1
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
2
|
Chen H, Teng D, Xu B, Wang C, Wang H, Jia W, Gong L, Dong H, Zhong L, Yang J. The SGLT2 Inhibitor Canagliflozin Reduces Atherosclerosis by Enhancing Macrophage Autophagy. J Cardiovasc Transl Res 2023; 16:999-1009. [PMID: 37126209 DOI: 10.1007/s12265-023-10390-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/23/2023] [Indexed: 05/02/2023]
Abstract
It has been shown that SGLT2 suppresses atherosclerosis (AS). Recent studies indicate that autophagy widely participates in atherogenesis. This study aimed to assess the effect of canagliflozin (CAN) on atherogenesis via autophagy. Macrophages and ApoE - / - mice were used in this study. In macrophages, the results showed that CAN promoted LC3II expression and autophagosome formation. Furthermore, the cholesterol efflux assay demonstrated that CAN enhanced cholesterol efflux from macrophages via autophagy, resulting in lower lipid droplet concentrations in macrophages. The western blot revealed that CAN regulated autophagy via the AMPK/ULK1/Beclin1 signaling pathway. CAN resulted in increased macrophage autophagy in atherosclerotic plaques of ApoE - / - mice, confirming that CAN could inhibit the progression of AS via promoting macrophage autophagy. The current study found that CAN reduced the production of atherosclerotic lesions, which adds to our understanding of how SGLT2 inhibitors function to delay the progression of AS.
Collapse
Affiliation(s)
- Hongping Chen
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Da Teng
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Bowen Xu
- Binzhou Medical University, Yantai, Shandong Province, China
| | - Chunxiao Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Hua Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Wenjuan Jia
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Lei Gong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Haibin Dong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China
| | - Lin Zhong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China.
| | - Jun Yang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Shandong Province, China.
| |
Collapse
|
3
|
Wang P, Li M, Gao T, Fan J, Zhang D, Zhao Y, Zhao Y, Wang Y, Guo T, Gao X, Liu Y, Gao Y, Guan X, Sun X, Zhao J, Li H, Yang L. Vascular Electrical Stimulation with Wireless, Battery-Free, and Fully Implantable Features Reduces Atherosclerotic Plaque Formation Through Sirt1-Mediated Autophagy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300584. [PMID: 37267941 DOI: 10.1002/smll.202300584] [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: 01/19/2023] [Revised: 05/18/2023] [Indexed: 06/04/2023]
Abstract
Electrical stimulation (ES) is a safe and effective procedure in clinical rehabilitation with few adverse effects. However, studies on ES for atherosclerosis (AS) are scarce because ES does not provide a long-term intervention for chronic disease processes. Battery-free implants and surgically mounted them in the abdominal aorta of high-fat-fed Apolipoprotein E (ApoE-/- ) mice are used, which are electrically stimulated for four weeks using a wireless ES device to observe changes in atherosclerotic plaques. Results showed that there is almost no growth of atherosclerotic plaque at the stimulated site in AopE-/- mice after ES. RNA-sequencing (RNA-seq) analysis of Thp-1 macrophages reveal that the transcriptional activity of autophagy-related genes increase substantially after ES. Additionally, ES reduces lipid accumulation in macrophages by restoring ABCA1- and ABCG1-mediated cholesterol efflux. Mechanistically, it is demonstrated that ES reduced lipid accumulation through Sirtuin 1 (Sirt1)/Autophagy related 5 (Atg5) pathway-mediated autophagy. Furthermore, ES reverse autophagic dysfunction in macrophages of AopE-/- mouse plaques by restoring Sirt1, blunting P62 accumulation, and inhibiting the secretion of interleukin (IL)-6, resulting in the alleviation of atherosclerotic lesion formation. Here, a novel approach is shown in which ES can be used as a promising therapeutic strategy for AS treatment through Sirt1/Atg5 pathway-mediated autophagy.
Collapse
Affiliation(s)
- Pengyu Wang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Manman Li
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Tielei Gao
- Department of Forensic Medicine, Harbin Medical University, Harbin, 150081, P. R. China
| | - Jiaying Fan
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Dengfeng Zhang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Ying Zhao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yajie Zhao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yuqin Wang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Tianwei Guo
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Xi Gao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yujun Liu
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Yang Gao
- Department of Pathology, Harbin Medical University-Daqing, Daqing, 163319, P. R. China
| | - Xue Guan
- School of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, P. R. China
| | - Xinyong Sun
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Jiyi Zhao
- Cardiovascular Care Unit, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Hong Li
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
| | - Liming Yang
- School of Basic Medical Sciences, Harbin Medical University, Harbin, 150081, P. R. China
- School of Basic Medical Sciences, Harbin Medical University-Daqing, Daqing, 163319, P. R. China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150081, P. R. China
| |
Collapse
|
4
|
Liu J, Chen Y, Chen H, Wang Y, Li D, Zhang Q, Chai L, Qiu Y, Zhang J, Shen N, Wang Q, Wang J, Li M. Macrophage migration inhibitory factor exacerbates asthmatic airway remodeling via dynamin-related protein 1-mediated autophagy activation. Respir Res 2023; 24:216. [PMID: 37674165 PMCID: PMC10481618 DOI: 10.1186/s12931-023-02526-y] [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: 06/07/2023] [Accepted: 08/31/2023] [Indexed: 09/08/2023] Open
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF) and GTPase dynamin-related protein 1 (Drp1)-dependent aberrant mitochondrial fission are closely linked to the pathogenesis of asthma. However, it is unclear whether Drp1-mediated mitochondrial fission and its downstream targets mediate MIF-induced proliferation of airway smooth muscle cells (ASMCs) in vitro and airway remodeling in chronic asthma models. The present study aims to clarify these issues. METHODS In this study, primary cultured ASMCs and ovalbumin (OVA)-induced asthmatic rats were applied. Cell proliferation was detected by CCK-8 and EdU assays. Western blotting was used to detect extracellular signal-regulated kinase (ERK) 1/2, Drp1, autophagy-related markers and E-cadherin protein phosphorylation and expression. Inflammatory cytokines production, airway reactivity test, histological staining and immunohistochemical staining were conducted to evaluate the development of asthma. Transmission electron microscopy was used to observe the mitochondrial ultrastructure. RESULTS In primary cultured ASMCs, MIF increased the phosphorylation level of Drp1 at the Ser616 site through activation of the ERK1/2 signaling pathway, which further activated autophagy and reduced E-cadherin expression, ultimately leading to ASMCs proliferation. In OVA-induced asthmatic rats, MIF inhibitor 4-iodo-6-phenylpyrimidine (4-IPP) treatment, suppression of mitochondrial fission by Mdivi-1 or inhibiting autophagy with chloroquine phosphate (CQ) all attenuated the development of airway remodeling. CONCLUSIONS The present study provides novel insights that MIF promotes airway remodeling in asthma by activating autophagy and degradation of E-cadherin via ERK/Drp1 signaling pathway, suggesting that targeting MIF/ERK/Drp1 might have potential therapeutic value for the prevention and treatment of asthma.
Collapse
Affiliation(s)
- Jin Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuqian Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, People's Republic of China.
| |
Collapse
|
5
|
Chen H, Zhang L, Mi S, Wang H, Wang C, Jia W, Gong L, Dong H, Xu B, Jing Y, Ge P, Pei Z, Zhong L, Yang J. FURIN suppresses the progression of atherosclerosis by promoting macrophage autophagy. FASEB J 2023; 37:e22933. [PMID: 37093709 DOI: 10.1096/fj.202201762rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/20/2023] [Accepted: 04/10/2023] [Indexed: 04/25/2023]
Abstract
FURIN, a member of the mammalian proprotein convertases (PCs) family, can promote the proteolytic maturation of proproteins. It has been shown that FURIN plays an important role in the progression of atherosclerosis (AS). Current evidence indicates that autophagy widely participates in atherogenesis. This study aimed to explore whether FURIN could affect atherogenesis via autophagy. The effect of FURIN on autophagy was studied using aortic tissues from aortic dissection patients who had BENTALL surgery, as well as macrophages and ApoE-/- mice. In atherosclerotic plaques of aortic tissues from patients, FURIN expression and autophagy were elevated. In macrophages, FURIN-shRNA and FURIN-overexpression lentivirus were used to intervene in FURIN expression. The results showed that FURIN overexpression accelerated LC3 formation in macrophages during the autophagosome formation phase. Furthermore, FURIN-induced autophagy resulted in lower lipid droplet concentrations in macrophages. The western blot revealed that FURIN regulated autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway. In vivo, FURIN overexpression resulted in increased macrophage LC3 formation in ApoE-/- mice atherosclerotic plaques, confirming that FURIN could inhibit the progression of AS by promoting macrophage autophagy. The present study demonstrated that FURIN suppressed the progression of AS by promoting macrophage autophagy via the AMPK/mTOR/ULK1/PI3KIII signaling pathway, which attenuated atherosclerotic lesion formation. Based on this data, current findings add to our understanding of the complexity of AS.
Collapse
Affiliation(s)
- Hongping Chen
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lihui Zhang
- School of Medicine, Qingdao University, Qingdao, China
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Shaohua Mi
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Hua Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Chunxiao Wang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Wenjuan Jia
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Lei Gong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Haibin Dong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Bowen Xu
- The 2nd Medical Colloge, Binzhou Medical University, Yantai, China
| | - Yanyan Jing
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Peipei Ge
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Zhigang Pei
- Department of Vascular Surgery, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, China
| | - Lin Zhong
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| | - Jun Yang
- Department of Cardiology, Yuhuangding Hospital, The Fourth School of Clinical Medicine of Qingdao University, Yantai, Yantai, China
| |
Collapse
|
6
|
Deng Y, Tu Y, Yang X, Liao X, Xia Z, Liao W. Anti-atherosclerosis effect of nobiletin via PINK1/Parkin-mediated mitophagy and NLRP3 inflammasome signaling pathway. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
7
|
Zhang F, Wang R, Liu B, Zhang L. A bibliometric analysis of autophagy in atherosclerosis from 2012 to 2021. Front Pharmacol 2022; 13:977870. [PMID: 36188570 PMCID: PMC9520361 DOI: 10.3389/fphar.2022.977870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
Abstract
Background: Regulation of autophagy affects the progression of atherosclerosis. In recent years, research on autophagy in atherosclerosis has been widely concerned. However, there is no bibliometric analysis in this field. Objective: The purpose of this study was to explore the general situation, hot spots, and trends of the research in this field through bibliometric analysis. Methods: Articles related to autophagy in atherosclerosis from 2012 to 2021 were retrieved from the Web of Science Core Collection. VOSviewer and CiteSpace were used for data analysis and visualization of countries, institutions, authors, keywords, journals, and citations. Results: A total of 988 articles were obtained in the last 10 years. The number of publications and citations increased rapidly from 2012 to 2021, especially after 2019. The most productive countries, institutions, journals, and authors were the People’s Republic of China, Shandong University, Arteriosclerosis Thrombosis and Vascular Biology, and Wim Martinet, respectively. The primary keywords were “oxidative stress,” “apoptosis,” “activated protein kinase,” and “inflammation.” The burst detection analysis of keywords found that “SIRT1” and “long non-coding RNA” might be regarded as the focus of future research. Conclusion: This is the first bibliometric analysis of autophagy in atherosclerosis, which reports the hot spots and emerging trends. The interaction between oxidative stress and autophagy, programmed cell death, and activated protein kinases are considered to be the current research priorities. Molecular mechanisms and therapeutic target for the intervention of atherosclerosis by regulating autophagy will become an emerging research direction.
Collapse
Affiliation(s)
| | | | | | - Lei Zhang
- *Correspondence: Baocheng Liu, ; Lei Zhang,
| |
Collapse
|
8
|
miR-99a-5p: A Potential New Therapy for Atherosclerosis by Targeting mTOR and Then Inhibiting NLRP3 Inflammasome Activation and Promoting Macrophage Autophagy. DISEASE MARKERS 2022; 2022:7172583. [PMID: 35968506 PMCID: PMC9374553 DOI: 10.1155/2022/7172583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022]
Abstract
Objective MicroRNAs have been revealed to be involved in the development of atherosclerosis. The present study is aimed at exploring the potential of miR-99a-5p as a therapy for atherosclerosis. We suspected that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy via constraining mTOR, therefore, alleviating atherosclerosis. Methods The cell viability in ox-LDL-induced THP-1 macrophages was assessed by CCK-8 assay. Bioinformatic analysis was used to predict the target genes of miR-99a-5p. The binding between miR-99a-5p and mTOR was confirmed by luciferase reporter assay. In vivo, a high-fat-diet-induced atherosclerosis model was established in apolipoprotein E knockout mice. Hematoxylin-eosin, oil red O, and Sirius red staining were performed for the determination of atherosclerotic lesions. MTOR and associated protein levels were detected by Western blot analysis. Results miR-99a-5p inhibited NLRP3 inflammasome activation and promoted macrophage autophagy by targeting mTOR. Enforced miR-99a-5p significantly reduced the levels of inflammasome complex and inflammatory cytokines. Furthermore, miR-99a-5p overexpression inhibited the expression of mTOR, whereas mTOR overexpression reversed the trend of the above behaviors. In vivo, the specific overexpression of miR-99a-5p significantly reduced atherosclerotic lesions, accompanied by a significant downregulation of autophagy marker CD68 protein expression. Conclusion We demonstrated for the first time that miR-99a-5p may be considered a therapy for atherosclerosis. The present study has revealed that miR-99a-5p might inhibit NLRP3 inflammasome activation and promote macrophage autophagy by targeting mTOR, therefore, alleviating atherosclerosis.
Collapse
|
9
|
Uyy E, Suica VI, Boteanu RM, Cerveanu-Hogas A, Ivan L, Hansen R, Antohe F. Regulated cell death joins in atherosclerotic plaque silent progression. Sci Rep 2022; 12:2814. [PMID: 35181730 PMCID: PMC8857202 DOI: 10.1038/s41598-022-06762-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 01/31/2022] [Indexed: 01/01/2023] Open
Abstract
Non-apoptotic regulated cell death (ferroptosis and necroptosis) leads to the release of damage-associated molecular patterns (DAMPs), which initiate and perpetuate a non-infectious inflammatory response. We hypothesize that DAMPs and non-apoptotic regulated cell death are critical players of atherosclerotic plaque progression with inadequate response to lipid-lowering treatment. We aimed to uncover the silent mechanisms that govern the existing residual risk of cardiovascular-related mortality in experimental atherosclerosis. Proteomic and genomic approaches were applied on the ascending aorta of hyperlipidemic rabbits and controls with and without lipid-lowering treatment. The hyperlipidemic animals, which presented numerous heterogeneous atherosclerotic lesions, exhibited high concentrations of serum lipids and increased lipid peroxidation oxidative stress markers. The analyses revealed the significant upregulation of DAMPs and proteins implicated in ferroptosis and necroptosis by hyperlipidemia. Some of them did not respond to lipid-lowering treatment. Dysregulation of five proteins involved in non-apoptotic regulated cell death proteins (VDAC1, VDAC3, FTL, TF and PCBP1) and nine associated DAMPs (HSP90AA1, HSP90AB1, ANXA1, LGALS3, HSP90B1, S100A11, FN, CALR, H3-3A) was not corrected by the treatment. These proteins could play a key role in the atherosclerotic silent evolution and may possess an unexplored therapeutic potential. Mass spectrometry data are available via ProteomeXchange with identifier PXD026379.
Collapse
Affiliation(s)
- Elena Uyy
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania
| | - Viorel I Suica
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania
| | - Raluca M Boteanu
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania
| | - Aurel Cerveanu-Hogas
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania
| | - Luminita Ivan
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania
| | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway.,Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Felicia Antohe
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, P.O. Box 35-14, 050568, Bucharest, Romania.
| |
Collapse
|
10
|
Fang S, Sun S, Cai H, Zou X, Wang S, Hao X, Wan X, Tian J, Li Z, He Z, Huang W, Liang C, Zhang Z, Yang L, Tian J, Yu B, Sun B. IRGM/Irgm1 facilitates macrophage apoptosis through ROS generation and MAPK signal transduction: Irgm1 +/- mice display increases atherosclerotic plaque stability. Theranostics 2021; 11:9358-9375. [PMID: 34646375 PMCID: PMC8490524 DOI: 10.7150/thno.62797] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 09/02/2021] [Indexed: 02/04/2023] Open
Abstract
Rationale: Atherosclerosis plaque rupture (PR) is the pathological basis and chief culprit of most acute cardiovascular events and death. Given the complex and important role of macrophage apoptosis and autophagy in affecting plaque stability, an important unanswered question include is whether, and how, immunity-related GTPase family M protein (IRGM) and its mouse orthologue IRGM1 affect macrophage survival and atherosclerotic plaque stability. Methods: To investigate whether serum IRGM of ST-segment elevation myocardial infarction (STEMI) patients is related to plaque morphology, we divided 85 STEMI patients into those with and without plaque rupture (PR and non-PR, respectively) based on OCT image analysis, and quantified the patients' serum IRGM levels. Next, we engineered Irgm1 deficient mice (Irgm1+/-) and chimera mice with Irgm1 deficiency in the bone marrow on an ApoE-/- background, which were then fed a high-fat diet for 16 weeks. Pathological staining was used to detect necrotic plaque cores, ratios of neutral lipids and cholesterol crystal, as well as collagen fiber contents in these mice to characterize plaque stability. In addition, immunofluorescence, immunohistochemical staining and western blot were used to detect the apoptosis of macrophages in the plaques. In vitro, THP-1 and RAW264.7 cells were stimulated with ox-LDL to mimic the in vivo environment, and IRGM/IRGM1 expression were modified by specific siRNA (knockdown) or IRGM plasmid (knocked-in). The effect of IRGM/Irgm1 on autophagy and apoptosis of macrophages induced by ox-LDL was then evaluated. In addition, we introduced inhibitors of the JNK/p38/ERK signaling pathway to verify the specific mechanism by which Irgm1 regulates RAW264.7 cell apoptosis. Results: The serum IRGM levels of PR patients is significantly higher than that of non-PR patients and healthy volunteers, which may be an effective predictor of PR. On a high-fat diet, Irgm1-deficient mice exhibit reduced necrotic plaque cores, as well as neutral lipid and cholesterol crystal ratios, with increased collagen fiber content. Additionally, macrophage apoptosis is inhibited in the plaques of Irgm1-deficient mice. In vitro, IRGM/Irgm1 deficiency rapidly inhibits ox-LDL-induced macrophage autophagy while inhibiting ox-LDL-induced macrophage apoptosis in late stages. Additionally, IRGM/Irgm1 deficiency suppresses reactive oxygen species (ROS) production in macrophages, while removal of ROS effectively inhibits macrophage apoptosis induced by IRGM overexpression. We further show that Irgm1 can affect macrophage apoptosis by regulating JNK/p38/ERK phosphorylation in the MAPK signaling pathway. Conclusions: Serum IRGM may be related to the process of PR in STEMI patients, and IRGM/Irgm1 deficiency increases plaque stability. In addition, IRGM/Irgm1 deficiency suppresses macrophage apoptosis by inhibiting ROS generation and MAPK signaling transduction. Cumulatively, these results suggest that targeting IRGM may represent a new treatment strategy for the prevention and treatment of acute cardiovascular deaths caused by PR.
Collapse
|
11
|
Li R, Wang F, Wei J, Lin Y, Tang G, Rao L, Ma L, Xu Q, Wu J, Lv Q, Zhou R, Lei H, Zhao X, Yao D, Xiao B, Huang H, Zhang J, Mo B. The Role of Macrophage Migration Inhibitory Factor (MIF) in Asthmatic Airway Remodeling. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:88-105. [PMID: 33191679 PMCID: PMC7680835 DOI: 10.4168/aair.2021.13.1.88] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/10/2020] [Accepted: 05/17/2020] [Indexed: 12/23/2022]
Abstract
Purpose Recent studies have demonstrated that macrophage migration inhibitory factor (MIF) is of importance in asthmatic inflammation. The role of MIF in modulating airway remodeling has not yet been thoroughly elucidated to date. In the present study, we hypothesized that MIF promoted airway remodeling by intensifying airway smooth muscle cell (ASMC) autophagy and explored the specific mechanisms. Methods MIF knockdown in the lung tissues of C57BL/6 mice was conducted by instilling intratracheally adeno-associated virus (AAV) vectors (MIF-mutant AAV9) into mouse lung tissues. Mice genetically deficient in the autophagy marker ATG5 (ATG5+/−) was used to detect the role of autophagy in ovalbumin (OVA)-asthmatic murine models. Moreover, to block the expression of MIF and CD74 in vitro models, inhibitors, antibodies and lentivirus transfection techniques were employed. Results First, MIF knockdown in the lung tissues of mice showed markedly reduced airway remodeling in OVA murine mice models. Secondly, ASMC autophagy was increased in the OVA-challenged models. Mice genetically deficient in the autophagy marker ATG5 (ATG5+/−) that were primed and challenged with OVA showed lower airway remodeling than genetically wild-type asthmatic mice. Thirdly, MIF can induce ASMC autophagy in vitro. Moreover, the cellular source of MIF which promoted ASMC autophagy was macrophages. Finally, MIF promoted ASMC autophagy in a CD74-dependent manner. Conclusions MIF can increase asthmatic airway remodeling by enhancing ASMC autophagy. Macrophage-derived MIF can promote ASMC autophagy by targeting CD74.
Collapse
Affiliation(s)
- Ruyi Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China
| | - Feiyun Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghong Wei
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yun Lin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Guofang Tang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lizong Rao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qing Xu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jingjie Wu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Qian Lv
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Rui Zhou
- Department of Respiratory and Critical Care Medicine, The Second Xiangya Hospital, Changsha, China
| | - Huiren Lei
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Xueqiang Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Dong Yao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Bo Xiao
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Haiming Huang
- Laboratory of Pulmonary Diseases, Guilin Medical University, Guilin, China
| | - Jiange Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.
| |
Collapse
|
12
|
Xu YL, Liu XY, Cheng SB, He PK, Hong MK, Chen YY, Zhou FH, Jia YH. Geniposide Enhances Macrophage Autophagy through Downregulation of TREM2 in Atherosclerosis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1821-1840. [DOI: 10.1142/s0192415x20500913] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Macrophage autophagy defect is closely related to the progression of atherosclerosis (AS) and is regulated by the triggering receptor expressed on myeloid cell 2 (TREM2). TREM2 is a key factor in the development of Alzheimer’s disease (AD), the deficiency of which leads to anomalous autophagy in microglia. However, the role of TREM2 in the autophagy of plaque macrophages is still unclear. Geniposide (GP) can inhibit AS progression and enhance macrophage autophagy, although the underlying mechanisms remain unknown. We found that high-fat diet (HFD) feeding significantly increased TREM2 levels and inhibited autophagy in the macrophages of ApoE[Formula: see text] mice. TREM2 overexpression in RAW264.7 macrophages decreased autophagy via activation of mTOR signaling. GP inhibited the progression of AS in ApoE[Formula: see text] mice, reinforced macrophage autophagy, and downregulated TREM2 by inhibiting mTOR signaling. Taken together, augmenting the autophagy levels in plaque macrophages by inhibiting the TREM2/mTOR axis can potentially impede atherosclerotic progression. The promising therapeutic effects of GP seen in this study should be validated in future trials, and the underlying mechanisms have to be elucidated in greater detail.
Collapse
Affiliation(s)
- Yu-Ling Xu
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Xiao-Yu Liu
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Sai-Bo Cheng
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Pei-Kun He
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Mu-Keng Hong
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Yu-Yao Chen
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Feng-Hua Zhou
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| | - Yu-Hua Jia
- School of Traditional Chinese Medicine, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
13
|
Autophagy, Hyperlipidemia, and Atherosclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:237-264. [PMID: 32671753 DOI: 10.1007/978-981-15-4272-5_18] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily conserved process in eukaryotes that processes the turnover of intracellular substances. Atherosclerosis is a disease caused by multiple factors, it mainly occurs on the walls of large and medium blood vessels and atherosclerotic plaques form in the intima of the blood vessels. Hyperlipidemia is considered to be a very dangerous factor leading to cardiovascular and cerebrovascular diseases, especially atherosclerosis. This chapter mainly introduces the key role of autophagy in hyperlipidemia and atherosclerosis, that is, impaired lipophagy affects the degradation of triacylglycerol, cholesterol, etc., leading to hyperlipidemia in atherosclerosis. In patients, excessive levels of autophagy accelerate the rupture of atherosclerotic plaque. This chapter also describes the advances in the treatment of atherosclerosis and hyperlipidemia by targeted autophagy.
Collapse
|
14
|
Bai J, Zhang Y, Fan Q, Xu J, Shan H, Gao X, Ma Q, Sheng L, Zheng X, Cheng W, Li D, Zhang M, Hao Y, Feng L, Chen Q, Zhou X, Wang C. Reactive Oxygen Species-Scavenging Scaffold with Rapamycin for Treatment of Intervertebral Disk Degeneration. Adv Healthc Mater 2020; 9:e1901186. [PMID: 31820852 DOI: 10.1002/adhm.201901186] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/10/2019] [Indexed: 12/15/2022]
Abstract
The chronic inflammatory microenvironment is characterized by the elevated level of reactive oxygen species (ROS). Here, it is hypothesized that developing an ROS-scavenging scaffold loaded with rapamycin (Rapa@Gel) may offer a new strategy for modulating the local inflammatory microenvironment to improve intervertebral disk tissue regeneration. The therapeutic scaffold consisting of ROS-degradable hydrogel can be injected into the injured degeneration site of intervertebral disk (IVD) and can release therapeutics in a programmed manner. The ROS scavenged by scaffold reduces the inflammatory responses. It is found that when rats are treated with Rapa@Gel, this results in an increase in the percentage of M2-like macrophages and a decrease in M1-like macrophages in the inflammatory environment, respectively. Regeneration of IVD is achieved by Rapa@Gel local treatment, due to the increased M2 macrophages and reduced inflammation. This strategy may be extended to the treatment of many other inflammatory diseases.
Collapse
Affiliation(s)
- Jinyu Bai
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Yingzi Zhang
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Qin Fan
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Jialu Xu
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Huajian Shan
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Xiang Gao
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Qingle Ma
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Lei Sheng
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Xin Zheng
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Weinan Cheng
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Dazhuang Li
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Mingchao Zhang
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Yu Hao
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Liangzhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| | - Xiaozhong Zhou
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow University Suzhou Jiangsu 215004 China
| | - Chao Wang
- Institute of Functional Nano & Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐based Functional Materials and DevicesSoochow University Suzhou Jiangsu 215123 China
| |
Collapse
|
15
|
Martinet W, Coornaert I, Puylaert P, De Meyer GRY. Macrophage Death as a Pharmacological Target in Atherosclerosis. Front Pharmacol 2019; 10:306. [PMID: 31019462 PMCID: PMC6458279 DOI: 10.3389/fphar.2019.00306] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disorder characterized by the gradual build-up of plaques within the vessel wall of middle-sized and large arteries. Over the past decades, treatment of atherosclerosis mainly focused on lowering lipid levels, which can be accomplished by the use of statins. However, some patients do not respond sufficiently to statin therapy and therefore still have a residual cardiovascular risk. This issue highlights the need for novel therapeutic strategies. As macrophages are implicated in all stages of atherosclerotic lesion development, they represent an important alternative drug target. A variety of anti-inflammatory strategies have recently emerged to treat or prevent atherosclerosis. Here, we review the canonical mechanisms of macrophage death and their impact on atherogenesis and plaque stability. Macrophage death is a prominent feature of advanced plaques and is a major contributor to necrotic core formation and plaque destabilization. Mechanisms of macrophage death in atherosclerosis include apoptosis, passive or accidental necrosis as well as secondary necrosis, a type of death that typically occurs when apoptotic cells are insufficiently cleared by neighboring cells via a phagocytic process termed efferocytosis. In addition, less-well characterized types of regulated necrosis in macrophages such as necroptosis, pyroptosis, ferroptosis, and parthanatos may occur in advanced plaques and are also discussed. Autophagy in plaque macrophages is an important survival pathway that protects against cell death, yet massive stimulation of autophagy promotes another type of death, usually referred to as autosis. Multiple lines of evidence indicate that a better insight into the different mechanisms of macrophage death, and how they mutually interact, will provide novel pharmacological strategies to resolve atherosclerosis and stabilize vulnerable, rupture-prone plaques.
Collapse
Affiliation(s)
- Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Isabelle Coornaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
16
|
Hawthorn Extract Alleviates Atherosclerosis through Regulating Inflammation and Apoptosis Related Factors: An Experimental Study. Chin J Integr Med 2018; 25:108-115. [DOI: 10.1007/s11655-018-3020-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2018] [Indexed: 10/27/2022]
|
17
|
Chang H, Yuan W, Wu H, Yin X, Xuan H. Bioactive components and mechanisms of Chinese poplar propolis alleviates oxidized low-density lipoprotein-induced endothelial cells injury. Altern Ther Health Med 2018; 18:142. [PMID: 29724195 PMCID: PMC5934819 DOI: 10.1186/s12906-018-2215-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/20/2018] [Indexed: 01/21/2023]
Abstract
Background Propolis, a polyphenol-rich natural product, has been used as a functional food in anti-inflammation. However, its bioactive components and mechanisms have not been fully elucidated. To discover the bioactive components and anti-inflammatory mechanism, we prepared and separated 8 subfractions from ethyl acetate extract of Chinese propolis (EACP) and investigated the mechanism in oxidized low density lipoprotein (ox-LDL) induced human umbilical vein endothelial cells (HUVECs) damage. Methods Eight subfractions were prepared and separated from ethyl acetate extract of Chinese propolis (EACP) with different concentrations of methanol-water solution, and analysed its chemical constituents by HPLC-DAD/Q-TOF-MS. Then 80% confluent HUVECs were stimulated with 40 μg/mL ox-LDL. Cell viability and apoptosis were evaluated by Sulforhodamine B (SRB) assay and Hoechst 33,258 staining, respectively. Levels of caspase 3, PARP, LC3B, p62, p-mTOR, p-p70S6K, p-PI3K, p-Akt, LOX-1 and p-p38 MAPK were assessed by western blotting and immunofluorescence assay, respectively. Reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were measured with fluorescent probes. Results Each subfraction exhibited similar protective effect although the contents of chemical constituents were different. EACP attenuated ox-LDL induced HUVECs apoptosis, depressed the ratio of LC3-II/LC3-I and enhanced the p62 level. In addition, treatment with EACP also activated the phosphorylation of PI3K/Akt/mTOR, and deactivated the level of LOX-1 and phosphorylation of p38 MAPK. The overproduction of ROS and the damage of MMP were also ameliorated after ECAP treatment. Conclusions These findings indicated that the bioactive component of propolis on anti-inflammatory activity was not determined by a single constituent, but a complex interaction including flavonoids, esters and phenolic acids. EACP attenuated ox-LDL induced HUVECs injury by inhibiting LOX-1 level and depressed ROS production against oxidative stress in ox-LDL induced HUVECs, further to activate PI3K/Akt/mTOR pathway and deactivate p38 MAPK to inhibit apoptosis and autophagy, which provide novel insights into the potential application of propolis on modulating chronic inflammation.
Collapse
|
18
|
Ko JH, Yoon SO, Lee HJ, Oh JY. Rapamycin regulates macrophage activation by inhibiting NLRP3 inflammasome-p38 MAPK-NFκB pathways in autophagy- and p62-dependent manners. Oncotarget 2018; 8:40817-40831. [PMID: 28489580 PMCID: PMC5522223 DOI: 10.18632/oncotarget.17256] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 04/11/2017] [Indexed: 01/07/2023] Open
Abstract
Excessive and prolonged activation of macrophages underlies many inflammatory and autoimmune diseases. To regulate activation and maintain homeostasis, macrophages have multiple intrinsic mechanisms, one of which is modulation through autophagy. Here we demonstrate that autophagy induction by rapamycin suppressed the production of IL-1β and IL-18 in lipopolysaccharide- and adenosine triphosphate-activated macrophages at the post-transcriptional level by eliminating mitochondrial ROS (mtROS) and pro-IL1β in a p62/SQSTM1-dependent manner. In addition, rapamycin activated Nrf2 through up-regulation of p62/SQSTM1, which further contributed to the reduction of mtROS. Reduced IL-1β subsequently diminished the activation of p38 MAPK-NFκB pathways, leading to transcriptional down-regulation of IL-6, IL-8, MCP-1, and IκBα in rapamycin-treated macrophages. Therefore, our results suggest that rapamycin negatively regulates macrophage activation by restricting a feedback loop of NLRP3 inflammasome-p38 MAPK-NFκB pathways in autophagy- and p62/SQSTM1-dependent manners.
Collapse
Affiliation(s)
- Jung Hwa Ko
- Department of Ophthalmology, Seoul National University Hospital, 03080, Seoul, Korea.,Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 03080, Seoul, Korea
| | - Sun-Ok Yoon
- R and D Laboratory, Eutilex Co., Ltd, 08594, Seoul, Korea
| | - Hyun Ju Lee
- Department of Ophthalmology, Seoul National University Hospital, 03080, Seoul, Korea.,Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 03080, Seoul, Korea
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University Hospital, 03080, Seoul, Korea.,Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 03080, Seoul, Korea
| |
Collapse
|
19
|
Tang F, Yang TL, Zhang Z, Li XG, Zhong QQ, Zhao TT, Gong L. MicroRNA-21 suppresses ox-LDL-induced human aortic endothelial cells injuries in atherosclerosis through enhancement of autophagic flux: Involvement in promotion of lysosomal function. Exp Cell Res 2017; 359:374-383. [PMID: 28823833 DOI: 10.1016/j.yexcr.2017.08.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/02/2017] [Accepted: 08/13/2017] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is a common pathological basis of cardiovascular disease and remains the leading cause of mortality. Endothelial cell (EC) injury and autophagy dysfunction have been proved to contribute to the development of atherosclerosis. Recently, accumulating evidence confirms that microRNAs (miRNAs) have emerged as vital regulators and fine-tuners of various pathophysiological cellular impacts and molecular signaling pathways involved in atherosclerosis. Herein, the objective of the present study was to explore the biological function of miR-21 in oxidized low-density lipoprotein (ox-LDL)-induced human aortic endothelial cells (HAECs) injury and the underlying molecular mechanism. The results showed that ox-LDL treatment significantly decreased HAECs viability, increased caspase-3 activity, apoptosis ratio and Bax protein expression, and reduced Bcl-2 protein expression resulting in EC injuries. Simultaneously, ox-LDL treatment obviously reduced miR-21 level in a time-and dose-dependent manner. Notably, ox-LDL-induced EC injuries were abolished by miR-21 mimics transfection. In addition, miR-21 mimics alleviated ox-LDL-induced impaired autophagic flux as illustrated by the increases in LC3-II/LC3-I ratio and Beclin-1 protein expression, and the decrease in p62 protein expression in HAECs. Moreover, ox-LDL suppressed the expressions of lysosomal membrane protein (LAMP1) and cathepsin D proteins, and attenuated cathepsin D activity in HAECs, leading to lysosomal dysfunction, while these effects were also blocked by miR-21 mimics. These findings indicated that miR-21 restored impaired autophagic flux and lysosomal dysfunction, thereby attenuating ox-LDL-induced HAECs injuries.
Collapse
Affiliation(s)
- Feng Tang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China; Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou, PR China
| | - Tian-Lun Yang
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhen Zhang
- Department of Centre for Experimental Medicine, Third Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Xiao-Gang Li
- Department of Cardiovascular Medicine, The Third Xiangya Hospital of Central South University, Changsha 410008, Hunan, PR China
| | - Qiao-Qing Zhong
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Ting-Ting Zhao
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Gong
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| |
Collapse
|
20
|
ROS-Dependent Activation of Autophagy through the PI3K/Akt/mTOR Pathway Is Induced by Hydroxysafflor Yellow A-Sonodynamic Therapy in THP-1 Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8519169. [PMID: 28191279 PMCID: PMC5278230 DOI: 10.1155/2017/8519169] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Revised: 12/10/2016] [Accepted: 12/21/2016] [Indexed: 12/11/2022]
Abstract
Monocyte-derived macrophages participate in infaust inflammatory responses by secreting various types of proinflammatory factors, resulting in further inflammatory reactions in atherosclerotic plaques. Autophagy plays an important role in inhibiting inflammation; thus, increasing autophagy may be a therapeutic strategy for atherosclerosis. In the present study, hydroxysafflor yellow A-mediated sonodynamic therapy was used to induce autophagy and inhibit inflammation in THP-1 macrophages. Following hydroxysafflor yellow A-mediated sonodynamic therapy, autophagy was induced as shown by the conversion of LC3-II/LC3-I, increased expression of beclin 1, degradation of p62, and the formation of autophagic vacuoles. In addition, inflammatory factors were inhibited. These effects were blocked by Atg5 siRNA, the autophagy inhibitor 3-methyladenine, and the reactive oxygen species scavenger N-acetyl cysteine. Moreover, AKT phosphorylation at Ser473 and mTOR phosphorylation at Ser2448 decreased significantly after HSYA-SDT. These effects were inhibited by the PI3K inhibitor LY294002, the AKT inhibitor triciribine, the mTOR inhibitor rapamycin, mTOR siRNA, and N-acetyl cysteine. Our results demonstrate that HSYA-SDT induces an autophagic response via the PI3K/Akt/mTOR signaling pathway and inhibits inflammation by reactive oxygen species in THP-1 macrophages.
Collapse
|
21
|
Sasaki Y, Ikeda Y, Iwabayashi M, Akasaki Y, Ohishi M. The Impact of Autophagy on Cardiovascular Senescence and Diseases. Int Heart J 2017; 58:666-673. [DOI: 10.1536/ihj.17-246] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Yuichi Sasaki
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Masaaki Iwabayashi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Yuichi Akasaki
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences, Kagoshima University
| |
Collapse
|
22
|
Systemic application of 3-methyladenine markedly inhibited atherosclerotic lesion in ApoE -/- mice by modulating autophagy, foam cell formation and immune-negative molecules. Cell Death Dis 2016; 7:e2498. [PMID: 27906187 PMCID: PMC5260998 DOI: 10.1038/cddis.2016.376] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/26/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022]
Abstract
A growing body of evidence demonstrates that autophagy, an evolutionarily conserved intracellular degradation process, is involved in the pathogenesis of atherosclerosis and has become a potential therapeutic target. Here we tested the effect of two inhibitors of phosphatidylinositol 3-kinase, 3-methyladenine (3-MA) and 2-(4-morpholinyl)-8-phenyl-chromone (LY294002), commonly used as inhibitors of autophagy, in atherosclerosis in apolipoprotein E−/− mice. Systemic application of 3-MA but not LY294002 markedly reduced the size of atherosclerotic plaque and increased the stability of lesions in high-fat diet-fed mice as compared with controls. Furthermore, 3-MA had multiple atheroprotective effects, including modulating macrophage autophagy and foam cell formation and altering the immune microenvironment. Long-term treatment with 3-MA promoted oxidized low-density lipoprotein (oxLDL)-induced macrophage autophagy and suppressed foam cell formation and cell viability in vitro. Furthermore, systemic application of 3-MA promoted lipid droplet breakdown and decreased apoptosis, most likely associated with autophagy. 3-MA treatment strikingly enhanced the expression of immune-negative molecules such as interleukin 10 (IL-10), transforming growth factor β and IL-35, as well as forkhead box P3 (Foxp3), the specific transcriptional factor for regulatory T cells, but did not affect the level of proinflammatory cytokines in the arterial wall. We provide strong evidence for the potential therapeutic benefit of 3-MA in inhibiting atherosclerosis development and improving plaque stability.
Collapse
|
23
|
Alloza I, Goikuria H, Freijo MDM, Vandenbroeck K. A role for autophagy in carotid atherosclerosis. Eur Stroke J 2016; 1:255-263. [PMID: 31008286 DOI: 10.1177/2396987316674085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
Purpose Autophagy has emerged in recent years as a critical cellular survival mechanism for cell homeostasis and may play a protective role in atherosclerosis. We aimed to review here the role autophagy plays in different cell types present in carotid atherosclerotic plaques and that may be associated with the development of unstable carotid atheroma plaque. Methods We performed a thorough literature exploration in this area of research covering the three main cell types present in carotid atheroma plaques. Findings Reviewed reports indicate that the role of autophagy in stable or unstable carotid atherosclerotic plaques depends on the different cell types and phenotypes, the stage and morphology of the plaque and the specific autophagy factor/s involved. Discussion Although defective autophagy could be one of the causes for carotid atheroma plaques to become unstable, it is important to take into account that autophagic players can act differentially in different cell types and different stages of the developed plaque. Conclusion This review provides an overview of the role of autophagy in the main cell types in carotid atherosclerosis (i.e. macrophages, endothelial cells and smooth muscle cells).
Collapse
Affiliation(s)
- Iraide Alloza
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| | - Haize Goikuria
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| | | | - Koen Vandenbroeck
- Neurogenomiks Group, Neuroscience Department, Faculty of Medicine and Odontology, University of Basque Country, UPV/EHU, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,ACHUCARRO Basque Centre for Neuroscience, Zamudio, Spain
| |
Collapse
|
24
|
Wang T, Zhang L, Hu J, Duan Y, Zhang M, Lin J, Man W, Pan X, Jiang Z, Zhang G, Gao B, Wang H, Sun D. Mst1 participates in the atherosclerosis progression through macrophage autophagy inhibition and macrophage apoptosis enhancement. J Mol Cell Cardiol 2016; 98:108-16. [DOI: 10.1016/j.yjmcc.2016.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/31/2016] [Accepted: 08/01/2016] [Indexed: 10/21/2022]
|
25
|
Leng S, Iwanowycz S, Saaoud F, Wang J, Wang Y, Sergin I, Razani B, Fan D. Ursolic acid enhances macrophage autophagy and attenuates atherogenesis. J Lipid Res 2016; 57:1006-16. [PMID: 27063951 PMCID: PMC4878185 DOI: 10.1194/jlr.m065888] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 04/01/2016] [Indexed: 12/29/2022] Open
Abstract
Macrophage autophagy has been shown to be protective against atherosclerosis. We previously discovered that ursolic acid (UA) promoted cancer cell autophagy. In the present study, we aimed to examine whether UA enhances macrophage autophagy in the context of atherogenesis. Cell culture study showed that UA enhanced autophagy of macrophages by increasing the expression of Atg5 and Atg16l1, which led to altered macrophage function. UA reduced pro-interleukin (IL)-1β protein levels and mature IL-1β secretion in macrophages in response to lipopolysaccharide (LPS), without reducing IL-1β mRNA expression. Confocal microscopy showed that in LPS-treated macrophages, UA increased LC3 protein levels and LC3 appeared to colocalize with IL-1β. In cholesterol-loaded macrophages, UA increased cholesterol efflux to apoAI, although it did not alter mRNA or protein levels of ABCA1 and ABCG1. Electron microscopy showed that UA induced lipophagy in acetylated LDL-loaded macrophages, which may result in increased cholesterol ester hydrolysis in autophagolysosomes and presentation of free cholesterol to the cell membrane. In LDLR(-/-) mice fed a Western diet to induce atherogenesis, UA treatment significantly reduced atherosclerotic lesion size, accompanied by increased macrophage autophagy. In conclusion, the data suggest that UA promotes macrophage autophagy and, thereby, suppresses IL-1β secretion, promotes cholesterol efflux, and attenuates atherosclerosis in mice.
Collapse
Affiliation(s)
- Shuilong Leng
- Department of Human Anatomy, School of Basic Science, Guangzhou Medical University, Guangzhou, Guangdong 510182, People's Republic of China Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Stephen Iwanowycz
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Fatma Saaoud
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Junfeng Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| | - Ismail Sergin
- Cardiovascular Division, Departments of Medicine and Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Babak Razani
- Cardiovascular Division, Departments of Medicine and Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC 29209
| |
Collapse
|
26
|
Hur SK, Park SH, Oh GT. The Role of Autophagy in the Pathogenesis of Atherosclerosis. J Lipid Atheroscler 2016. [DOI: 10.12997/jla.2016.5.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Shin Kyoung Hur
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Seung Hee Park
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
27
|
Zhao S, Yang G, Liu PN, Deng YY, Zhao Z, Sun T, Zhuo XZ, Liu JH, Tian Y, Zhou J, Yuan Z, Wu Y. miR-590-3p Is a Novel MicroRNA in Myocarditis by Targeting Nuclear Factor Kappa-B in vivo. Cardiology 2015; 132:182-8. [PMID: 26278103 DOI: 10.1159/000433596] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/21/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Nuclear factor kappa-B (NF-954;B)-induced inflammation leads to myocarditis and heart dysfunction. How microRNAs (miRNAs) contribute to this process is poorly defined. The aim of this study was to investigate whether miRNAs regulate NF-954;B-induced inflammation in experimental autoimmune myocarditis (EAM) in vivo. METHODS AND RESULTS NF-954;B and its related proinflammatory genes, including interleukin-6 (IL-6) and tumor necrosis factor-a (TNF-a), were activated in EAM. Profiling of NF-954;B-related miRNAs revealed that miR-590-3p was strikingly reduced in EAM. We found IL-6-induced proinflammatory signaling via miR-590-3p reduction, p50 induction, NF-954;B activation and IL-6/TNF-a expression. Moreover, a luciferase reporter assay demonstrated that miR-590-3p directly interacted with the 3' UTR (untranslated region) of the p50 subunit, and that miR-590-3p overexpression inhibited p50 expression. Finally, miR-590-3p transfection through adeno-associated virus significantly inhibited p50 expression, suppressed NF-954;B activity and blocked IL-6/TNF-a expression in vivo, reducing the lesion area and improving cardiac function in EAM. CONCLUSION miR-590-3p is a novel NF-954;B-related miRNA that directly targets the p50 subunit. This may provide a novel strategy for the treatment of myocarditis.
Collapse
Affiliation(s)
- Sijia Zhao
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Immune evasion in cancer: Mechanistic basis and therapeutic strategies. Semin Cancer Biol 2015; 35 Suppl:S185-S198. [PMID: 25818339 DOI: 10.1016/j.semcancer.2015.03.004] [Citation(s) in RCA: 1030] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/27/2022]
Abstract
Cancer immune evasion is a major stumbling block in designing effective anticancer therapeutic strategies. Although considerable progress has been made in understanding how cancers evade destructive immunity, measures to counteract tumor escape have not kept pace. There are a number of factors that contribute to tumor persistence despite having a normal host immune system. Immune editing is one of the key aspects why tumors evade surveillance causing the tumors to lie dormant in patients for years through "equilibrium" and "senescence" before re-emerging. In addition, tumors exploit several immunological processes such as targeting the regulatory T cell function or their secretions, antigen presentation, modifying the production of immune suppressive mediators, tolerance and immune deviation. Besides these, tumor heterogeneity and metastasis also play a critical role in tumor growth. A number of potential targets like promoting Th1, NK cell, γδ T cell responses, inhibiting Treg functionality, induction of IL-12, use of drugs including phytochemicals have been designed to counter tumor progression with much success. Some natural agents and phytochemicals merit further study. For example, use of certain key polysaccharide components from mushrooms and plants have shown to possess therapeutic impact on tumor-imposed genetic instability, anti-growth signaling, replicative immortality, dysregulated metabolism etc. In this review, we will discuss the advances made toward understanding the basis of cancer immune evasion and summarize the efficacy of various therapeutic measures and targets that have been developed or are being investigated to enhance tumor rejection.
Collapse
|
29
|
Michiels CF, Fransen P, De Munck DG, De Meyer GRY, Martinet W. Defective autophagy in vascular smooth muscle cells alters contractility and Ca2+ homeostasis in mice. Am J Physiol Heart Circ Physiol 2015; 308:H557-67. [DOI: 10.1152/ajpheart.00659.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Autophagy is an evolutionary preserved process that prevents the accumulation of unwanted cytosolic material through the formation of autophagosomes. Although autophagy has been extensively studied to understand its function in normal physiology, the role of vascular smooth muscle (SM) cell (VSMC) autophagy in Ca2+ mobilization and contraction remains poorly understood. Recent evidence shows that autophagy is involved in controlling contractile function and Ca2+ homeostasis in certain cell types. Therefore, autophagy might also regulate contractile capacity and Ca2+-mobilizing pathways in VSMCs. Contractility (organ chambers) and Ca2+ homeostasis (myograph) were investigated in aortic segments of 3.5-mo-old mice containing a SM cell-specific deletion of autophagy-related 7 ( Atg7; Atg7 fl/ fl SM22α -Cre+ mice) and in segments of corresponding control mice ( Atg7+/+ SM22α -Cre+). Our results indicate that voltage-gated Ca2+ channels (VGCCs) of Atg7 fl/ fl SM22α -Cre+ VSMCs were more sensitive to depolarization, independent of changes in resting membrane potential. Contractions elicited with K+ (50 mM) or the VGCC agonist BAY K8644 (100 nM) were significantly higher due to increased VGCC expression and activity. Interestingly, the sarcoplasmic reticulum of Atg7 fl/ fl SM22α -Cre+ VSMCs was enlarged, which, combined with increased sarco(endo)plasmic reticulum Ca2+-ATPase 2 expression and higher store-operated Ca2+ entry, promoted inositol 1,4,5-trisphosphate-mediated contractions of Atg7 fl/ fl SM22α -Cre+ segments and maximized the Ca2+ storing capacity of the sarcoplasmic reticulum. Moreover, decreased plasma membrane Ca2+-ATPase expression in Atg7 fl/ fl SM22α -Cre+ VSMCs hampered Ca2+ extrusion to the extracellular environment. Overall, our study indicates that defective autophagy in VSMCs leads to an imbalance between Ca2+ release/influx and Ca2+ reuptake/extrusion, resulting in higher basal Ca2+ concentrations and significant effects on vascular reactivity.
Collapse
Affiliation(s)
| | - Paul Fransen
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Dorien G. De Munck
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
30
|
Abstract
Autophagy is a reparative, life-sustaining process by which cytoplasmic components are sequestered in double-membrane vesicles and degraded on fusion with lysosomal compartments. Growing evidence reveals that basal autophagy is an essential in vivo process mediating proper vascular function. Moreover, autophagy is stimulated by many stress-related stimuli in the arterial wall to protect endothelial cells and smooth muscle cells against cell death and the initiation of vascular disease, in particular atherosclerosis. Basal autophagy is atheroprotective during early atherosclerosis but becomes dysfunctional in advanced atherosclerotic plaques. Little is known about autophagy in other vascular disorders, such as aneurysm formation, arterial aging, vascular stiffness, and chronic venous disease, even though autophagy is often impaired. This finding highlights the need for pharmacological interventions with compounds that stimulate the prosurvival effects of autophagy in the vasculature. A large number of animal studies and clinical trials have indicated that oral or stent-based delivery of the autophagy inducer rapamycin or derivatives thereof, collectively known as rapalogs, effectively inhibit the basic mechanisms that control growth and destabilization of atherosclerotic plaques. Other autophagy-inducing drugs, such as spermidine or add-on therapy with widely used antiatherogenic compounds, including statins and metformin, are potentially useful to prevent vascular disease with minimal adverse effects.
Collapse
Affiliation(s)
- Guido R.Y. De Meyer
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Mandy O.J. Grootaert
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Cédéric F. Michiels
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Ammar Kurdi
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Dorien M. Schrijvers
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- From the Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
31
|
Abderrazak A, Couchie D, Mahmood DFD, Elhage R, Vindis C, Laffargue M, Matéo V, Büchele B, Ayala MR, El Gaafary M, Syrovets T, Slimane MN, Friguet B, Fulop T, Simmet T, El Hadri K, Rouis M. Anti-inflammatory and antiatherogenic effects of the NLRP3 inflammasome inhibitor arglabin in ApoE2.Ki mice fed a high-fat diet. Circulation 2015; 131:1061-70. [PMID: 25613820 DOI: 10.1161/circulationaha.114.013730] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND This study was designed to evaluate the effect of arglabin on the NLRP3 inflammasome inhibition and atherosclerotic lesion in ApoE2Ki mice fed a high-fat Western-type diet. METHODS AND RESULTS Arglabin was purified, and its chemical identity was confirmed by mass spectrometry. It inhibited, in a concentration-dependent manner, interleukin (IL)-1β and IL-18, but not IL-6 and IL-12, production in lipopolysaccharide and cholesterol crystal-activated cultured mouse peritoneal macrophages, with a maximum effect at ≈50 nmol/L and EC50 values for both cytokines of ≈ 10 nmol/L. Lipopolysaccharide and cholesterol crystals did not induce IL-1β and IL-18 production in Nlrp3(-/-) macrophages. In addition, arglabin activated autophagy as evidenced by the increase in LC3-II protein. Intraperitoneal injection of arglabin (2.5 ng/g body weight twice daily for 13 weeks) into female ApoE2.Ki mice fed a high-fat diet resulted in a decreased IL-1β plasma level compared with vehicle-treated mice (5.2±1.0 versus 11.7±1.1 pg/mL). Surprisingly, arglabin also reduced plasma levels of total cholesterol and triglycerides to 41% and 42%, respectively. Moreover, arglabin oriented the proinflammatory M1 macrophages into the anti-inflammatory M2 phenotype in spleen and arterial lesions. Finally, arglabin treatment markedly reduced the median lesion areas in the sinus and whole aorta to 54% (P=0.02) and 41% (P=0.02), respectively. CONCLUSIONS Arglabin reduces inflammation and plasma lipids, increases autophagy, and orients tissue macrophages into an anti-inflammatory phenotype in ApoE2.Ki mice fed a high-fat diet. Consequently, a marked reduction in atherosclerotic lesions was observed. Thus, arglabin may represent a promising new drug to treat inflammation and atherosclerosis.
Collapse
Affiliation(s)
- Amna Abderrazak
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Dominique Couchie
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Dler Faieeq Darweesh Mahmood
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Rima Elhage
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Cécile Vindis
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Muriel Laffargue
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Véronique Matéo
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Berthold Büchele
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Monica Rubio Ayala
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Menna El Gaafary
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Tatiana Syrovets
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Mohamed-Naceur Slimane
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Bertrand Friguet
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Tamas Fulop
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Thomas Simmet
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Khadija El Hadri
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.)
| | - Mustapha Rouis
- From Sorbonne Universités, UPMC Université Paris 06, Biological Adaptation and Ageing-IBPS, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); CNRS-UMR 8256, Paris, France (A.A., D.C., D.F.D.M., R.E.H., F.B., K.E.H., M.R.); Inserm U1164, Paris, France (A.A., D.C., D.F.D.M., R.E.H., B.F., K.E.H., M.R.); INSERM-UPS UMR 1048, Institut des Maladies Métaboliques et Cardiovasculaires, Toulouse, France (C.V., M.L.); Centre de Recherche d'Immunologie et Maladies Infectieuses, CIMI-Paris UPMC UMRS CR7, INSERM U-1135, CNRS ERL 8255, Hôpital Pitié-Salpêtrière, Paris, France (V.M.); Ulm University, Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm, Germany (B.B., M.R.A., M.E.G., T.S., T.S.); Biochemistry Laboratory, Faculty of Medicine, TN-Monastir, Tunisia (M.-N.S.); and Centre de Recherche sur le Vieillissement, Service Gériatrique, Département de Médecine, Université de Sherbrooke, Sherbrooke, QC, Canada (T.F.).
| |
Collapse
|
32
|
Swaminathan B, Goikuria H, Vega R, Rodríguez-Antigüedad A, López Medina A, Freijo MDM, Vandenbroeck K, Alloza I. Autophagic marker MAP1LC3B expression levels are associated with carotid atherosclerosis symptomatology. PLoS One 2014; 9:e115176. [PMID: 25503069 PMCID: PMC4264866 DOI: 10.1371/journal.pone.0115176] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 11/19/2014] [Indexed: 12/31/2022] Open
Abstract
Objectives The mechanism by which atheroma plaque becomes unstable is not completely understood to date but analysis of differentially expressed genes in stable versus unstable plaques may provide clues. This will be crucial toward disclosing the mechanistic basis of plaque instability, and may help to identify prognostic biomarkers for ischaemic events. The objective of our study was to identify differences in expression levels of 59 selected genes between symptomatic patients (unstable plaques) and asymptomatic patients (stable plaques). Methods 80 carotid plaques obtained by carotid endarterectomy and classified as symptomatic (>70% stenosis) or asymptomatic (>80% stenosis) were used in this study. The expression levels of 59 genes were quantified by qPCR on RNA extracted from the carotid plaques obtained by endarterectomy and analyzed by means of various bioinformatic tools. Results Several genes associated with autophagy pathways displayed differential expression levels between asymptomatic and symptomatic (i.e. MAP1LC3B, RAB24, EVA1A). In particular, mRNA levels of MAP1LC3B, an autophagic marker, showed a 5−fold decrease in symptomatic samples, which was confirmed in protein blots. Immune system−related factors and endoplasmic reticulum-associated markers (i.e. ERP27, ITPR1, ERO1LB, TIMP1, IL12B) emerged as differently expressed genes between asymptomatic and symptomatic patients. Conclusions Carotid atherosclerotic plaques in which MAP1LC3B is underexpressed would not be able to benefit from MAP1LC3B−associated autophagy. This may lead to accumulation of dead cells at lesion site with subsequent plaque destabilization leading to cerebrovascular events. Identified biomarkers and network interactions may represent novel targets for development of treatments against plaque destabilization and thus for the prevention of cerebrovascular events.
Collapse
Affiliation(s)
- Bhairavi Swaminathan
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
| | - Haize Goikuria
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
| | - Reyes Vega
- Department of Neurology, Basurto Hospital, Bilbao, Spain
| | | | | | | | - Koen Vandenbroeck
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- IKERBASQUE, Basque Foundation for Sciences, Bilbao, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
| | - Iraide Alloza
- Neurogenomiks, Neurosciences Department, Faculty of Medicine and Odontology, University of Basque Country, Leioa, Spain
- IKERBASQUE, Basque Foundation for Sciences, Bilbao, Spain
- Achucarro Basque Center for Neurosciences, Zamudio, Spain
- * E-mail:
| |
Collapse
|
33
|
Abstract
Autophagy is an essential cellular pathway by which protein aggregates, long-lived proteins, or defective organelles are sequestered in double membrane vesicles and then degraded upon fusion of those vesicles with lysosomes. Although autophagy plays a critical role in maintaining intracellular homeostasis and keeping the cell in a healthy state, this key pathway can become dysregulated in various cardiometabolic disorders, such as; obesity, dyslipidemia, inflammation, and insulin resistance. In these conditions, autophagy may actually worsen the pathological state instead of protecting the cell or organism. In this review, we discuss how dysregulated autophagy may be linked to increases in cardiovascular risk factors, and how manipulation of the autophagic machinery might reduce those risks.
Collapse
Affiliation(s)
- Juan G. Juárez-Rojas
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY USA
- Endocrinolgy Department, National Institute of Cardiology “Ignacio Chávez”, Mexico City, Mexico
| | - Gissette Reyes-Soffer
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY USA
| | - Donna Conlon
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY USA
- PH10-305, Irving Institute for Clinical and Translational Research, 630 West 168 Street, New York, NY 10032 USA
| |
Collapse
|
34
|
HUANG BAOJUN, JIN MENGXING, YAN HAI, CHENG YANWEI, HUANG DAKE, YING SONGCHENG, ZHANG LINJIE. Simvastatin enhances oxidized-low density lipoprotein-induced macrophage autophagy and attenuates lipid aggregation. Mol Med Rep 2014; 11:1093-8. [DOI: 10.3892/mmr.2014.2790] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 10/02/2014] [Indexed: 11/06/2022] Open
|
35
|
Kain V, Prabhu SD, Halade GV. Inflammation revisited: inflammation versus resolution of inflammation following myocardial infarction. Basic Res Cardiol 2014; 109:444. [PMID: 25248433 DOI: 10.1007/s00395-014-0444-7] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 12/14/2022]
Abstract
Myocardial infarction (MI) is the main cause for the progression of the left ventricle towards congestive heart failure. The optimal healing after MI requires timely induction and resolution of inflammation. Primarily, there have been a number of strategies applied to inhibit the post-MI inflammation but approaches that focus on the resolution of inflammation have sparsely been used in the treatment of heart failure. The early attempts to inhibit post-MI inflammation resulted in adverse outcomes that were realized in heart failure trials. We provide here an overview on the cyclooxygenase (COX)- and lipoxygenase (LOX)-derived lipid mediators that are either impairing or resolving the post-MI inflammation. With the evolution of lipidomics there has been emerging novel bioactive-specialized lipid mediators that promise to resolve chronic inflammation rather than promoting inhibition. The current review is focused on post-MI immune cells kinetics and the unexplored array of lipid mediators that are coordinated by COX and LOX. Thus, an emphasis on COX and LOX poses key questions and potential for the development of novel targets in the heart failure treatment strategy. This updated dynamic approach aims to fuse basic pre-clinical discoveries and translational bioactive lipid-based resolvin discoveries that could be potentially used in the clinic for the treatment of heart failure.
Collapse
Affiliation(s)
- Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, 703 19th Street South, Birmingham, AL, 35233, USA
| | | | | |
Collapse
|
36
|
Peng N, Meng N, Wang S, Zhao F, Zhao J, Su L, Zhang S, Zhang Y, Zhao B, Miao J. An activator of mTOR inhibits oxLDL-induced autophagy and apoptosis in vascular endothelial cells and restricts atherosclerosis in apolipoprotein E⁻/⁻ mice. Sci Rep 2014; 4:5519. [PMID: 24980430 PMCID: PMC4076681 DOI: 10.1038/srep05519] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 06/13/2014] [Indexed: 12/28/2022] Open
Abstract
Oxidized low-density lipoprotein (oxLDL) inhibits mammalian target of rapamycin (mTOR) and induces autophagy and apoptosis in vascular endothelial cells (VECs) that play very critical roles for the cardiovascular homostasis. We recently defined 3-benzyl-5-((2-nitrophenoxy) methyl)-dihydrofuran-2(3H)-one (3BDO) as a new activator of mTOR. Therefore, we hypothesized that 3BDO had a protective role in VECs and thus stabilized atherosclerotic lesions in apolipoprotein E-/- (apoE-/-) mice. Our results showed that oxLDL inhibited the activity of mTOR and increased the protein level of autophagy-related 13 (ATG13) and its dephosphorylation, thus inducing autophagy in human umbilical vein endothelial cells (HUVECs). All of these effects were strongly inhibited by 3BDO. In vivo experiments confirmed that 3BDO activated mTOR and decreased the protein level of ATG13 in the plaque endothelium of apoE-/- mice. Importantly, 3BDO did not affect the activity of mTOR and autophagy in macrophage cell line RAW246.7 and vascular smooth muscle cells of apoE-/- mice, but suppressed plaque endothelial cell death and restricted atherosclerosis development in the mice. 3BDO protected VECs by activating mTOR and thus stabilized atherosclerotic lesions in apoE-/- mice.
Collapse
Affiliation(s)
- Nan Peng
- 1] Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China [2]
| | - Ning Meng
- 1] School of Biological Science and Technology, University of Jinan, Jinan 250022, China [2] Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China [3]
| | - ShengQing Wang
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - Fei Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Jing Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Le Su
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - ShangLi Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, 250012, China
| | - BaoXiang Zhao
- Institute of Organic Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China
| | - JunYing Miao
- 1] Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Science, Shandong University, Jinan 250100, China [2] The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, 250012, China
| |
Collapse
|
37
|
Abstract
Oxidative stress and inflammation underpin most diseases; their mechanisms are inextricably linked. Chronic inflammation is associated with oxidation, anti-inflammatory cascades are linked to decreased oxidation, increased oxidative stress triggers inflammation, and redox balance inhibits the inflammatory cellular response. Whether or not oxidative stress and inflammation represent the cause or consequence of cellular pathology, they contribute significantly to the pathogenesis of noncommunicable diseases (NCD). The incidence of obesity and other related metabolic disturbances are increasing, as are age-related diseases due to a progressively aging population. Relationships between oxidative stress, inflammatory signaling, and metabolism are, in the broad sense of energy transformation, being increasingly recognized as part of the problem in NCD. In this chapter, we summarize the pathologic consequences of an imbalance between circulating and cellular paraoxonases, the system for scavenging excessive reactive oxygen species and circulating chemokines. They act as inducers of migration and infiltration of immune cells in target tissues as well as in the pathogenesis of disease that perturbs normal metabolic function. This disruption involves pathways controlling lipid and glucose homeostasis as well as metabolically driven chronic inflammatory states that encompass several response pathways. Dysfunction in the endoplasmic reticulum and/or mitochondria represents an important feature of chronic disease linked to oxidation and inflammation seen as self-reinforcing in NCD. Therefore, correct management requires a thorough understanding of these relationships and precise interpretation of laboratory test results.
Collapse
|
38
|
Cheng P, Wang F, Chen K, Shen M, Dai W, Xu L, Zhang Y, Wang C, Li J, Yang J, Zhu R, Zhang H, Zheng Y, Lu J, Zhou Y, Guo C. Hydrogen sulfide ameliorates ischemia/reperfusion-induced hepatitis by inhibiting apoptosis and autophagy pathways. Mediators Inflamm 2014; 2014:935251. [PMID: 24966472 PMCID: PMC4055384 DOI: 10.1155/2014/935251] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 04/08/2014] [Accepted: 04/08/2014] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Hepatic ischemia/reperfusion (I/R) injury is an important clinical problem, and its consequences can seriously threaten human health. Apoptosis and autophagy have been shown to contribute to cell death in hepatic I/R injury. Hydrogen sulfide (H2S) is the third most common endogenously produced gaseous signaling molecule and is known to exert a protective effect against hepatic I/R injury. In this study, the purpose is to explore both the effect and mechanism of H2S on hepatic I/R injury. METHODS Balb/c mice were randomized into Sham, I/R, or two doses (14 μmol/kg and 28 μmol/kg) of sodium hydrosulfide (NaHS, an H2S donor) preconditioning groups. RESULTS NaHS significantly reduced the levels of TNF- α and IL-6 at 12 h and 24 h after injection compared with ischemia/reperfusion challenge alone. The expression of Bcl-2, Bax, Beclin-1, and LC3, which play important roles in the regulation of the apoptosis and autophagy pathways, was also clearly affected by NaHS. Furthermore, NaHS affected the p-JNK1, p-ERK1, and p-p38. CONCLUSION Our results indicate that H2S attenuates hepatic I/R injury, at least in part, by regulating apoptosis through inhibiting JNK1 signaling. The autophagy agonist rapamycin potentiated this hepatoprotective effect by reversing the inhibition of autophagy by H2S.
Collapse
Affiliation(s)
- Ping Cheng
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Miao Shen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ling Xu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
- Department of Gastroenterology, The Tongren Hospital of Shanghai Jiaotong University, Shanghai 200050, China
| | - Yan Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chengfen Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jing Yang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Rong Zhu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
- Department of Gastroenterology, Clinical Medical College of the Tenth People's Hospital of Nangjing Medical University, Shanghai 200072, China
| | - Huawei Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yuanyuan Zheng
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| |
Collapse
|
39
|
CD40L contributes to angiotensin II-induced pro-thrombotic state, vascular inflammation, oxidative stress and endothelial dysfunction. Basic Res Cardiol 2013; 108:386. [DOI: 10.1007/s00395-013-0386-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 08/14/2013] [Accepted: 09/06/2013] [Indexed: 12/27/2022]
|
40
|
Lavandero S, Troncoso R, Rothermel BA, Martinet W, Sadoshima J, Hill JA. Cardiovascular autophagy: concepts, controversies, and perspectives. Autophagy 2013; 9:1455-66. [PMID: 23959233 DOI: 10.4161/auto.25969] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Despite recent scientific and technological advances, cardiovascular disease remains the leading cause of morbidity and mortality worldwide. Autophagy, an evolutionarily ancient response to cellular stress, has been implicated in the pathogenesis of a wide range of heart pathologies. However, the precise role of autophagy in these contexts remains obscure owing to its multifarious actions. Here, we review recently derived insights regarding the role of autophagy in multiple manifestations of cardiac plasticity and disease.
Collapse
Affiliation(s)
- Sergio Lavandero
- Center for Molecular Studies of the Cell; Faculty of Chemical & Pharmaceutical Sciences/ Faculty of Medicine; University of Chile; Santiago, Chile; Department of Internal Medicine (Cardiology Division); University of Texas Southwestern Medical Center; Dallas, TX USA
| | - Rodrigo Troncoso
- Center for Molecular Studies of the Cell; Faculty of Chemical & Pharmaceutical Sciences/ Faculty of Medicine; University of Chile; Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division); University of Texas Southwestern Medical Center; Dallas, TX USA; Department of Molecular Biology; University of Texas Southwestern Medical Center; Dallas, TX USA
| | - Wim Martinet
- Laboratory of Physiopharmacology; University of Antwerp; Antwerp, Belgium
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine; Rutgers New Jersey Medical School; Newark, NJ USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology Division); University of Texas Southwestern Medical Center; Dallas, TX USA; Department of Molecular Biology; University of Texas Southwestern Medical Center; Dallas, TX USA
| |
Collapse
|
41
|
Macrophage autophagy in atherosclerosis. Mediators Inflamm 2013; 2013:584715. [PMID: 23401644 PMCID: PMC3563164 DOI: 10.1155/2013/584715] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 12/27/2012] [Indexed: 12/14/2022] Open
Abstract
Macrophages play crucial roles in atherosclerotic immune responses. Recent investigation into macrophage autophagy (AP) in atherosclerosis has demonstrated a novel pathway through which these cells contribute to vascular inflammation.
AP is a cellular catabolic process involving the delivery of cytoplasmic contents to the lysosomal machinery for ultimate degradation and recycling. Basal levels of macrophage AP play an essential role in atheroprotection during early atherosclerosis. However, AP becomes dysfunctional in the more advanced stages of the pathology and its deficiency promotes vascular inflammation, oxidative stress, and plaque necrosis. In this paper, we will discuss the role of macrophages and AP in atherosclerosis and the emerging evidence demonstrating the contribution of macrophage AP to vascular pathology. Finally, we will discuss how AP could be targeted for therapeutic utility.
Collapse
|