1
|
Johnson E, Albakri JS, Allemailem KS, Sultan A, Alwanian WM, Alrumaihi F, Almansour NM, Aldakheel FM, Khalil FMA, Abduallah AM, Smith O. Mitochondrial dysfunction and calcium homeostasis in heart failure: Exploring the interplay between oxidative stress and cardiac remodeling for future therapeutic innovations. Curr Probl Cardiol 2024; 50:102968. [PMID: 39653095 DOI: 10.1016/j.cpcardiol.2024.102968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
Heart failure (HF) is a multifaceted clinical syndrome characterized by the heart's inability to pump sufficient blood to meet the body's metabolic demands. It arises from various etiologies, including myocardial injury, hypertension, and valvular heart disease. A critical aspect of HF pathophysiology involves mitochondrial dysfunction, particularly concerning calcium (Ca2+) homeostasis and oxidative stress. This review highlights the pivotal role of excess mitochondrial Ca2+ in exacerbating oxidative stress, contributing significantly to HF progression. Novel insights are provided regarding the mechanisms by which mitochondrial Ca2+ overload leads to increased production of reactive oxygen species (ROS) and impaired cellular function. Despite this understanding, key gaps in research remain, particularly in elucidating the complex interplay between mitochondrial dynamics and oxidative stress across different HF phenotypes. Furthermore, therapeutic strategies targeting mitochondrial dysfunction are still in their infancy, with limited applications in clinical practice. By summarizing recent findings and identifying these critical research gaps, this review aims to pave the way for innovative therapeutic approaches that improve the management of heart failure, ultimately enhancing patient outcomes through targeted interventions.
Collapse
Affiliation(s)
- Emily Johnson
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | | | - Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Abdulaziz Sultan
- Family Medicine Senior Registrar, Ministry of Health, Saudi Arabia
| | - Wanian M Alwanian
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Nahlah Makki Almansour
- Department of Biology, College of Science, University of Hafr Al Batin, Hafr Al Batin 31991, Saudi Arabia
| | - Fahad M Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Fatma Mohamed Ameen Khalil
- King Khalid University, Applied College, Unit of health specialties, basic sciences and their applications, Mohayil Asir Abha, 61421, Saudi Arabia
| | - Alduwish Manal Abduallah
- Department of Biology, College of Science and Humanities in Al-Kharj, Prince Sattam Bin Abdulaziz University, Alkarj 11942, Saudi Arabia
| | - Oliver Smith
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
2
|
Zhang M, Li J, Hu W. The complex interplay between ferroptosis and atherosclerosis. Biomed Pharmacother 2024; 178:117183. [PMID: 39079265 DOI: 10.1016/j.biopha.2024.117183] [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: 06/06/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024] Open
Abstract
Atherosclerosis, characterized by the accumulation of plaque within the arterial walls, is an intricate cardiovascular disease that often results in severe health issues. Recent studies have emphasized the importance of ferroptosis, a controlled type of cell death dependent on iron, as a critical factor in this disease state. Ferroptosis, distinguished by its reliance on iron and the accumulation of lipid hydroperoxides, offers a unique insight into the pathology of atherosclerotic lesions. This summary encapsulates the current knowledge of the intricate role ferroptosis plays in the onset and progression of atherosclerosis. It explores the molecular processes through which lipid peroxidation and iron metabolism contribute to the development of atheromatous plaques and evaluates the possibility of utilizing ferroptosis as a novel treatment approach for atherosclerosis. By illuminating the intricate relationship between ferroptosis-related processes and atherosclerosis, this review paves the way for future clinical applications and personalized medicine approaches aimed at alleviating the effects of atherosclerosis.
Collapse
Affiliation(s)
- Mao Zhang
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiangping Li
- Department of Oncological Radiotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Hu
- Department of Vascular Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
3
|
Li Y, Du L, Meng L, Lv C, Tian X. High expression of CASP1 induces atherosclerosis. Medicine (Baltimore) 2024; 103:e37616. [PMID: 38640260 PMCID: PMC11030018 DOI: 10.1097/md.0000000000037616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/17/2024] [Accepted: 02/23/2024] [Indexed: 04/21/2024] Open
Abstract
Atherosclerosis is a chronic, progressive vascular disease. The relationship between CASP1 gene expression and atherosclerosis remains unclear. The atherosclerosis dataset GSE132651 and GSE202625 profiles were downloaded from gene expression omnibus. Differentially expressed genes (DEGs) were screened. The construction and analysis of protein-protein interaction network, functional enrichment analysis, gene set enrichment analysis, and Comparative Toxicogenomics Database analysis were performed. Gene expression heatmap was drawn. TargetScan was used to screen miRNAs that regulate central DEG. 47 DEGs were identified. According to gene ontology analysis, they were mainly enriched in the regulation of stimulus response, response to organic matter, extracellular region, extracellular region, and the same protein binding. Kyoto Encyclopedia of Gene and Genome analysis results showed that the target cells were mainly enriched in the PI3K-Akt signaling pathway, Ras signaling pathway, and PPAR signaling pathway. In the enrichment project of Metascape, vascular development, regulation of body fluid levels, and positive regulation of cell motility can be seen in the gene ontology enrichment project. Eleven core genes (CASP1, NLRP3, MRC1, IRS1, PPARG, APOE, IL13, FGF2, CCR2, ICAM1, HIF1A) were obtained. IRS1, PPARG, APOE, FGF2, CCR2, and HIF1A genes are identified as core genes. Gene expression heatmap showed that CASP1 was highly expressed in atherosclerosis samples and low expressed in normal samples. NLRP3, MRC1, IRS1, PPARG, APOE, IL13, FGF2, CCR2, ICAM1, HIF1A were low expressed in atherosclerosis samples. CTD analysis showed that 5 genes (CASP1, NLRP3, CCR2, ICAM1, HIF1A) were found to be associated with pneumonia, inflammation, cardiac enlargement, and tumor invasiveness. CASP1 gene is highly expressed in atherosclerosis. The higher the CASP1 gene, the worse the prognosis.
Collapse
Affiliation(s)
- Yongchao Li
- Department of Cardiac Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Lihong Du
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Lingbing Meng
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Chao Lv
- Department of neurology, Pizhou Hospital Affiliated to Xuzhou Medical University, Pizhou People's Hospital, Pizhou, Jiangsu Province, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
4
|
Li X, Cai P, Tang X, Wu Y, Zhang Y, Rong X. Lactylation Modification in Cardiometabolic Disorders: Function and Mechanism. Metabolites 2024; 14:217. [PMID: 38668345 PMCID: PMC11052226 DOI: 10.3390/metabo14040217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiovascular disease (CVD) is recognized as the primary cause of mortality and morbidity on a global scale, and developing a clear treatment is an important tool for improving it. Cardiometabolic disorder (CMD) is a syndrome resulting from the combination of cardiovascular, endocrine, pro-thrombotic, and inflammatory health hazards. Due to their complex pathological mechanisms, there is a lack of effective diagnostic and treatment methods for cardiac metabolic disorders. Lactylation is a type of post-translational modification (PTM) that plays a regulatory role in various cellular physiological processes by inducing changes in the spatial conformation of proteins. Numerous studies have reported that lactylation modification plays a crucial role in post-translational modifications and is closely related to cardiac metabolic diseases. This article discusses the molecular biology of lactylation modifications and outlines the roles and mechanisms of lactylation modifications in cardiometabolic disorders, offering valuable insights for the diagnosis and treatment of such conditions.
Collapse
Affiliation(s)
- Xu Li
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingdong Cai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xinyuan Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yingzi Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xianglu Rong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangzhou 510006, China; (X.L.); (P.C.); (X.T.); (Y.W.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, Guangzhou 510006, China
- Key Unit of Modulating Liver to Treat Hyperlipemia SATCM, State Administration of Traditional Chinese Medicine, Guangzhou 510006, China
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
5
|
Reiter RJ, Sharma R, Chuffa LGDA, Simko F, Dominguez-Rodriguez A. Mitochondrial Melatonin: Beneficial Effects in Protecting against Heart Failure. Life (Basel) 2024; 14:88. [PMID: 38255703 PMCID: PMC10820220 DOI: 10.3390/life14010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/01/2024] [Accepted: 01/03/2024] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease is the cause of physical infirmity and thousands of deaths annually. Typically, during heart failure, cardiomyocyte mitochondria falter in terms of energy production and metabolic processing. Additionally, inflammation and the accumulation of non-contractile fibrous tissue contribute to cardiac malfunction. Melatonin, an endogenously produced molecule, experimentally reduces the initiation and progression of atherosclerotic lesions, which are often the basis of coronary artery disease. The current review critically analyzes published data related to the experimental use of melatonin to forestall coronary artery pathologies. Collectively, these studies document melatonin's anti-atherosclerotic actions in reducing LDL oxidation and triglyceride levels, lowering endothelial malfunction, limiting adhesion molecule formation, preventing macrophage polarization to the M1 pro-inflammatory phenotype, changing cellular metabolism, scavenging destructive reactive oxygen species, preventing the proliferation and invasion of arterial smooth muscle cells into the lesioned area, restricting the ingrowth of blood vessels from the vasa vasorum, and solidifying the plaque cap to reduce the chance of its rupture. Diabetic hyperglycemia, which aggravates atherosclerotic plaque formation, is also inhibited by melatonin supplementation in experimental animals. The potential value of non-toxic melatonin as a possible inhibitor of cardiac pathology in humans should be seriously considered by performing clinical trials using this multifunctional molecule.
Collapse
Affiliation(s)
- Russel J. Reiter
- Department of Cell Systems and Anatomy, Long School of Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Ramaswamy Sharma
- Applied Biomedical Sciences, School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology-IBB/UNESP, Institute of Biosciences of Botucatu, Universidade Estadual Paulista, Botucatu 18618-689, São Paulo, Brazil;
| | - Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, 81108 Bratislava, Slovakia;
| | | |
Collapse
|
6
|
Yang Y, Li JH, Yao BC, Chen QL, Jiang N, Wang LQ, Guo ZG. NDUFB11 and NDUFS3 play a role in atherosclerosis and chronic stress. Aging (Albany NY) 2023; 15:8026-8043. [PMID: 37642954 PMCID: PMC10496984 DOI: 10.18632/aging.204947] [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: 05/11/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023]
Abstract
OBJECTIVE Atherosclerosis is characterized by the formation of fibrofatty plaques in the intima of arteries, resulting in thickening of the vessel wall and narrowing of the lumen. Chronic stress refers to individuals in a state of long-term chronic stress. However, the relationship between NDUFB11 and NDUFS3 and atherosclerosis and chronic stress is unclear. METHOD The atherosclerosis with chronic stress group data file was used. DEGs were screened and WGCNA was performed. Construction and analysis of PPI Network. Functional enrichment analysis, GSEA, gene expression heatmap, immune infiltration analysis and mRNA analysis were performed. CTD was used to find diseases most related to core genes. WB was performed. TargetScan was used to screen miRNAs of DEGs. RESULTS 1708 DEGs were identified. According to GO analysis, they were mainly enriched in catabolic processes, organic acid metabolism processes, carboxylic acid metabolism processes. KEGG analysis showed that they were mainly enriched in metabolic pathways, fatty acid metabolism, pentose phosphate pathway, glycolysis / gluconeogenesis, fructose and mannose metabolism. Gene expression heatmap showed that the core genes (NDUFB11, NDUFS3) were lowly expressed in samples of those with atherosclerosis accompanied by chronic stress and highly expressed in the normal samples. NDUFB11 and NDUFS3 were associated with necrosis, hyperplasia, inflammation, renal disease, weight loss, memory impairment, and cognitive impairment. WB showed that the expression level of NDUFS3 in atherosclerosis and chronic stress was lower than that in control group. CONCLUSIONS NDUFB11 and NDUFS3 are underexpressed in atherosclerosis and chronic stress; the lower NDUFB11 and NDUFS3 levels, the worse the prognosis.
Collapse
Affiliation(s)
- Yin Yang
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Jing-Hui Li
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Bo-Chen Yao
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Qing-Liang Chen
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Nan Jiang
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Lian-Qun Wang
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| | - Zhi-Gang Guo
- Clinical School of Thoracic, Tianjin Medical University, Tianjin Chest Hospital, Jinnan, Tianjin 300222, P.R. China
| |
Collapse
|
7
|
Xu J, Yang Y, Li X, Ding S, Zheng L, Xiong C, Yang Y. Pleiotropic activities of succinate: The interplay between gut microbiota and cardiovascular diseases. IMETA 2023; 2:e124. [PMID: 38867936 PMCID: PMC10989957 DOI: 10.1002/imt2.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2024]
Abstract
Cardiovascular diseases (CVDs) continue to be a significant contributor to global mortality, imposing a substantial burden and emphasizing the urgent need for disease control to save lives and prevent disability. With advancements in technology and scientific research, novel mechanisms underlying CVDs have been uncovered, leading to the exploration of promising treatment targets aimed at reducing the global burden of the disease. One of the most intriguing findings is the relationship between CVDs and gut microbiota, challenging the traditional understanding of CVDs mechanisms and introducing the concept of the gut-heart axis. The gut microbiota, through changes in microbial compositions and functions, plays a crucial role in influencing local and systemic effects on host physiology and disease development, with its metabolites acting as key regulators. In previous studies, we have emphasized the importance of specific metabolites such as betaine, putrescine, trimethylamine oxide, and N,N,N-trimethyl-5-aminovaleric acid in the potential treatment of CVDs. Particularly noteworthy is the gut microbiota-associated metabolite succinate, which has garnered significant attention due to its involvement in various pathophysiological pathways closely related to CVDs pathogenesis, including immunoinflammatory responses, oxidative stress, and energy metabolism. Furthermore, we have identified succinate as a potential biomarker, highlighting its therapeutic feasibility in managing aortic dissection and aneurysm. This review aims to comprehensively outline the characteristics of succinate, including its biosynthetic process, summarize the current evidence linking it to CVDs causation, and emphasize the host-microbial crosstalk involved in modulating CVDs. The insights presented here offer a novel paradigm for future management and control of CVDs.
Collapse
Affiliation(s)
- Jing Xu
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yicheng Yang
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xin Li
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain ProtectionThe Capital Medical UniversityBeijingChina
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain ProtectionThe Capital Medical UniversityBeijingChina
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Health Science CenterPeking UniversityBeijingChina
| | - Changming Xiong
- Respiratory and Pulmonary Vascular Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
8
|
The Mechanism Study of Moxa Combustion Products on Regulating Vascular Endothelial Function in Atherosclerotic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1303978. [PMID: 36225192 PMCID: PMC9550439 DOI: 10.1155/2022/1303978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/30/2022] [Indexed: 11/06/2022]
Abstract
Objective To evaluate the anti-atherogenic effect of moxa combustion products (MCPs) and whether it is mediated through improving the vascular endothelial function in ApoE−/− mice. Methods A total of 60 male ApoE−/− mice were randomly divided into the moxa smoke (MS) group, filtered moxa smoke (FMS) group, moxa floss volatile (MFV) group, essential oil of Artemisia argyi (EOAA) group, and model group (n = 12/group), while 12 male C57BL/6 mice were used as the control group. The six groups were intervened for 20 min/day, 6 days/week. After 14 weeks of intervention, the mice were euthanized and their blood lipids were measured. The aortic roots and thoracic aortas were collected for haematoxylin and eosin (HE) or Oil Red O staining, respectively. The contents of AMPK, PI3K, Akt, and eNOS mRNA in the thoracic aortas were examined by RT-qPCR. Results The MS group and FMS group showed significantly lower plaque area percentage in the aortic roots and thoracic aortas and higher contents of AMPK-mRNA and eNOS-mRNA in the thoracic aortas compared with the model group. Conclusion MS and FMS equally suppressed the progression of atherosclerotic lesions in ApoE−/− mice. It was suggested that the particulate matter in MS may not be the key components of moxibustion.
Collapse
|
9
|
Xu R, Yuan W, Wang Z. Advances in Glycolysis Metabolism of Atherosclerosis. J Cardiovasc Transl Res 2022; 16:476-490. [PMID: 36068370 DOI: 10.1007/s12265-022-10311-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022]
Abstract
Glycolysis is an important way for various cells such as vascular wall endothelial cells, smooth muscle cells, macrophages, and other cells to obtain energy. In pathological conditions, it can participate in the process of AS by regulating lipid deposition, calcification, angiogenesis in plaques, etc., together with its metabolite lactic acid. Recent studies have shown that lactate-related lactylation modifications are ubiquitous in the human proteome and are involved in the regulation of various inflammatory diseases. Combined with the distribution and metabolic characteristics of cells in the plaque in the process of AS, glycolysis-lactate-lactylation modification may be a new entry point for targeted intervention in atherosclerosis in the future. Therefore, this article intends to elaborate on the role and mechanism of glycolysis-lactate-lactylation modification in AS, as well as the opportunities and challenges in targeted therapy, hoping to bring some help to relevant scholars in this field. In atherosclerosis, glycolysis, lactate, and lactylation modification as a metabolic sequence affect the functions of macrophages, smooth muscle cells, endothelial cells, lymphocytes, and other cells and interfere with processes such as vascular calcification and intraplaque neovascularization to influence the progression of atherosclerosis.
Collapse
Affiliation(s)
- Ruhan Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
10
|
LINC00460 Stimulates the Proliferation of Vascular Endothelial Cells by Downregulating miRNA-24-3p. DISEASE MARKERS 2022; 2022:2524156. [PMID: 35222741 PMCID: PMC8881155 DOI: 10.1155/2022/2524156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/23/2021] [Indexed: 11/18/2022]
Abstract
Objective To clarify the effect of LINC00460 on mediating the proliferative ability of vascular endothelial cells (ECs) by targeting microRNA-24-3p (miRNA-24-3p), thus influencing the progression of atherosclerotic diseases. Methods Relative levels of LINC00460 and miRNA-24-3p in ECs induced with different doses of ox-LDL (oxidized low density lipoprotein) for different time points were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Influences of LINC00460 and miRNA-24-3p on the viability of ECs were assessed by Cell Counting Kit-8 (CCK-8) and 5-ethynyl-2′-deoxyuridine (EdU) assay. Through dual-luciferase reporter gene assay, the binding between LINC00460 and miRNA-24-3p was evaluated. At last, rescue experiments were performed to identify the function of the LINC00460/miRNA-24-3p axis in regulating the proliferative ability of ECs. Results LINC00460 was upregulated after ox-LDL treatment in a dose- and time-dependent manner. Viability of ECs gradually increased with the prolongation of ox-LDL treatment and the treatment of increased dose. The overexpression of LINC00460 enhanced the viability and EdU-positive rate in ECs treated with ox-LDL. miRNA-24-3p was the direct target of LINC00460, which was negatively regulated by LINC00460. miRNA-24-3p was downregulated with the prolongation of ox-LDL treatment. The overexpression of miRNA-24-3p could reverse the effect of LINC00460 on regulating the proliferative ability of ECs. Conclusions LINC00460 regulates the proliferative ability of ECs and thus the occurrence and development of coronary atherosclerotic diseases by targeting miRNA-24-3p.
Collapse
|
11
|
Song D, Li M, Yu X, Wang Y, Fan J, Yang W, Yang L, Li H. The Molecular Pathways of Pyroptosis in Atherosclerosis. Front Cell Dev Biol 2022; 10:824165. [PMID: 35237603 PMCID: PMC8884404 DOI: 10.3389/fcell.2022.824165] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease seriously endangering human health, whose occurrence and development is related to many factors. Pyroptosis is a recently identified novel programmed cell death associated with an inflammatory response and involved in the formation and progression of AS by activating different signaling pathways. Protein modifications of the sirtuin family and microRNAs (miRNAs) can directly or indirectly affect pyroptosis-related molecules. It is important to link atherosclerosis, thermogenesis and molecular modifications. This article will systematically review the molecular pathways of pyroptosis in AS, which can provide a new perspective for AS prevention and treatment.
Collapse
Affiliation(s)
- Dan Song
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Manman Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xue Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yuqin Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jiaying Fan
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Wei Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, China
- *Correspondence: Hong Li, ; Liming Yang,
| | - Hong Li
- Department of Pathophysiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
- *Correspondence: Hong Li, ; Liming Yang,
| |
Collapse
|
12
|
Zhang S, Liang Y, Li L, Chen Y, Wu P, Wei D. Succinate: A Novel Mediator to Promote Atherosclerotic Lesion Progression. DNA Cell Biol 2022; 41:285-291. [PMID: 35138943 DOI: 10.1089/dna.2021.0345] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Succinate is an important intermediate product of mitochondrial energy metabolism. Recent studies revealed that beyond its known traditional metabolic functions, succinate plays important roles in signal transduction, immunity, inflammation, and posttranslational modification. Recent studies showed that patients and mouse models with cardiovascular disease have high levels of serum succinate and succinate accumulation. Atherosclerosis (As) is the pathological basis of cardiovascular and peripheral vascular diseases, such as coronary heart disease, cerebral infarction, and peripheral vascular disease, and is a major factor affecting human health. This article reviews the progression of succinate in As diseases and its underlying mechanisms.
Collapse
Affiliation(s)
- Shulei Zhang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| | - Yamin Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| | - Lu Li
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| | - Yanmei Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| | - Peng Wu
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| | - Dangheng Wei
- Key Laboratory for Arteriosclerology of Hunan Province, Institute of Cardiovascular Disease, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, University of South China, Hengyang, China
| |
Collapse
|
13
|
Razeghian-Jahromi I, Karimi Akhormeh A, Razmkhah M, Zibaeenezhad MJ. Immune system and atherosclerosis: Hostile or friendly relationship. Int J Immunopathol Pharmacol 2022; 36:3946320221092188. [PMID: 35410514 PMCID: PMC9009140 DOI: 10.1177/03946320221092188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Coronary artery disease has remained a major health challenge despite enormous
progress in prevention, diagnosis, and treatment strategies. Formation of
atherosclerotic plaque is a chronic process that is developmentally influenced
by intrinsic and extrinsic determinants. Inflammation triggers atherosclerosis,
and the fundamental element of inflammation is the immune system. The immune
system involves in the atherosclerosis process by a variety of immune cells and
a cocktail of mediators. It is believed that almost all main components of this
system possess a profound contribution to the atherosclerosis. However, they
play contradictory roles, either protective or progressive, in different stages
of atherosclerosis progression. It is evident that monocytes are the first
immune cells appeared in the atherosclerotic lesion. With the plaque growth,
other types of the immune cells such as mast cells, and T lymphocytes are
gradually involved. Each cell releases several cytokines which cause the
recruitment of other immune cells to the lesion site. This is followed by
affecting the expression of other cytokines as well as altering certain
signaling pathways. All in all, a mix of intertwined interactions determine the
final outcome in terms of mild or severe manifestations, either clinical or
subclinical. Therefore, it is of utmost importance to precisely understand the
kind and degree of contribution which is made by each immune component in order
to stop the growing burden of cardiovascular morbidity and mortality. In this
review, we present a comprehensive appraisal on the role of immune cells in the
atherosclerosis initiation and development.
Collapse
Affiliation(s)
- Iman Razeghian-Jahromi
- Cardiovascular Research Center, 571605Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Karimi Akhormeh
- Cardiovascular Research Center, 571605Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, 48435Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
14
|
Li Z, Xu C, Sun D. MicroRNA-488 serves as a diagnostic marker for atherosclerosis and regulates the biological behavior of vascular smooth muscle cells. Bioengineered 2021; 12:4092-4099. [PMID: 34288824 PMCID: PMC8806555 DOI: 10.1080/21655979.2021.1953212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis (AS) is one of the main causes of cerebral infarction. Researches on AS mainly focus on the gene level, among which microRNA is the research hotspot nowadays. This study investigated the diagnostic value of aberrant serum miR-488 in AS patients, and further explored the effect of abnormally expressed miR-488 on the biological behavior of vascular smooth muscle (VSMCs) cells by cell transfection. The qRT-PCR was used to investigate the expression level of miR-488 in 125 AS patients and 60 healthy controls. The diagnostic value of miR-488 was analyzed by the receiver operator characteristic (ROC) curve. CCK-8 and Transwell assays were used to detect the ability of miR-488 on the proliferation and migration ability of VSMCs cells. Serum expression of miR-488 in AS patients was higher than that in healthy controls. The expression level of miR-488 was significantly positively correlated with the Carotid Intima-Media Thickness (CIMT) value. The AUC of the ROC curve was 0.892, specificity was 99.3%, and sensitivity was 77.6%. In VSMCs cells, overexpression of miR-488 significantly promoted the proliferation and migration ability. The high expression of miR-488 is a good diagnostic marker for AS. The upregulation of miR-488 promotes VSMCs cell proliferation, and migration, which may provide a new theory for the treatment of AS.
Collapse
Affiliation(s)
- Zhen Li
- Department of Cardiology, Shengli Oilfield Central Hospital, Shandong, China
| | - Congjian Xu
- Department of Cardiology, Shengli Oilfield Central Hospital, Shandong, China
| | - Di Sun
- Department of Cardiology, Shengli Oilfield Central Hospital, Shandong, China
| |
Collapse
|
15
|
Liu Z, Li J, Lin S, Wu Y, He D, Qu P. PI3K regulates the activation of NLRP3 inflammasome in atherosclerosis through part-dependent AKT signaling pathway. Exp Anim 2021; 70:488-497. [PMID: 34162771 PMCID: PMC8614019 DOI: 10.1538/expanim.21-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PI3K is a downstream target of multiple cell-surface receptors, which acts as a crucial modulator of both cell polarization and survival. PI3K/AKT signaling pathway is commonly involved in cancer, atherosclerosis, and other diseases. However, its role in cardiovascular diseases, especially in atherosclerosis, remains to be further investigated. To determine the effect of PI3K/AKT signaling pathway on cellular inflammatory response and oxidative stress, PI3K inhibitor (GDC0941) and AKT inhibitor (MK2206) were used. First, THP-1 cells were incubated with ox-LDL (100 µg/ml) to establish an in vitro atherosclerosis model. The inflammatory factors and foam cell formation were then evaluated to ascertain and compare the effects of PI3K and AKT inhibition. ApoE−/− mice fed a high-fat diet were used to assess the roles of PI3K and AKT in aortic plaque formation. Our results showed that the inhibition of PI3K or AKT could suppress the activation of NLRP3, decreased the expression levels of p-p65/p65 and reduced the production of mitochondrial reaction oxygen species (mitoROS) in THP-1 cells. Inhibition of PI3K or AKT could also reduced atherosclerosis lesion and plaque area, and decreased the levels of NLRP3 and IL-1β in ApoE−/− mice. The effect of PI3K inhibition was more significant than AKT. Therefore, PI3K inhibition can retard the progress of atherosclerosis. Besides, there may be other AKT-independent pathways that regulate the formation of atherosclerosis.
Collapse
Affiliation(s)
- Zhenzhu Liu
- Department of Cardiology, 2nd Affiliated Hospital of Dalian Medical University
| | - Jing Li
- Department of Cardiology, 2nd Affiliated Hospital of Dalian Medical University
| | - Shu Lin
- Department of Cardiology, The University of Hong Kong-Shenzhen Hospital
| | - Yuhang Wu
- Department of Cardiology, 2nd Affiliated Hospital of Dalian Medical University
| | - Dan He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education
| | - Peng Qu
- Department of Cardiology, 2nd Affiliated Hospital of Dalian Medical University
| |
Collapse
|
16
|
Chen X, Wang D, Qian L. LncRNA Fetal-Lethal Noncoding Developmental Regulatory RNA (FENDRR) Suppresses Cell Proliferation and Promotes Apoptosis in Platelet Derived Growth Factor BB/Tumor Necrosis Factor α Induced Vascular Smooth Muscle Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Atherosclerosis is one of the primary causes that lead to cardiovascular disease. LncRNAs have been regarded as key modulators in many pathological processes. The study aims to identify the regulatory role of LncRNA fetal-lethal noncoding developmental regulatory RNA (FENDRR) in atherosclerosis.
Cell viability proliferation, cell cycle and cell apoptosis were evaluated by Cell Counting Kit-8 (CCK-8) assay flow cytometric analysis and western blot analysis. Quantitative real-time PCR (qRT-PCR) was carried out to determine FENDRR expression in PDGF-BB/TNF-α induced VSMCs.
Levels of TNF-α, IL-1, IL-6, MCP-1 and ICAM-1 were investigated by enzyme-linked immunosorbent assay (ELISA). The results showed that cell viability was enhanced and FENDRR expression was downregulated after VSMCs were induced by platelet derived growth factor BB (PDGF-BB) or
tumor necrosis factor a (TNF-α). Cell proliferation was inhibited by FENDRR overexpression in a time-dependent manner in PDGF-BB or TNF-α induced VSMCs. Moreover, FENDRR overexpression blocked cell cycle, suppressed the generations of TNF-α, IL-1, IL-6,
MCP-1 and ICAM-1, and facilitated cell apoptosis in VSMCs induced by PDGF-BB or TNF-α. These findings indicate the functional role of LncRNA FENDRR in atherosclerosis that attenuates cell proliferation and accelerates cell apoptosis.
Collapse
Affiliation(s)
- Xiaofang Chen
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dongjin Wang
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lingmei Qian
- Department of Cardiology The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
17
|
Jiang X, Li J, Zhang X, Chen H. Acute coronary syndrome in a young woman with a giant coronary aneurysm and mitral valve prolapse: a case report and literature review. J Int Med Res 2021; 49:300060521999525. [PMID: 33752500 PMCID: PMC7995495 DOI: 10.1177/0300060521999525] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
Acute coronary syndrome in the young population is infrequently seen and has a different etiology from that in the elderly population. Giant coronary artery aneurysms are rare and usually asymptomatic, but they can cause acute clinical symptoms such as chest pain or chest tightness. We herein describe a young woman with a history of mitral valve prolapse who developed sudden-onset chest pain. She had mild elevations of her creatine kinase and cardiac troponin levels; however, no ST segment alteration was found on an electrocardiogram, and no abnormal regional wall movement was noted on echocardiography. Cardiac magnetic resonance imaging with late gadolinium enhancement revealed a "mass" at the right coronary artery and linear subendocardial enhancement at the posterior wall. Coronary angiography later confirmed a giant coronary aneurysm with a substantial thrombus. The combined presence of the coronary artery aneurysm and mitral valve prolapse in this patient was likely a sequela of Kawasaki disease.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Zhejiang, China
- Department of Medical Examinations, First People’s Hospital of Wenling, Zhejiang, China
| | - Jiamin Li
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Zhejiang, China
| | - Xuehua Zhang
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Zhejiang, China
| | - Han Chen
- Department of Cardiology, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Lab of Cardiovascular Disease Diagnosis and Treatment, Zhejiang, China
| |
Collapse
|
18
|
Tang D, Geng F, Yu C, Zhang R. Recent Application of Zebrafish Models in Atherosclerosis Research. Front Cell Dev Biol 2021; 9:643697. [PMID: 33718384 PMCID: PMC7947229 DOI: 10.3389/fcell.2021.643697] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
Atherosclerotic cardiovascular disease is one of the leading causes of death worldwide. Establishing animal models of atherosclerosis is of great benefit for studying its complicated pathogenesis and screening and evaluating related drugs. Although researchers have generated a variety of models for atherosclerosis study in rabbits, mice and rats, the limitations of these models make it difficult to monitor the development of atherosclerosis, and these models are unsuitable for large scale screening of potential therapeutic targets. On the contrast, zebrafish can fulfill these purposes thanks to their fecundity, rapid development ex utero, embryonic transparency, and conserved lipid metabolism process. Thus, zebrafish have become a popular alternative animal model for atherosclerosis research. In this mini review, we summarize different zebrafish models used to study atherosclerosis, focusing on the latest applications of these models to the dynamic monitoring of atherosclerosis progression, mechanistic study of therapeutic intervention and drug screening, and assessment of the impacts of other risk factors.
Collapse
Affiliation(s)
- Dandan Tang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Fang Geng
- School of Life Sciences, Fudan University, Shanghai, China
| | - Chunxiao Yu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Gegunde S, Alfonso A, Alvariño R, Alonso E, González-Juanatey C, Botana LM. Crosstalk between cyclophilins and T lymphocytes in coronary artery disease. Exp Cell Res 2021; 400:112514. [PMID: 33582093 DOI: 10.1016/j.yexcr.2021.112514] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/13/2022]
Abstract
Cardiovascular diseases and atherosclerosis are currently some of the most widespread diseases of our time. Within cardiovascular disease, coronary artery disease and underlying atherosclerosis were recently linked with systemic and local inflammation. Cyclophilins participate in the initiation and progression of these inflammatory-related diseases. Cyclophilins are released into the extracellular space upon inflammatory stimuli and participate in the pathology of cardiovascular diseases. The cell surface receptor for extracellular cyclophilins, the CD147 receptor, also contributes to coronary artery disease pathogenesis. Nevertheless, the physiological relevance of cyclophilin's family and their receptor in cardiovascular diseases remains unclear. The present study aimed to better understand the role of cyclophilins in cardiovascular artery disease and their relationship with inflammation. Hence, cyclophilins and pro-inflammatory interleukins were measured in the serum of 30 subjects (divided into three groups according to coronary artery disease status: 10 patients with acute coronary syndrome, 10 patients with chronic coronary artery disease, and 10 control volunteers). In addition, cyclophilin levels and CD147 receptor expression were measured in T lymphocytes purified from these subjects. Cyclophilin A, B, and C, pro-inflammatory interleukins, and CD147 membrane expression were significantly elevated in patients with coronary artery disease.
Collapse
Affiliation(s)
- Sandra Gegunde
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, 27002, Spain; Grupo Investigación Biosdiscovery, IDIS, Spain
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, 27002, Spain; Grupo Investigación Biosdiscovery, IDIS, Spain.
| | - Rebeca Alvariño
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, 27002, Spain; Grupo Investigación Biosdiscovery, IDIS, Spain
| | - Eva Alonso
- Grupo Investigación Biosdiscovery, IDIS, Spain
| | - Carlos González-Juanatey
- Grupo Investigación Biosdiscovery, IDIS, Spain; Servicio de Cardiología, Hospital Universitario Lucus Augusti, Lugo, 27004, Spain
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, Universidad de Santiago de Compostela, Lugo, 27002, Spain; Grupo Investigación Biosdiscovery, IDIS, Spain
| |
Collapse
|
20
|
Li W, Su SA, Chen J, Ma H, Xiang M. Emerging roles of fibroblasts in cardiovascular calcification. J Cell Mol Med 2020; 25:1808-1816. [PMID: 33369201 PMCID: PMC7882970 DOI: 10.1111/jcmm.16150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/16/2020] [Accepted: 11/22/2020] [Indexed: 12/18/2022] Open
Abstract
Cardiovascular calcification, a kind of ectopic mineralization in cardiovascular system, including atherosclerotic calcification, arterial medial calcification, valve calcification and the gradually recognized heart muscle calcification, is a complex pathophysiological process correlated with poor prognosis. Although several cell types such as smooth muscle cells have been proven critical in vascular calcification, the aetiology of cardiovascular calcification remains to be clarified due to the diversity of cellular origin. Fibroblasts, which possess remarkable phenotypic plasticity that allows rapid adaption to fluctuating environment cues, have been demonstrated to play important roles in calcification of vasculature, valve and heart though our knowledge of the mechanisms controlling fibroblast phenotypic switching in the calcified process is far from complete. Indeed, the lack of definitive fibroblast lineage‐tracing studies and typical expression markers of fibroblasts raise major concerns regarding the contributions of fibroblasts during all the stages of cardiovascular calcification. The goal of this review was to rigorously summarize the current knowledge regarding possible phenotypes exhibited by fibroblasts within calcified cardiovascular system and evaluate the potential therapeutic targets that may control the phenotypic transition of fibroblasts in cardiovascular calcification.
Collapse
Affiliation(s)
- Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng-An Su
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Zhang Y, Li L, Liu X, Ding L, Wu X, Wang J, He M, Hou H, Ruan G, Lai J, Chen C. Examination of the Effect of a 50-Hz Electromagnetic Field at 500 μT on Parameters Related With the Cardiovascular System in Rats. Front Public Health 2020; 8:87. [PMID: 32318532 PMCID: PMC7154052 DOI: 10.3389/fpubh.2020.00087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/03/2020] [Indexed: 11/21/2022] Open
Abstract
Background: Whether electromagnetic field (EMF) exposure affects the function of the cardiovascular system is under debate. The present study aimed to investigate the effects of 500 μT EMF exposure on the cardiovascular system in rats. Methods: Forty-eight-week-old male Sprague-Dawley rats were randomly divided into two groups: the sham group and the exposure group. During 24-week EMF exposure (20 h per day), the blood pressure and pulse rate were recorded every 4 weeks. Before sacrifice, electrocardiography, echocardiography, and cardiac catheterization analysis were conducted to evaluate the cardiac function. Meanwhile, hematoxylin-eosin (HE) staining, Western blot, and real-time polymerase chain reaction (PCR) were performed to identify morphological and molecular changes indicative of cardiac remodeling. Results: The heart rate, blood pressure, and pulse rate were not influenced by EMF exposure compared with the control group. In addition, HE staining showed no change in the morphology and arrangement of cardiomyocytes. Further, we found that the mRNA and protein levels of cardiac hypertrophy-related genes were not affected by EMF exposure. Finally, no significant difference was observed in cardiac function between the two groups by echocardiography and cardiac catheterization detection. Conclusion: The 24-week exposure to EMF at 500 μT did not have apparent effects on the cardiovascular system in rats, at least for the variables studied.
Collapse
Affiliation(s)
- Yemao Zhang
- School of Electrical Engineering and Automation, Hefei University of Technology, Hefei, China.,State Key Laboratory of Power Grid Environmental Protection, High Voltage Research Institute, China Electric Power Research Institute, Wuhan, China
| | - Lijuan Li
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingfa Liu
- State Key Laboratory of Power Grid Environmental Protection, High Voltage Research Institute, China Electric Power Research Institute, Wuhan, China
| | - Lijian Ding
- School of Electrical Engineering and Automation, Hefei University of Technology, Hefei, China
| | - Xiong Wu
- State Key Laboratory of Power Grid Environmental Protection, High Voltage Research Institute, China Electric Power Research Institute, Wuhan, China
| | - Jin Wang
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengying He
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiying Hou
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoran Ruan
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinsheng Lai
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology and Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Guo Y, Yang Q, Weng XG, Wang YJ, Hu XQ, Zheng XJ, Li YJ, Zhu XX. Shenlian Extract Against Myocardial Injury Induced by Ischemia Through the Regulation of NF-κB/IκB Signaling Axis. Front Pharmacol 2020; 11:134. [PMID: 32210797 PMCID: PMC7069067 DOI: 10.3389/fphar.2020.00134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 01/31/2020] [Indexed: 12/22/2022] Open
Abstract
Ischemic heart disease (IHD), caused predominantly by atherosclerosis, is a leading cause of global mortality. Our previous studies showed that Shenlian extract (SL) could prevent the formation of atherosclerosis and enhance the stability of atherosclerotic plaques. To further investigate the protective effects of SL on myocardial ischemic injury and its possible mechanisms, anesthetized dogs, ex vivo rat hearts, and H9c2 cardiomyocytes were used as models. The results showed that SL had a significant protective effect on the anesthetized dog ligating coronary artery model, reduced the degree of myocardial ischemia (Σ-ST), and reduced the scope of myocardial ischemia (N-ST). Meanwhile, SL alleviated ischemic reperfusion damage in ex vivo rat hearts with improved LVEDP and ± dp/dtmax values of the left ventricle. SL reduced the pathological changes of LDH, IL-1β, MDA, and NO contents, all of which are related to the expression of NF-κB. Further analysis by Bio-Plex array and signal pathway blocker revealed that the phosphorylation of IκB was a key factor for SL to inhibit myocardial ischemic injury, and the regulation of SL on IκB was primarily related to degradation of the IκB protein. These results provided dependable evidence that SL could protect against myocardial ischemic injury through the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuan Guo
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qing Yang
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Gang Weng
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ya-Jie Wang
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue-Qi Hu
- College of Pharmacy, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao-Jun Zheng
- Pharmacy Department, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yu-Jie Li
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Xin Zhu
- Pharmacokinetics Laboratory, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Ma L, Nelson ER. Oxysterols and nuclear receptors. Mol Cell Endocrinol 2019; 484:42-51. [PMID: 30660701 DOI: 10.1016/j.mce.2019.01.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/08/2019] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
Oxysterols are derivatives of cholesterol and an important regulator of cholesterol metabolism, in part due to their role as ligands for nuclear receptors, such as the liver X receptors. Oxysterols are also known to be ligands for the RAR-related orphan receptors, involved in normal T cell differentiation. However, increasing evidence supports a role for oxysterols in the progression of several diseases. Here, we review recent developments in oxysterol research, highlighting the biological functions that oxysterols exert through their target nuclear receptors: the liver X receptors, estrogen receptors, RAR-related orphan receptors and the glucocorticoid receptor. We also bring the regulation of the immune system into the context of interaction between oxysterols and nuclear receptors, discussing the effect of such interaction on the pro-inflammatory function of macrophages and the development of T cells. Finally, we examine the impact that oxysterols have on various disease models, including cancer, Alzheimer's disease and atherosclerosis, stressing the role of nuclear receptors if previously identified. This review underscores the need to consider the multifaceted roles of oxysterols in terms of multiple receptor engagements and selective modulation of these receptors.
Collapse
Affiliation(s)
- Liqian Ma
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, Chicago, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, IL, United States.
| |
Collapse
|
24
|
Zhang J, Liang R, Wang L, Yang B. Effects and mechanisms of Danshen-Shanzha herb-pair for atherosclerosis treatment using network pharmacology and experimental pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2019; 229:104-114. [PMID: 30312741 DOI: 10.1016/j.jep.2018.10.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/29/2018] [Accepted: 10/01/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The danshen (the root of Salvia miltiorrhiza Bge.)-shanzha (the fruit of Crataegus pinnatifida Bge. var. major N.E.Br.) (DS) herb combination is a commonly used traditional Chinese medicine with cardiovascular disease (CVD) treatment potential. MATERIALS AND METHODS In this study, we investigated the anti-atherosclerotic effects and mechanisms of DS by the integration of network pharmacology and polypharmacology. Eight main components were selected for target fishing by PharmMapper. RESULTS The network pharmacological study indicated that DS may target 41 proteins and 16 pathways associated with inflammation, lipid metabolism and endothelial protection, which indicates that DS probably adjusts these processes as part of its anti-atherosclerotic activities. Furthermore, this hypothesis was verified by polypharmacology using an atherosclerotic model. Histopathological examination showed that DS inhibited pathological changes in the arteries of atherosclerotic rats and reduced the intima-media thickness (IMT). DS significantly reduced the levels of total cholesterol (TC), triglyceride (TG), and low-density lipoprotein-cholesterol (LDL-C) and increased the high-density lipoprotein-cholesterol (HDL-C) level in the blood. DS also decreased the concentrations of interleukin (IL)-1β and IL-18, indicating anti-inflammation activity. In addition, DS increased the serum levels of nitric oxide (NO) and 6-keto-prostaglandin F1α (6-keto-PGF1α) and decreased the serum levels of endothelin (ET) and thromboxane B2 (TXB2), indicating an endothelial protective effect. CONCLUSIONS In conclusion, DS has an anti-atherosclerotic ability to lower lipid concentrations and to protect endothelial function, and it also has anti-inflammatory activity.
Collapse
Affiliation(s)
- Jianyong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Rixin Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lan Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bin Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|