on the role of cardiovascular autophagy in physiological and pathophysiological conditions.
Collapse
|
46
|
Wang D, Yu W, Liu Y, Zhong G, Zhao Z, Yan X, Liu Q. Roles of Autophagy in Ischemic Heart Diseases and the Modulatory Effects of Chinese Herbal Medicine. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:1401-1419. [PMID: 28946768 DOI: 10.1142/s0192415x17500768] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autophagy is an evolutionarily conserved degradation process which eliminates dysfunctional proteins and cytoplasmic components to maintain homeostasis for cell survival. Increasing evidence has demonstrated the modulatory role of autophagy in ischemic heart diseases (IHDs). Traditionally, this process has been recognized as having protective functions, such as inhibiting atherosclerosis progression and reducing cell death during the ischemic phase. However, recent studies have suggested its dual roles in myocardial ischemia/reperfusion (MIR) injury. Excessive autophagy may play a deleterious role in cardiac function, due to overwhelming clearance of cellular constituents and proteins. Hence modulation of autophagy to increase cardiomyocyte survival and improve cardiac function is meaningful for the treatment of IHD. Chinese herbal medicine, including extractive compounds and patented drugs, has shown its potential role in treating IHD by addressing autophagy-related mechanisms. This review summarizes the updated knowledge on the molecular basis and modulatory role of autophagy in IHD and the recent progress of Chinese herbal medicine in its treatment.
Collapse
Affiliation(s)
- Dawei Wang
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.,† Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510405, China
| | - Weiqing Yu
- ‡ Department of Cardiology, Guangdong Second Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510095, China
| | - Yuntao Liu
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.,† Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510405, China
| | - Guofu Zhong
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Zhen Zhao
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Xia Yan
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.,† Guangdong Provincial Key Laboratory of Research on Emergency in TCM, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510405, China
| | - Qing Liu
- * The Second Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.,§ Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
47
|
Ding S, Jiang J, Yu P, Zhang G, Zhang G, Liu X. Green tea polyphenol treatment attenuates atherosclerosis in high-fat diet-fed apolipoprotein E-knockout mice via alleviating dyslipidemia and up-regulating autophagy. PLoS One 2017; 12:e0181666. [PMID: 28777810 PMCID: PMC5544182 DOI: 10.1371/journal.pone.0181666] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/05/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Green tea polyphenol (GTP) is a polyphenol source from green tea that has drawn wide attention owing to epidemiological evidence of its beneficial effects in the prevention of cardiovascular disease; the underlying molecular mechanisms of these effects are not well understood. This study aimed to investigate the effects of GTP treatment on autophagy regulation in the vessel wall and lipid metabolism of HFD-fed male ApoE-knockout mice. Methods: Adult male ApoE-knockout mice (n = 30) fed with a high-fat diet (HFD) were treated with either vehicle or GTP (3.2 or 6.4 g/L) administered via drinking water for 15 weeks, and C57BL/6J mice fed with standard chow diet (STD) were used as the control group. Metabolic parameters, expression of key mRNAs and proteins of hepatic lipid metabolism and autophagy in the vessel wall of mice were determined after the 15-week treatment. Results: A HFD induced atherosclerosis formation and lipid metabolism disorders as well as reduced autophagy expression in the vessel wall of ApoE-knockout mice, but GTP treatment alleviated the lipid metabolism disorders, decreased the oxLDL levels in serum, and increased the mRNA and protein expressions of hepatic PPARα and autophagy markers (LC3, Beclin1 and p62) in the vessel wall of ApoE-knockout mice. Conclusions: Our findings suggest that GTP supplementation showed marked suppression of atherogenesis through improved lipid metabolism as well as through a direct impact on oxLDL and autophagy flux in the vessel wall.
Collapse
Affiliation(s)
- Shibin Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
- Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
- * E-mail:
| | - Jinjin Jiang
- School of Public Health, Capital Medical University, Beijing, PR, China
| | - Pengxin Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
| | - Guofu Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
| | - Guanghui Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
| | - Xiaoting Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, PR, China
| |
Collapse
|
48
|
Sachdev U, Lotze MT. Perpetual change: autophagy, the endothelium, and response to vascular injury. J Leukoc Biol 2017; 102:221-235. [PMID: 28626046 PMCID: PMC6608075 DOI: 10.1189/jlb.3ru1116-484rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Current studies of vascular health, aging, and autophagy emphasize how the endothelium adapts to stress and contributes to disease. The endothelium is far from an inert barrier to blood-borne cells, pathogens, and chemical signals; rather, it actively translates circulating mediators into tissue responses, changing rapidly in response to physiologic stressors. Macroautophagy-the cellular ingestion of effete organelles and protein aggregates to provide anabolic substrates to fuel bioenergetics in times of stress-plays an important role in endothelial cell homeostasis, vascular remodeling, and disease. These roles include regulating vascular tone, sustaining or limiting cell survival, and contributing to the development of atherosclerosis secondary to infection, inflammation, and angiogenesis. Autophagy modulates these critical functions of the endothelium in a dynamic and perpetual response to tissue and intravascular cues.
Collapse
Affiliation(s)
- Ulka Sachdev
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
49
|
Zhang L, Zhao S, Yuan L, Wu H, Jiang H, Luo G. Hyperoxia-mediated LC3B activation contributes to the impaired transdifferentiation of type II alveolar epithelial cells (AECIIs) to type I cells (AECIs). Clin Exp Pharmacol Physiol 2017; 43:834-43. [PMID: 27187184 DOI: 10.1111/1440-1681.12592] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/05/2016] [Accepted: 05/12/2016] [Indexed: 12/14/2022]
Abstract
Life-saving mechanical ventilation can also cause lung injury through the overproduction of reactive oxygen species (ROS), leading to bronchopulmonary dysplasia (BPD)-like symptoms in preterm infants. It is reported that the autophagic protein microtubule-associated protein-1 light chain (LC)-3B can confer protection against hyperoxia-induced DNA damage in lung alveolar epithelium. However, its role in the transdifferentiation of type II alveolar epithelial cells (AECIIs) to type I cells (AECIs) is unclear and requires further investigation. In this study, newborn Sprague-Dawley rats were exposed to 90% oxygen for up to 14 days to mimic BPD in human infants, with neonatal pups exposed to room air (21% oxygen) as controls. Primary rat AECIIs were cultured under hyperoxic conditions for up to 24 hours to further investigate the underlying mechanisms. This study found that hyperoxia promoted a significant and time-dependent increase of AECII marker surfactant protein (SP)-C in the lung. The increase of AECI marker T1α was repressed by hyperoxia during lung development. These results indicated an impaired AECII transdifferentiation. Pulmonary ROS concentration and expression of autophagic protein LC-3B were increased gradually in response to hyperoxia exposure. Furthermore, AECIIs produced more ROS when cultured under hyperoxic conditions in vitro. Both the LC3B expression and the conversion from LC3BI to LC3BII were enhanced in hyperoxic AECs. Interestingly, inhibition of LC3B either by ROS inhibitor N-acetyl-l-cysteine (NAC) or adenovirus-mediated LC3B shRNA could partly restore AECII transdifferentiation under hyperoxia condition. In summary, the current study reveals a novel role of activated LC3B induced by hyperoxia in AECII transdifferentiation.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Zhao
- Department of Paediatrics, Shenyang Fourth People's Hospital, Shenyang, China
| | - Lijie Yuan
- Department of Biochemistry and Molecular Biology, Harbin Medical University (Daqing Campus), Daqing, China
| | - Hongmin Wu
- Department of Neonatology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hong Jiang
- Department of Paediatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Gang Luo
- Department of Paediatrics, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Role of autophagy in advanced atherosclerosis. Mol Med Rep 2017; 15:2903-2908. [DOI: 10.3892/mmr.2017.6403] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 02/15/2017] [Indexed: 11/05/2022] Open
|