1
|
Gu Y, Bi X, Liu X, Qian Q, Wen Y, Hua S, Fu Q, Zheng Y, Sun S. Roles of ABCA1 in Chronic Obstructive Pulmonary Disease. COPD 2025; 22:2493701. [PMID: 40302380 DOI: 10.1080/15412555.2025.2493701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the common chronic respiratory diseases, which causes a heavy burden to patients and society. Increasing studies suggest that ABCA1 plays an important role in COPD. ABCA1 belongs to a large class of ATP-binding (ABC) transporters. It is not only involved in the reverse transport of cholesterol, but also in the regulation of apoptosis, pyroptosis, cellular inflammation and cellular immunity. Meanwhile, ABCA1 is involved in several signaling pathways, such as SREBP pathway, LXR pathway, MAPK pathway, p62/mTOR pathway, CTRP1 pathway and so on. In addition, the ABCA1 participates in the disorder of lipid metabolism in COPD by regulating the formation of RCT and HDL, regulates the inflammation of COPD by removing excess cholesterol in macrophages, and promotes the differentiation of COPD phenotype into emphysema type. Accordingly, the ABCA1 may be a therapeutic target for COPD.
Collapse
Affiliation(s)
- Ying Gu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaoqing Bi
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaofei Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qingqing Qian
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qiaoli Fu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yuanyuan Zheng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Song C, Ling H, Yang G, Ding J. Microenvironments‐Targeted Nanomaterials for Atherosclerosis Therapy. ADVANCED FUNCTIONAL MATERIALS 2025. [DOI: 10.1002/adfm.202421512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Indexed: 05/14/2025]
Abstract
AbstractAtherosclerosis significantly contributes to cardiovascular disease. Traditional treatments for atherosclerosis, such as pharmacological interventions and surgical procedures, have demonstrated limited efficacy and often yield unsatisfactory results. Consequently, safe and effective therapeutic strategies are urgently needed. The atherosclerotic microenvironments, characterized by inflammation driven by foam cells, damaged endothelial cells, recruited leukocytes, lipoproteins, and inflammatory mediators, play a key role in disease progression. By leveraging the biological components and physicochemical properties of these microenvironments, researchers have developed microenvironments‐targeted nanomaterials as a promising approach to treat atherosclerosis. These nanomaterials aim to address and eliminate inflammatory processes. Their functions include repairing endothelial damage, reducing lipoprotein accumulation, inhibiting leukocyte chemotaxis, suppressing foam cell formation, delaying plaque rupture, and preventing thrombosis within the plaque. This review highlights the therapeutic mechanisms and effects of nanomaterials targeting key processes in atherosclerotic microenvironments. Finally, the challenges and prospects of nanomaterial‐based therapies for atherosclerosis are discussed to inspire the development of nanomaterials that modulate atherosclerotic microenvironments, potentially leading to promising clinical applications.
Collapse
Affiliation(s)
- Chunli Song
- Department of General Practice The Second Hospital of Jilin University 4026 Yatai Street Changchun 130041 P. R. China
| | - Hao Ling
- Department of General Practice The Second Hospital of Jilin University 4026 Yatai Street Changchun 130041 P. R. China
| | - Guanqing Yang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| |
Collapse
|
3
|
Sun X, Zhou Q, Xiao C, Mao C, Liu Y, Chen G, Song Y. Role of post-translational modifications of Sp1 in cardiovascular diseases. Front Cell Dev Biol 2024; 12:1453901. [PMID: 39252788 PMCID: PMC11381397 DOI: 10.3389/fcell.2024.1453901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Specific protein 1 (Sp1) is pivotal in sustaining baseline transcription as well as modulating cell signaling pathways and transcription factors activity. Through interactions with various proteins, especially transcription factors, Sp1 controls the expression of target genes, influencing numerous biological processes. Numerous studies have confirmed Sp1's significant regulatory role in the pathogenesis of cardiovascular disorders. Post-translational modifications (PTMs) of Sp1, such as phosphorylation, ubiquitination, acetylation, glycosylation, SUMOylation, and S-sulfhydration, can enhance or modify its transcriptional activity and DNA-binding stability. These modifications also regulate Sp1 expression across different cell types. Sp1 is crucial in regulating non-coding gene expression and the activity of proteins in response to pathophysiological stimuli. Understanding Sp1 PTMs advances our knowledge of cell signaling pathways in controlling Sp1 stability during cardiovascular disease onset and progression. It also aids in identifying novel pharmaceutical targets and biomarkers essential for preventing and managing cardiovascular diseases.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Synopsis of the Golden Chamber, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Zhou
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Typhoid, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caiyun Mao
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Liu
- The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan, China
| | - Guozhen Chen
- Department of Pediatrics, Yantai Yuhuangding Hospital, Shandong, China
| | - Yunjia Song
- Department of Pharmacology, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Zhang Z, Li L, Shi H, Chen B, Li X, Zhang Y, Liu F, Wei W, Zhou Y, Liu K, Xia W, Gu X, Huang J, Tu S, Yin C, Shao A, Jiang L. Role of Circular RNAs in Atherosclerosis through Regulation of Inflammation, Cell Proliferation, Migration, and Apoptosis: Focus on Atherosclerotic Cerebrovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1461. [PMID: 37629751 PMCID: PMC10456328 DOI: 10.3390/medicina59081461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
Atherosclerosis (AS) is a disease dangerous to human health and the main pathological cause of ischemic cardiovascular diseases. Although its pathogenesis is not fully understood, numerous basic and clinical studies have shown that AS is a chronic inflammatory disease existing in all stages of atherogenesis. It may be a common link or pathway in the pathogenesis of multiple atherogenic factors. Inflammation is associated with AS complications, such as plaque rupture and ischemic cerebral infarction. In addition to inflammation, apoptosis plays an important role in AS. Apoptosis is a type of programmed cell death, and different apoptotic cells have different or even opposite roles in the process of AS. Unlike linear RNA, circular RNA (circRNA) a covalently closed circular non-coding RNA, is stable and can sponge miRNA, which can affect the stages of AS by regulating downstream pathways. Ultimately, circRNAs play very important roles in AS by regulating inflammation, apoptosis, and some other mechanisms. The study of circular RNAs can provide new ideas for the prediction, prevention, and treatment of AS.
Collapse
Affiliation(s)
- Zheng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Huanqing Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Biao Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Xiaoqin Li
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Yuyao Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Fei Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wan Wei
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Yongji Zhou
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Keqin Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wenqing Xia
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Xin Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Jinyu Huang
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China;
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310006, China;
| | - Congguo Yin
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Disease, Hangzhou 310009, China
| | - Lin Jiang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| |
Collapse
|
5
|
Ghai S, Young A, Su KH. Proteotoxic stress response in atherosclerotic cardiovascular disease: Emerging role of heat shock factor 1. Front Cardiovasc Med 2023; 10:1155444. [PMID: 37077734 PMCID: PMC10106699 DOI: 10.3389/fcvm.2023.1155444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023] Open
Abstract
Atherosclerosis is a major risk factor for cardiovascular diseases. Hypercholesterolemia has been both clinically and experimentally linked to cardiovascular disease and is involved in the initiation of atherosclerosis. Heat shock factor 1 (HSF1) is involved in the control of atherosclerosis. HSF1 is a critical transcriptional factor of the proteotoxic stress response that regulates the production of heat shock proteins (HSPs) and other important activities such as lipid metabolism. Recently, HSF1 is reported to directly interact with and inhibit AMP-activated protein kinase (AMPK) to promote lipogenesis and cholesterol synthesis. This review highlights roles of HSF1 and HSPs in critical metabolic pathways of atherosclerosis, including lipogenesis and proteome homeostasis.
Collapse
|
6
|
Dai T, He W, Tu S, Han J, Yuan B, Yao C, Ren W, Wu A. Black TiO2 nanoprobe-mediated mild phototherapy reduces intracellular lipid levels in atherosclerotic foam cells via cholesterol regulation pathways instead of apoptosis. Bioact Mater 2022; 17:18-28. [PMID: 35386468 PMCID: PMC8958315 DOI: 10.1016/j.bioactmat.2022.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
Given that apoptosis increases the risk of plaque rupture, strategies that reduce intracellular lipid levels without killing foam cells are warranted for safe and effective treatment of atherosclerosis. In this study, a mild phototherapy strategy is carried out to achieve the hypothesis. Foam cell-targeted nanoprobes that allow photothermal therapy (PTT) and/or photodynamic therapy (PDT) were prepared by loading hyaluronan and porphine onto black TiO2 nanoparticles. The results showed that when temperatures below 45 °C, PTT alone and PTT + PDT significantly reduced the intracellular lipid burden without inducing evidently apoptosis or necrosis. In contrast, the use of PDT alone resulted in only a slight reduction in lipid levels and induced massive apoptosis or necrosis. The protective effect against apoptosis or necrosis after mild-temperature PTT and PTT + PDT was correlated with the upregulation of heat shock protein 27. Further, mild-temperature PTT and PTT + PDT attenuated intracellular cholesterol biosynthesis and excess cholesterol uptake via the SREBP2/LDLR pathway, and also triggered ABCA1-mediated cholesterol efflux, ultimately inhibiting lipid accumulation in foam cells. Our results offer new insights into the mechanism of lipid regulation in foam cells and indicate that the black TiO2 nanoprobes could allow safer and more effective phototherapy of atherosclerosis. Mild phototherapy reduced the intracellular lipid in foam cells without inducing obvious apoptosis or necrosis. HSP27 was upregulated in foam cells treated by mild phototherapy, which could protect cells against apoptosis or necrosis. Mild phototherapy attenuated intracellular cholesterol biosynthesis and excess uptake, also boosted cholesterol efflux.
Collapse
Affiliation(s)
- Ting Dai
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
| | - Wenming He
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
| | - Shuangshuang Tu
- Department of Cardiology, The Affiliated Hospital of Medical School, Ningbo University, 247 Renmin Road, Jiangbei District, Ningbo, Zhejiang Province, 315020, China
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
| | - Jinru Han
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- University of Chinese Academy of Sciences, No. 1 Yanqihu East Road, Huairou District, Beijing, 101408, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Bo Yuan
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Chenyang Yao
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
| | - Wenzhi Ren
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
- Corresponding author. Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China.
| | - Aiguo Wu
- Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, 516000, China
- Corresponding author. Cixi Institute of Biomedical Engineering, International Cooperation Base of Biomedical Materials Technology and Application, CAS Key Laboratory of Magnetic Materials and Devices & Zhejiang Engineering Research Center for Biomedical Materials, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, 1219 ZhongGuan West Road, Ningbo, 315201, China.
| |
Collapse
|
7
|
Jiang JF, Zhou ZY, Liu YZ, Wu L, Nie BB, Huang L, Zhang C. Role of Sp1 in atherosclerosis. Mol Biol Rep 2022; 49:9893-9902. [PMID: 35715606 DOI: 10.1007/s11033-022-07516-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 10/18/2022]
Abstract
Specificity protein (Sp) is a famous family of transcription factors including Sp1, Sp2 and Sp3. Sp1 is the first one of Sp family proteins to be characterized and cloned in mammalian. It has been proposed that Sp1 acts as a modulator of the expression of target gene through interacting with a series of proteins, especially with transcriptional factors, and thereby contributes to the regulation of diverse biological processes. Notably, growing evidence indicates that Sp1 is involved in the main events in the development of atherosclerosis (AS), such as inflammation, lipid metabolism, plaque stability, vascular smooth muscle cells (VSMCs) proliferation and endothelial dysfunction. This review is designed to provide useful clues to further understanding roles of Sp1 in the pathogenesis of AS, and may be helpful for the design of novel efficacious therapeutics agents targeting Sp1.
Collapse
Affiliation(s)
- Jie-Feng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
| | - Zheng-Yang Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
| | - Yi-Zhang Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
| | - Li Wu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
| | - Bin-Bin Nie
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, 421001, Hengyang, Hunan, People's Republic of China.
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, 421001, Hengyang, Hunan, People's Republic of China.
| |
Collapse
|
8
|
Chen L, Zhao ZW, Zeng PH, Zhou YJ, Yin WJ. Molecular mechanisms for ABCA1-mediated cholesterol efflux. Cell Cycle 2022; 21:1121-1139. [PMID: 35192423 PMCID: PMC9103275 DOI: 10.1080/15384101.2022.2042777] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The maintenance of cellular cholesterol homeostasis is essential for normal cell function and viability. Excessive cholesterol accumulation is detrimental to cells and serves as the molecular basis of many diseases, such as atherosclerosis, Alzheimer's disease, and diabetes mellitus. The peripheral cells do not have the ability to degrade cholesterol. Cholesterol efflux is therefore the only pathway to eliminate excessive cholesterol from these cells. This process is predominantly mediated by ATP-binding cassette transporter A1 (ABCA1), an integral membrane protein. ABCA1 is known to transfer intracellular free cholesterol and phospholipids to apolipoprotein A-I (apoA-I) for generating nascent high-density lipoprotein (nHDL) particles. nHDL can accept more free cholesterol from peripheral cells. Free cholesterol is then converted to cholesteryl ester by lecithin:cholesterol acyltransferase to form mature HDL. HDL-bound cholesterol enters the liver for biliary secretion and fecal excretion. Although how cholesterol is transported by ABCA1 to apoA-I remains incompletely understood, nine models have been proposed to explain this effect. In this review, we focus on the current view of the mechanisms underlying ABCA1-mediated cholesterol efflux to provide an important framework for future investigation and lipid-lowering therapy.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Zhen-Wang Zhao
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Peng-Hui Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying-Jie Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wen-Jun Yin
- Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China,CONTACT Wen-Jun Yin Department of Clinical Laboratory, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan421001, China
| |
Collapse
|
9
|
Matsuo M. ABCA1 and ABCG1 as potential therapeutic targets for the prevention of atherosclerosis. J Pharmacol Sci 2022; 148:197-203. [DOI: 10.1016/j.jphs.2021.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
|
10
|
Chen W, Wang S, Xing D. New Horizons for the Roles and Association of APE1/Ref-1 and ABCA1 in Atherosclerosis. J Inflamm Res 2021; 14:5251-5271. [PMID: 34703267 PMCID: PMC8526300 DOI: 10.2147/jir.s330147] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/25/2021] [Indexed: 12/27/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide. APE1/Ref-1 and ABCA1 play key roles in the progression of atherosclerosis. APE1/Ref-1 suppresses atherosclerosis via multiple mechanisms, including reducing the IL-6-, TNF-α-, and IL-1β-mediated proinflammatory responses, suppressing ROS-mediated oxidant activity and Bax/Bcl-2-mediated vascular calcification and apoptosis, and reducing LOX-1-mediated cholesterol uptake. However, APE1/Ref-1 also promotes atherosclerosis by increasing the activity of the NK-κB and S1PR1 pathways. APE1/Ref-1 localizes to the nucleus, cytoplasm, and mitochondria and can be secreted from the cell. APE1/Ref-1 localization is dynamically regulated by the disease state and may be responsible for its proatherogenic and antiatherogenic effects. ABCA1 promotes cholesterol efflux and anti-inflammatory responses by binding to apoA-I and regulates apoptotic cell clearance and HSPC proliferation to protect against inflammatory responses. Interestingly, in addition to mediating these functions, ABCA1 promotes the secretion of acetylated APE1/Ref-1 (AcAPE1/Ref-1), a therapeutic target, which protects against atherosclerosis development. The APE1/Ref-1 inhibitor APX3330 is being evaluated in a phase II clinical trial. The LXR agonist LXR-623 (WAY-252623) is an agonist of ABCA1 and the first LXR-targeting compound to be evaluated in clinical trials. In this article, we review the roles of ABCA1 and APE1/Ref-1 in atherosclerosis and focus on new insights into the ABCA1-APE1/Ref-1 axis and its potential as a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266071, People's Republic of China
| | - Shuai Wang
- School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong, 261053, People's Republic of China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, 266071, People's Republic of China.,School of Life Sciences, Tsinghua University, Beijing, 100084, People's Republic of China
| |
Collapse
|
11
|
Tan C, Zhou L, Wen W, Xiao N. Curcumin promotes cholesterol efflux by regulating ABCA1 expression through miR-125a-5p/SIRT6 axis in THP-1 macrophage to prevent atherosclerosis. J Toxicol Sci 2021; 46:209-222. [PMID: 33952798 DOI: 10.2131/jts.46.209] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
OBJECTIVE To seek out the effect of curcumin on cholesterol efflux in THP-1 macrophages and clarify its specific mechanism. METHODS THP-1 macrophages were cultured with curcumin at different concentrations, followed by detection of the toxicity of curcumin to cells utilizing CCK-8 assay. Following culturing with serum-free ox-LDL, THP-1 macrophages were transfected with mi-miR-125a-5p, or in-miR-125a-5p, or pcDNA3.1-SIRT6, or si-SIRT6 for 24 hr, prior to treatment with curcumin at different concentrations. Oil red O staining was applied to examine the formation rate of foam cells, the kits were used for measuring intracellular lipid content of THP-1 macrophages, and the fluorescence detection kit for observing the cholesterol efflux rate. The expressions of miR-125a-5p, SIRT6, and ABCA1 were assayed by qRT-PCR and Western blot. ELISA was adopted to assess the contents of TNF-α, IL-6, and MCP-1. The interaction between miR-125a-5p and SIRT6 was evaluated by dual-luciferase reporter gene assay. RESULTS The optimal dosage of curcumin could reduce foam cell formation and intracellular lipid content, and promote cholesterol efflux in THP-1 macrophages. Meanwhile, curcumin markedly suppressed the expression of miR-125a-5p and upregulated the expression of SIRT6. MiR-125a-5p negatively targeted SIRT6. Overexpression of SIRT6 partially reversed the inhibition role of miR-125a-5p mimic in the biological function of curcumin. Silencing of SIRT6 could partially reverse the effect of the miR-125a-5p inhibitor on the biological function of curcumin. CONCLUSION urcumin could promote cholesterol efflux of THP-1 macrophages through miR-125a-5p/SIRT6 axis and regulate the expression of ABCA1.
Collapse
Affiliation(s)
- Chao Tan
- Department of Internal Medicine, the First Affiliated Hospital of Hunan University of Chinese Medicine, China.,The Domestic First-class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, China.,Inherit Workroom of Medical Master Professor Jibo Xiong's Experiences, China
| | - Lan Zhou
- Graduate School of Hunan University of Chinese Medicine, China
| | - Weinong Wen
- Inherit Workroom of Medical Master Professor Jibo Xiong's Experiences, China
| | - Nan Xiao
- Graduate School of Hunan University of Chinese Medicine, China
| |
Collapse
|
12
|
Aminian AR, Forouzanfar F. Interplay between Heat Shock Proteins, Inflammation, and Pain: A promising Therapeutic Approach. Curr Mol Pharmacol 2021; 15:170-178. [PMID: 34781874 DOI: 10.2174/1874467214666210719143150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 11/22/2022]
Abstract
Heat Shock Proteins (HSPs) are important molecular chaperones that facilitate many functions of the cells. They also play a pivotal role in cell survival, especially in the presence of stressors, including nutritional deprivation, lack of oxygen, fever, alcohol, inflammation, oxidative stress, heavy metals, as well as conditions that cause injury and necrosis. In the face of a painful stimulus encounter, many factors could be associated with pain that may include nitric oxide, excitatory amino acids, reactive oxygen species (ROS) formation, prostaglandins, and inflammatory cytokines. One influential factor affecting pain reduction is the expression of HSPs that act as a ROS scavenger, regulate the inflammatory cytokines, and reduce pain responses subsequently. Hence, we assembled information on the painkilling attributes of HSPs. In this field of research, new painkillers could be developed by targetting HSPs to alleviate pain and widen our grasp of pain in pathological conditions and neurological diseases.
Collapse
Affiliation(s)
- Ahmad Reza Aminian
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad. Iran
| |
Collapse
|
13
|
Lien CF, Chen SJ, Tsai MC, Lin CS. Potential Role of Protein Kinase C in the Pathophysiology of Diabetes-Associated Atherosclerosis. Front Pharmacol 2021; 12:716332. [PMID: 34276388 PMCID: PMC8283198 DOI: 10.3389/fphar.2021.716332] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus is a metabolic syndrome that affects millions of people worldwide. Recent studies have demonstrated that protein kinase C (PKC) activation plays an important role in hyperglycemia-induced atherosclerosis. PKC activation is involved in several cellular responses such as the expression of various growth factors, activation of signaling pathways, and enhancement of oxidative stress in hyperglycemia. However, the role of PKC activation in pro-atherogenic and anti-atherogenic mechanisms remains controversial, especially under hyperglycemic condition. In this review, we discuss the role of different PKC isoforms in lipid regulation, oxidative stress, inflammatory response, and apoptosis. These intracellular events are linked to the pathogenesis of atherosclerosis in diabetes. PKC deletion or treatment with PKC inhibitors has been studied in the regulation of atherosclerotic plaque formation and evolution. Furthermore, some preclinical and clinical studies have indicated that PKCβ and PKCδ are potential targets for the treatment of diabetic vascular complications. The current review summarizes these multiple signaling pathways and cellular responses regulated by PKC activation and the potential therapeutic targets of PKC in diabetic complications.
Collapse
Affiliation(s)
- Chih-Feng Lien
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
14
|
Li H, Yu XH, Ou X, Ouyang XP, Tang CK. Hepatic cholesterol transport and its role in non-alcoholic fatty liver disease and atherosclerosis. Prog Lipid Res 2021; 83:101109. [PMID: 34097928 DOI: 10.1016/j.plipres.2021.101109] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a quickly emerging global health problem representing the most common chronic liver disease in the world. Atherosclerotic cardiovascular disease represents the leading cause of mortality in NAFLD patients. Cholesterol metabolism has a crucial role in the pathogenesis of both NAFLD and atherosclerosis. The liver is the major organ for cholesterol metabolism. Abnormal hepatic cholesterol metabolism not only leads to NAFLD but also drives the development of atherosclerotic dyslipidemia. The cholesterol level in hepatocytes reflects the dynamic balance between endogenous synthesis, uptake, esterification, and export, a process in which cholesterol is converted to neutral cholesteryl esters either for storage in cytosolic lipid droplets or for secretion as a major constituent of plasma lipoproteins, including very-low-density lipoproteins, chylomicrons, high-density lipoproteins, and low-density lipoproteins. In this review, we describe decades of research aimed at identifying key molecules and cellular players involved in each main aspect of hepatic cholesterol metabolism. Furthermore, we summarize the recent advances regarding the biological processes of hepatic cholesterol transport and its role in NAFLD and atherosclerosis.
Collapse
Affiliation(s)
- Heng Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 460106, China
| | - Xiang Ou
- Department of Endocrinology, the First Hospital of Changsha, Changsha, Hunan 410005, China
| | - Xin-Ping Ouyang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| | - Chao-Ke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
15
|
Chen W, Li L, Wang J, Zhang R, Zhang T, Wu Y, Wang S, Xing D. The ABCA1-efferocytosis axis: A new strategy to protect against atherosclerosis. Clin Chim Acta 2021; 518:1-8. [PMID: 33741356 DOI: 10.1016/j.cca.2021.02.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a disease process characterized by lipid accumulation and inflammation, is the main cause of coronary heart disease (CHD) and myocardial infarction (MI). Efferocytosis involves the clearance of apoptotic cells by phagocytes. Successful engulfment triggers the release of anti-inflammatory cytokines to suppress atherosclerosis. ABCA1 is a key mediator of cholesterol efflux to apoA-I for the generation of HDL-C in reverse cholesterol transport (RCT). Intriguingly, ABCA1 promotes not only cholesterol efflux but also efferocytosis. ABCA1 promotes efferocytosis by regulating the release of "find-me" ligands, including LPC, and the exposure, release, and expression of "eat-me" ligands, including PtdSer, ANXA1, ANXA5, MEGF10, and GULP1. ABCA1 has a pathway similar to TG2, which is an "eat-me" ligand. ABCA1 has the highest known homology to ABCA7, which controls efferocytosis as the engulfment and processing ligand. In addition, ABCA1 can form several regulatory feedback axes with ANXA1, MEGF10, GULP1, TNFα, and IL-6. Therefore, ABCA1 is the central factor that links cholesterol efflux and apoptotic cell clearance. Several drugs have been studied or approved for apoptotic cell clearance, such as CD47 antibody and PD1-/PD-L1 antibody. In this article, we review the role and mechanism of action of ABCA1 in efferocytosis and focus on new insights into the ABCA1-efferocytosis axis and its potential as a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Lu Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China.
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
16
|
Zhao Y, Qian Y, Sun Z, Shen X, Cai Y, Li L, Wang Z. Role of PI3K in the Progression and Regression of Atherosclerosis. Front Pharmacol 2021; 12:632378. [PMID: 33767629 PMCID: PMC7985550 DOI: 10.3389/fphar.2021.632378] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
Phosphatidylinositol 3 kinase (PI3K) is a key molecule in the initiation of signal transduction pathways after the binding of extracellular signals to cell surface receptors. An intracellular kinase, PI3K activates multiple intracellular signaling pathways that affect cell growth, proliferation, migration, secretion, differentiation, transcription and translation. Dysregulation of PI3K activity, and as aberrant PI3K signaling, lead to a broad range of human diseases, such as cancer, immune disorders, diabetes, and cardiovascular diseases. A growing number of studies have shown that PI3K and its signaling pathways play key roles in the pathophysiological process of atherosclerosis. Furthermore, drugs targeting PI3K and its related signaling pathways are promising treatments for atherosclerosis. Therefore, we have reviewed how PI3K, an important regulatory factor, mediates the development of atherosclerosis and how targeting PI3K can be used to prevent and treat atherosclerosis.
Collapse
Affiliation(s)
- Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xinyi Shen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
17
|
Zhang S, Li L, Wang J, Zhang T, Ye T, Wang S, Xing D, Chen W. Recent advances in the regulation of ABCA1 and ABCG1 by lncRNAs. Clin Chim Acta 2021; 516:100-110. [PMID: 33545111 DOI: 10.1016/j.cca.2021.01.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
Coronary heart disease (CHD) with atherosclerosis is the leading cause of death worldwide. ABCA1 and ABCG1 promote cholesterol efflux to suppress foam cell generation and reduce atherosclerosis development. Long noncoding RNAs (lncRNAs) are emerging as a unique group of RNA transcripts that longer than 200 nucleotides and have no protein-coding potential. Many studies have found that lncRNAs regulate cholesterol efflux to influence atherosclerosis development. ABCA1 is regulated by different lncRNAs, including MeXis, GAS5, TUG1, MEG3, MALAT1, Lnc-HC, RP5-833A20.1, LOXL1-AS1, CHROME, DAPK1-IT1, SIRT1 AS lncRNA, DYNLRB2-2, DANCR, LeXis, LOC286367, and LncOR13C9. ABCG1 is also regulated by different lncRNAs, including TUG1, GAS5, RP5-833A20.1, DYNLRB2-2, ENST00000602558.1, and AC096664.3. Thus, various lncRNAs are associated with the roles of ABCA1 and ABCG1 on cholesterol efflux in atherosclerosis regulation. However, some lncRNAs play dual roles in ABCA1 expression and atherosclerosis, and the functions of some lncRNAs in atherosclerosis have not been investigated in vivo. In this article, we review the roles of lncRNAs in atherosclerosis and focus on new insights into lncRNAs associated with the roles of ABCA1 and ABCG1 on cholesterol efflux and the potential of these lncRNAs as novel therapeutic targets in atherosclerosis.
Collapse
Affiliation(s)
- Shun Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Lu Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Ting Ye
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong 261053, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China.
| |
Collapse
|
18
|
Shan R, Liu N, Yan Y, Liu B. Apoptosis, autophagy and atherosclerosis: Relationships and the role of Hsp27. Pharmacol Res 2020; 166:105169. [PMID: 33053445 DOI: 10.1016/j.phrs.2020.105169] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a multifactorial chronic inflammatory disease of the arterial wall, and an important pathological basis of coronary heart disease. Endothelial cells, vascular smooth muscle cells, and macrophages play important roles in the development of atherosclerosis. Of note, apoptosis and autophagy, two types of programmed cell death, influence the development and progression of atherosclerosis via the modulation of such cells. The small heat shock protein Hsp27 is a multifunctional protein induced by various stress factors and has a protective effect on cells. A large number of studies have demonstrated that Hsp27 plays an important role in regulating apoptosis. Recently, some studies have suggested that Hsp27 also participates in the autophagic process. Moreover, Hsp27 is closely related to the occurrence and development of atherosclerosis. Here, we summarize the molecular mechanisms of apoptosis and autophagy and discuss their effects on endothelial cells, vascular smooth muscle cells, and macrophages in the context of atherosclerotic procession. We further explore the involvement of Hsp27 in apoptosis, autophagy, and atherosclerosis. We speculate that Hsp27 may exert its anti-atherosclerotic role via the regulation of apoptosis and autophagy; this may provide the basis for the development of new approaches for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Ruiting Shan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Youyou Yan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
19
|
Shen X, Zhang S, Guo Z, Xing D, Chen W. The crosstalk of ABCA1 and ANXA1: a potential mechanism for protection against atherosclerosis. Mol Med 2020; 26:84. [PMID: 32894039 PMCID: PMC7487582 DOI: 10.1186/s10020-020-00213-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis, characterized by the formation of fat-laden plaques, is a chronic inflammatory disease. ABCA1 promotes cholesterol efflux, reduces cellular cholesterol accumulation, and regulates anti-inflammatory activities in an apoA-I- or ANXA1-dependent manner. The latter activity occurs by mediating the efflux of ANXA1, which plays a critical role in anti-inflammatory effects, cholesterol transport, exosome and microparticle secretion, and apoptotic cell clearance. ApoA-I increases ANXA1 expression via the ERK, p38MAPK, AKT, and PKC pathways. ApoA-I regulates the signaling pathways by binding to ABCA1, suggesting that apoA-I increases ANXA1 expression by binding to ABCA1. Furthermore, ANXA1 may increase ABCA1 expression. ANXA1 increases PPARγ expression by modulating STAT6 phosphorylation. PPARγ also increases ANXA1 expression by binding to the promoter of ANXA1. Therefore, ABCA1, PPARγ, and ANXA1 may form a feedback loop and regulate each other. Interestingly, the ANXA1 needs to be externalized to the cell membrane or secreted into the extracellular fluids to exert its anti-inflammatory properties. ABCA1 transports ANXA1 from the cytoplasm to the cell membrane by regulating lipidization and serine phosphorylation, thereby mediating ANXA1 efflux, likely by promoting microparticle and exosome release. The direct role of ABCA1 expression and ANXA1 release in atherosclerosis has been unclear. In this review, we focus on the role of ANXA1 in atheroprogression and its novel interaction with ABCA1, which may be useful for providing basic knowledge for the development of novel therapeutic targets for atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Xin Shen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Shun Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Zhu Guo
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.,Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China. .,School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| |
Collapse
|
20
|
Chen W, Wu Y, Lu Q, Wang S, Xing D. Endogenous ApoA-I expression in macrophages: A potential target for protection against atherosclerosis. Clin Chim Acta 2020; 505:55-59. [PMID: 32092318 DOI: 10.1016/j.cca.2020.02.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/20/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022]
Abstract
ApoA-I is a major protein component of high-density lipoprotein (HDL) that is widely known for regulating cholesterol trafficking and inflammatory and immune responses and for protecting against atherosclerosis. ApoA-I is generally considered to be synthesized in the liver (hepatocytes) and small intestine (enterocytes). However, computer analysis of ApoA-I has shown that the ApoA-I gene may be expressed in not only hepatocytes and enterocytes but also monocyte-macrophage cells, dendritic cells (DCs) and T cells. ApoA-I expression has been detected in THP-1 monocytes and macrophages, peripheral blood mononuclear cells (PBMCs) from postmenopausal women, human PBMC-derived monocytes and macrophages, mouse peritoneal macrophages, etc. Endogenous ApoA-I in macrophages has anti-inflammatory and cholesterol efflux effects. However, our understanding of the detailed roles of macrophage-synthesized ApoA-I is still at an early stage and very limited. More experiments are needed to elucidate the exact roles of endogenous ApoA-I in macrophages. Several lines of evidence indicate that recombinant exogenous human ApoA-I in mouse macrophages increases cholesterol efflux and thus reduces atherosclerosis development. Considering the antiatherogenic effect of exogenous ApoA-I overexpression in mouse macrophages, better understanding the role and mechanisms underlying macrophage-synthesized ApoA-I in atherosclerosis will enable macrophage-synthesized ApoA-I therapy to open new avenues for reducing the risk of atherosclerosis.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Qi Lu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; Department of Oncology, Weifang Traditional Chinese Medicine Hospital, Weifang, Shandong 261041, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
21
|
Bacterial Pathogens Hijack the Innate Immune Response by Activation of the Reverse Transsulfuration Pathway. mBio 2019; 10:mBio.02174-19. [PMID: 31662455 PMCID: PMC6819659 DOI: 10.1128/mbio.02174-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Macrophages are professional immune cells that ingest and kill microbes. In this study, we show that different pathogenic bacteria induce the expression of cystathionine γ-lyase (CTH) in macrophages. This enzyme is involved in a metabolic pathway called the reverse transsulfuration pathway, which leads to the production of numerous metabolites, including cystathionine. Phagocytized bacteria use cystathionine to better survive in macrophages. In addition, the induction of CTH results in dysregulation of the metabolism of polyamines, which in turn dampens the proinflammatory response of macrophages. In conclusion, pathogenic bacteria can evade the host immune response by inducing CTH in macrophages. The reverse transsulfuration pathway is the major route for the metabolism of sulfur-containing amino acids. The role of this metabolic pathway in macrophage response and function is unknown. We show that the enzyme cystathionine γ-lyase (CTH) is induced in macrophages infected with pathogenic bacteria through signaling involving phosphatidylinositol 3-kinase (PI3K)/MTOR and the transcription factor SP1. This results in the synthesis of cystathionine, which facilitates the survival of pathogens within myeloid cells. Our data demonstrate that the expression of CTH leads to defective macrophage activation by (i) dysregulation of polyamine metabolism by depletion of S-adenosylmethionine, resulting in immunosuppressive putrescine accumulation and inhibition of spermidine and spermine synthesis, and (ii) increased histone H3K9, H3K27, and H3K36 di/trimethylation, which is associated with gene expression silencing. Thus, CTH is a pivotal enzyme of the innate immune response that disrupts host defense. The induction of the reverse transsulfuration pathway by bacterial pathogens can be considered an unrecognized mechanism for immune escape.
Collapse
|
22
|
IL-8 negatively regulates ABCA1 expression and cholesterol efflux via upregulating miR-183 in THP-1 macrophage-derived foam cells. Cytokine 2019; 122:154385. [DOI: 10.1016/j.cyto.2018.04.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 04/18/2018] [Accepted: 04/20/2018] [Indexed: 02/08/2023]
|
23
|
He XW, Zhao Y, Shi YH, Zhao R, Liu YS, Hu Y, Zhuang MT, Wu YL, Li GF, Yin JW, Cui GH, Liu JR. DNA Methylation Analysis Identifies Differentially Methylated Sites Associated with Early-Onset Intracranial Atherosclerotic Stenosis. J Atheroscler Thromb 2019; 27:71-99. [PMID: 31142690 PMCID: PMC6976716 DOI: 10.5551/jat.47704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Studies have suggested that genetic and environmental factors do not account for all risks and mechanisms of intracranial atherosclerotic stenosis (ICAS). DNA methylation may play a role in the progression of ICAS. Methods: DNA methylation profiles of peripheral blood leucocytes from 7 patients with early-onset ICAS and 7 perfectly matched controls were interrogated for the first time using the Illumina Infinium Human MethylationEPIC BeadChip. Afterward, functional analysis for differentially methylated genes was conducted. In addition, pyrosequencing verification was performed in an independent cohort comprising 21 patients with earlyonset ICAS and 21 age- and gender-matched controls. Results: A total of 318 cytosine-phosphate-guanine sites were found to be differentially methylated based on the established standards. Functional analysis annotated differentially methylated sites to atherosclerosis-related processes and pathways, such as the negative regulation of hydrolase activity (GO 0051346), type II diabetes mellitus (KEGG hsa04930), and the insulin signaling pathway (KEGG hsa04910). In addition, a differentially methylated site was also validated, cg22443212 in gene Rnf213, which showed significant hypermethylation in patients with early-onset ICAS compared with controls 59.56% (49.77%, 88.55%) vs. 44.65% (25.07%, 53.21%), respectively; P = 0.010). Receiver operating characteristic curve analysis showed that the area under the curve value of cg22443212 was 0.744 (95% confidence interval, 0.586–0.866; P = 0.002). Conclusions: We revealed that altered DNA methylation may play a role in the occurrence and development of ICAS. These results provided new epigenetic insights into ICAS.
Collapse
Affiliation(s)
- Xin-Wei He
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Ying Zhao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Yan-Hui Shi
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Rong Zhao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Yi-Sheng Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Yue Hu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Mei-Ting Zhuang
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Yi-Lan Wu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Ge-Fei Li
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Jia-Wen Yin
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Guo-Hong Cui
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| | - Jian-Ren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine.,Clinical Research Center, Shanghai Jiao Tong University School of Medicine
| |
Collapse
|
24
|
Lv Y, Yang J, Gao A, Sun S, Zheng X, Chen X, Wan W, Tang C, Xie W, Li S, Guo D, Peng T, Zhao G, Zhong L. Sortilin promotes macrophage cholesterol accumulation and aortic atherosclerosis through lysosomal degradation of ATP-binding cassette transporter A1 protein. Acta Biochim Biophys Sin (Shanghai) 2019; 51:471-483. [PMID: 30950489 DOI: 10.1093/abbs/gmz029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Indexed: 11/13/2022] Open
Abstract
Sortilin is closely associated with hyperlipidemia and the risk of atherosclerosis (AS). The role of sortilin and the underlying mechanism in peripheral macrophage are not fully understood. In this study, we investigated the effect of macrophage sortilin on ATP-binding cassette transporter A1 (ABCA1) expression, ABCA1-mediated cholesterol efflux, and aortic AS. Macrophage sortilin expression was upregulated by oxidized low-density lipoproteins (ox-LDLs) in both concentration- and time-dependent manners. Its expression reached the peak level when cells were incubated with 50 μg/ml ox-LDL for 24 h. Overexpression of sortilin in macrophage reduced cholesterol efflux, leading to an increase in intracellular total cholesterol, free cholesterol, and cholesterol ester. Sortilin was found to bind with ABCA1 protein and suppress macrophage ABCA1 expression, resulting in a decrease in cholesterol efflux from macrophages. The inhibitory effect of sortilin in cholesterol efflux was partially reversed by treatment with chloroquine, a lysosomal inhibitor. On the contrary, the ABCA1 protein level and ABCA1-mediated cholesterol efflux is increased by sortilin short hairpin RNA transfection. The fecal and biliary cholesterol 3H-sterol from cholesterol-laden mouse peritoneal macrophage was reduced by sortilin overexpression through lentivirus vector (LV)-sortilin in low-density lipoprotein receptor knockout mice, which was prevented by co-treatment with chloroquine. Treatment with LV-sortilin reduced plasma high-density lipoprotein and increased plasma ox-LDL levels. Accordingly, aortic lipid deposition and plaque area were exacerbated, and ABCA1 expression was reduced in mice in response to infection with LV-sortilin alone. These effects of LV-sortilin were partially reversed by chloroquine. Sortilin enhances lysosomal degradation of ABCA1 protein and suppresses ABCA1-mediated cholesterol efflux from macrophages, leading to foam cell formation and AS development.
Collapse
Affiliation(s)
- Yuncheng Lv
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Jing Yang
- Clinical Medical Research Institute of the First Affiliated Hospital, University of South China, Hengyang, China
| | - Anbo Gao
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Sha Sun
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Xilong Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Canada
| | - Xi Chen
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Wei Wan
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Chaoke Tang
- Institute of Cardiovascular Research, Medical Research Center, University of South China, Hengyang, China
| | - Wei Xie
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Suyun Li
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Dongming Guo
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Tianhong Peng
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| | - Guojun Zhao
- Department of Histology and Embryology, Guilin Medical University, Guilin, China
| | - Liyuan Zhong
- Clinical Anatomy and Reproductive Medicine Application Institute, University of South China, Hengyang, China
| |
Collapse
|
25
|
Is there any association between Serum anti-HSP27 antibody level and the presence of metabolic syndrome; population based case-control study. REV ROMANA MED LAB 2019. [DOI: 10.2478/rrlm-2019-0016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Abstract
Background: Heat shock protein 27 (HSP27) is an intracellular chaperone constitutively expressed in many cell types including cardio myocytes and endothelial cells. Circulating levels of HSP27 and anti-HSP27 antibody are higher in patients with CVD. Anti-HSP27 antibody concentrations were also reported to be increased in atherogenesis. We aimed to evaluate serum anti-HSP27 antibody titers in individuals with, or without, MetS in the MASHAD study cohort with large sample size in 6,568 subjects.
Methods: Participants with MetS were identified from MASHAD cohort (n=3358) using the IDF criteria, and the control group were those individuals who did not meet these criteria (n=3210). In-house enzyme-linked immune sorbent assay (ELISA) method was used for measuring Anti-HSP27 antibody levels. The two groups were matched for age, sex and smoking habit.
Results: As expected, there were significant differences in height (p= 0.004), waist and hip circumference, weight, BMI, systolic and diastolic blood pressure, TGs, TC, HDL-C, Hs-CRP, glucose, with the presence of diabetes mellitus, hypertension, hyperlipidemia (p<0.001) between the two groups. Serum HSP27 antibody titers did not show significant difference between the groups with and without metabolic syndrome (p= 0.740).
Conclusion: In conclusion, our results revealed serum anti-HSP27 antibody titers were not statistically different between individuals with and without MetS. However, it is possible that drug treatment may affect antibody titers and confound our findings in this population sample..
Collapse
|
26
|
Lynn EG, Lhoták Š, Lebeau P, Byun JH, Chen J, Platko K, Shi C, O'Brien RE, Austin RC. 4‐Phenylbutyrate protects against atherosclerotic lesion growth by increasing the expression of HSP25 in macrophages and in the circulation of
Apoe
−/−
mice. FASEB J 2019; 33:8406-8422. [DOI: 10.1096/fj.201802293rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Edward G. Lynn
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Šárka Lhoták
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Paul Lebeau
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Jae Hyun Byun
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Jack Chen
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Khrystyna Platko
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| | - Chunhua Shi
- Department of Cardiac SciencesLibin Cardiovascular Institute of AlbertaCumming School of MedicineUniversity of Calgary Calgary Alberta Canada
| | - R. Edward O'Brien
- Department of Cardiac SciencesLibin Cardiovascular Institute of AlbertaCumming School of MedicineUniversity of Calgary Calgary Alberta Canada
| | - Richard C. Austin
- Division of NephrologyDepartment of MedicineMcMaster UniversityThe Research Institute of St. Joe's HamiltonHamilton Centre for Kidney Research Hamilton Ontario Canada
| |
Collapse
|