1
|
Wu W, Gong M, Liu P, Yu H, Gao X, Zhao X. Hypomagnesemia: exploring its multifaceted health impacts and associations with blood pressure regulation and metabolic syndrome. Diabetol Metab Syndr 2025; 17:217. [PMID: 40524199 PMCID: PMC12168336 DOI: 10.1186/s13098-025-01772-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 05/30/2025] [Indexed: 06/19/2025] Open
Abstract
This article provides a comprehensive and in-depth exploration of the multifaceted effects of Hypomagnesemia on human health, with a specific focus on its intricate associations with mechanisms regulating blood pressure and metabolic syndrome. Firstly, the fundamental concept of hypomagnesemia is elucidated, followed by a detailed analysis of its prevalence, risk factors, and Magnesium Deficiency Score. Furthermore, this article delves into the intricate relationship between hypomagnesemia and blood pressure regulation, encompassing its impact on endothelial function, vascular calcification, oxidative stress and inflammatory response, sympathetic nervous system activity as well as the renin-angiotensin-aldosterone system (RAAS). Additionally, it explores the correlation between hypomagnesemia and insulin resistance, metabolic syndrome along with other health issues. Notably noteworthy is that this paper also places special emphasis on exploring the potential role of hypomagnesemia in specific diseases such as renal hypertension and preeclampsia while providing novel insights for their prevention and treatment. Finally, this article summarizes the diverse effects of hypomagnesemia on health while anticipating future research directions. Future studies should further investigate the pathogenesis underlying hypomagnesemia while optimizing assessment methods for magnesium deficiency to develop targeted intervention strategies aimed at offering improved treatment options alongside preventive measures for patients.
Collapse
Affiliation(s)
- Wenlong Wu
- Department of Cardiology, the Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Shaku District, Dalian, 116023, China
- Department of Cardiology, Liao Ning University of Traditional Chinese Medicine, No.79, Chong Shan East Road, Huanggu District, Shenyang, 110085, China
| | - Ming Gong
- Department of Cardiology, the Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Shaku District, Dalian, 116023, China
| | - Pan Liu
- Laboratory of Basic Medicine, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shen He District, Shenyang, 110016, China
| | - Huiying Yu
- Laboratory of Basic Medicine, General Hospital of Northern Theater Command, No.83, Wenhua Road, Shen He District, Shenyang, 110016, China
| | - Xue Gao
- Department of Cardiology, the Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Shaku District, Dalian, 116023, China
- Department of Cardiology, Liao Ning University of Traditional Chinese Medicine, No.79, Chong Shan East Road, Huanggu District, Shenyang, 110085, China
| | - Xin Zhao
- Department of Cardiology, the Second Hospital of Dalian Medical University, No.467, Zhongshan Road, Shaku District, Dalian, 116023, China.
| |
Collapse
|
2
|
Liao W, Huang Y, Wang X, Hu Z, Zhao C, Wang G. Multidimensional excavation of the current status and trends of mechanobiology in cardiovascular homeostasis and remodeling within 20 years. MECHANOBIOLOGY IN MEDICINE 2025; 3:100127. [PMID: 40395770 PMCID: PMC12067904 DOI: 10.1016/j.mbm.2025.100127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/12/2025] [Accepted: 03/09/2025] [Indexed: 05/22/2025]
Abstract
Mechanobiology is essential for cardiovascular structure and function and regulates the normal physiological and pathological processes of the cardiovascular system. Cells in the cardiovascular system are extremely sensitive to their mechanical environment, and once mechanical stimulation is abnormal, the homeostasis mechanism is damaged or lost, leading to the occurrence of pathological remodeling diseases. In the past 20 years, many articles concerning the mechanobiology of cardiovascular homeostasis and remodeling have been published. To better understand the current development status, research hotspots and future development trends in the field, this paper uses CiteSpace software for bibliometric analysis, quantifies and visualizes the articles published in this field in the past 20 years, and reviews the research hotspots and emerging trends. The regulatory effects of mechanical stimulation on the biological behavior of endothelial cells, smooth muscle cells and the extracellular matrix, as well as the mechanical-related remodeling mechanism in heart failure, have always been research hotspots in this field. This paper reviews the research advances of these research hotspots in detail. This paper also introduces the research status of emerging hotspots, such as those related to cardiac fibrosis, homeostasis, mechanosensitive transcription factors and mechanosensitive ion channels. We hope to provide a systematic framework and new ideas for follow-up research on mechanobiology in the field of cardiovascular homeostasis and remodeling and promote the discovery of more therapeutic targets and novel markers of mechanobiology in the cardiovascular system.
Collapse
Affiliation(s)
- Wei Liao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Yuxi Huang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | | | - Ziqiu Hu
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
| | - Chuanrong Zhao
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Guixue Wang
- Key Laboratory of Biorheological and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
3
|
Tang Y, Cai Y, Peng F, Li M, Mo Z. Midkine Promote Atherosclerosis by Regulating the Expression of ATP-Binding Cassette Transporter A1 via Activator Protein-1. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07717-x. [PMID: 40402391 DOI: 10.1007/s10557-025-07717-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/23/2025]
Abstract
PURPOSE Midkine (MK) has been shown to facilitate atherosclerotic plaque formation by downregulating the expression of ATP-binding cassette transporter A1 (ABCA1). However, the mechanism by which MK regulates ABCA1 to promote atherosclerosis remains incompletely understood. In this study, we sought to investigate the molecular mechanism by which MK's regulation of ABCA1 influences the pathogenesis of atherosclerosis. METHODS Male apoE-/- mice were subjected to a high-fat diet to establish an atherosclerosis model. The model mice received intraperitoneal injections of MK and activator protein-1 (AP-l) inhibitor SR11302. The ATP-binding cassette transporter A1 (ABCA1) and AP1 expression were detected using immunohistochemistry (IHC), quantitative polymerase chain reaction (qPCR), and western blotting (WB). RAW264.7 macrophages were incubated with oxidized low-density lipoprotein (ox-LDL) to generate foam cells. These foam cells were treated with MK, SR11302, JNK inhibitor SP600125, and PI3K inhibitor wortmannin. The expression of ABCA1, AP-1, JNK, and PI3K were detected using qPCR and WB. The cholesterol efflux and lipid accumulation of cells were analyzed using scintillation counting and oil red O staining, respectively. RESULTS MK-treated mice exhibited an accelerated development of atherosclerotic lesion (30% in the MK group vs. 20% in the control group), along with hepatic steatosis and lipid disorder. The expression of c-fos and AP-1 were up-regulated by MK in macrophages. Compared with the MK-treated group, inhibition of AP-1 using SR11302 or transfection with c-fos siRNA markedly enhanced the cholesterol efflux (12.73% in the MK + SR11302 group vs. 9.98% in the MK group, 12.73% in the MK + si-c-fos group vs. 10.02 % in the MK group), reduced lipid accumulation, and increased the protein levels of ABCA1 in macrophages. Compared to the MK-treated group, mice treated with both MK and SR11302 showed downregulated ABCA1 expression in aortic sinus lesions, a larger lesion area (22.59% vs. 18.54%), and significantly elevated levels of plasma total cholesterol (TC), low-density lipoprotein (LDL), and triglycerides (TG). These results suggest that MK-induced pharmacological inhibition of AP-1 augmented ABCA1 expression in plaques, ameliorated lipid disorders, and abrogated atherosclerosis progression in apoE-/- mice. In addition, in vitro experiments revealed that the MK-induced up-regulation of c-fos expression was effectively suppressed by inhibitors of JNK and PI3K. CONCLUSIONS Our findings unveil a novel mechanistic pathway in atherosclerosis, whereby MK promotes the development of atherosclerosis by up-regulating AP-1 in macrophages via the PI3K/AKT/JNK signaling cascade.
Collapse
Affiliation(s)
- Yixin Tang
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, China
- Department of Cardiovascular Medicine, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yaqin Cai
- Clinical Laboratory of Zhuhai 5th People's Hospital, Zhuhai, 519055, China
| | - Fengling Peng
- Department of Cardiovascular Medicine, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ming Li
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, China.
- Department of Histology and Embryology, Hunan University of Medicine, Huaihua, 418000, China.
| | - Zhongcheng Mo
- Guangxi Key Laboratory of Diabetic Systems Medicine, Department of Histology and Embryology, Guilin Medical University, Guilin, 541199, China.
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin, 541001, China.
| |
Collapse
|
4
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Ballal S, Pathak PK, Bareja L, Aminov Z, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Exosomal non-coding RNAs: key regulators of inflammation-related cardiovascular disorders. Eur J Med Res 2025; 30:395. [PMID: 40390035 PMCID: PMC12087048 DOI: 10.1186/s40001-025-02649-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 04/30/2025] [Indexed: 05/21/2025] Open
Abstract
Inflammation is a complex, tightly regulated process involving biochemical and cellular reactions to harmful stimuli. Often termed "the internal fire", it is crucial for protecting the body and facilitating tissue healing. While inflammation is essential for survival, chronic inflammation can be detrimental, leading to tissue damage and reduced survival. The innate immune system triggers inflammation, closely linked to the development of heart diseases, with significant consequences for individuals. Inflammation in arterial walls or the body substantially contributes to atherosclerotic disease progression, affecting the cardiovascular system. Altered lipoproteins increase the risk of excessive blood clotting, a hallmark of atherosclerotic cardiovascular disease and its complications. Integrating inflammatory biomarkers with established risk assessment techniques can enhance our ability to identify at-risk individuals, assess their risk severity, and recommend appropriate CVD prevention strategies. Exosomes, a type of extracellular vesicle, are released by various cells and mediate cell communication locally and systemically. In the past decade, exosomes have been increasingly studied for their vital roles in health maintenance and disease processes. They can transport substances like non-coding RNAs, lipids, and proteins between cells, influencing immune responses and inflammation to elicit harmful or healing effects. This study focuses on the critical role of inflammation in heart disease progression and how non-coding RNAs in exosomes modulate the inflammatory process, either exacerbating or alleviating inflammation-related damage in the cardiovascular system.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Piyus Kumar Pathak
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
5
|
Nieddu G, Formato M, Lepedda AJ. Deciphering Acute Coronary Syndromes Pathobiology Through Proteomics. J Cardiovasc Dev Dis 2025; 12:188. [PMID: 40422959 DOI: 10.3390/jcdd12050188] [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: 01/29/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/28/2025] Open
Abstract
Acute coronary syndrome (ACS) refers to a spectrum of conditions characterized by a sudden decrease in blood flow to the heart. This includes unstable angina, the mildest form, as well as non-ST- and ST-segment elevation myocardial infarction. The primary cause of ACS is typically the rupture or erosion of an atherosclerotic plaque in a coronary artery, resulting in the formation of a blood clot that can, partially or completely, block the blood flow to the heart muscle. The ongoing discovery and comprehension of emerging biomarkers for atherosclerosis could enhance our capacity to predict future events, particularly when integrated alongside traditional risk factors in assessing overall risk profiles. With advancements in proteomic technologies, large-scale approaches have been increasingly instrumental in unraveling pathways implicated in atherosclerotic degeneration and identifying novel circulating markers, which may serve as early diagnostic indicators or targets for innovative therapies. Over recent decades, numerous matrices including plasma, urine, microparticles, lipoproteins, atherosclerotic plaque extracts and secretomes, as well as thrombi, have been examined to address these questions. Furthermore, proteomics has been applied to various experimental models of atherosclerosis to deepen our understanding of the mechanisms underlying atherogenesis. This review offers a critical overview of the past two decades of untargeted omics research focused on identifying circulating and tissue biomarkers relevant to ACS.
Collapse
Affiliation(s)
- Gabriele Nieddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Marilena Formato
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | | |
Collapse
|
6
|
Eid AH. A Game Changer for Resistant Hypertension: The Rise of Aprocitentan. J Cardiovasc Pharmacol 2025; 85:312-315. [PMID: 39903701 DOI: 10.1097/fjc.0000000000001679] [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: 10/18/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
ABSTRACT Hypertension is a major risk factor for cardiovascular disease, and a major contributor to global morbidity and mortality. In particular, resistant hypertension (rHTN), defined as blood pressure that remains elevated despite treatment with at least three antihypertensive agents including a diuretic, continues to be a major pharmacotherapeutic challenge. Traditional antihypertensive drugs often fail in patients with rHTN, underscoring the need for novel therapies. This is a brief mini-review of aprocitentan, a new drug that promises a glimmer of hope for patients with rHTN. This drug is a dual endothelin (ET) receptor antagonist that blocks both ET A and ET B receptors. Given that these two receptors are critical players in vasotone regulation, antagonizing them, such as by aprocitentan, would be expected to significantly reduce blood pressure in patients with rHTN. Indeed, the PRECISION clinical trial demonstrated aprocitentan's superior effectiveness in reducing blood pressure in resistant patients, and the effects were sustained. Aprocitentan has been recently FDA-approved, marking a major milestone in hypertension management, offering hope for patients with difficult-to-treat hypertension.
Collapse
Affiliation(s)
- Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Zhang M, Lei R, Wang L, Jiang Y, Zhou X, Wang Y. KLF4 regulates FAM3A to promotes angiotensin II-induced proliferation and migration of vascular smooth muscle cells through the PI3K/AKT signaling pathway. Peptides 2025; 187:171379. [PMID: 40015605 DOI: 10.1016/j.peptides.2025.171379] [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: 12/03/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Hypertension, a major cause of cardiovascular disease, is linked to vascular remodeling, which is influenced by phenotypic changes in vascular smooth muscle cells (VSMCs). Studies have shown that KLF4 influences vascular remodeling by promoting VSMC dedifferentiation, increasing proliferation, and enhancing inflammatory responses, while FAM3 may play a key role in VSMC migration and proliferation. Angiotensin II (Ang II) contributes to remodeling, but the mechanisms are unclear. METHODS Ang II was used to stimulate VSMCs in order to evaluate the expression levels of KLF4 and FAM3A. EdU assays, transwell and scratch wound healing assays measured proliferation and migration. KLF4 knockdown and overexpression experiments were performed to examine the effects on FAM3A expression and VSMC behavior. Western blotting was conducted to analyze protein expression levels of KLF4, FAM3A, and PI3K/AKT signaling components. Bioinformatics analysis was used to predict KLF4 binding sites on the FAM3A promoter. Luciferase and CHIP assays confirmed regulation. RESULTS Ang II stimulation increased VSMC proliferation, migration, and the expression of KLF4 and FAM3A. Knockdown of KLF4 reduced Ang II-induced proliferation and migration of VSMCs, accompanied by decreased FAM3A expression. Conversely, overexpression of KLF4 enhanced FAM3A levels, promoting VSMC proliferation and migration. Bioinformatics, luciferase reporter assays and CHIP assay confirmed that KLF4 directly binds to the FAM3A promoter. FAM3A knockdown inhibited Ang II-induced VSMC proliferation and migration by reducing PI3K/AKT pathway activation, whereas FAM3A overexpression reversed the inhibitory effects of KLF4 knockdown. CONCLUSION KLF4 transcriptionally regulates FAM3A, modulating Ang II-induced VSMC proliferation and migration through the PI3K/AKT signaling pathway.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Kruppel-Like Factor 4
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Signal Transduction/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphatidylinositol 3-Kinases/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Animals
- Humans
- Cells, Cultured
Collapse
Affiliation(s)
- Min Zhang
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Rong Lei
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Liqiong Wang
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Yimin Jiang
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Xiaoyan Zhou
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China
| | - Yuquan Wang
- Physical Examination Center, Department of Cardiology, Affiliated Hospital of North Sichuan Medical College, Nanchong City, Sichuan Province, China.
| |
Collapse
|
8
|
Heller K, Doukas P, Uhl C, Gombert A. Sex-Specific Characteristics of Perivascular Fat in Aortic Aneurysms. J Clin Med 2025; 14:3071. [PMID: 40364103 PMCID: PMC12072766 DOI: 10.3390/jcm14093071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/17/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Aortic aneurysms (AAs), the dilation or widening of the aorta, lead to dissection or rupture with high morbidity and mortality if untreated. AA displays gender disparities in its prevalence, progression and outcomes, with women having worse outcomes and faster aneurysm growth. However, current guidelines do not address gender dimorphism, emphasizing the urgent need for personalized treatment strategies and further research. Perivascular adipose tissue (PVAT), a unique type of fat surrounding blood vessels, plays a critical role in maintaining vasomotor tone and vascular homeostasis, with dysfunction associated with chronic inflammation and vessel-wall remodeling. Indeed, PVAT dysfunction promotes the development of aortic aneurysms, with hormonal and biomechanical factors exacerbating the pathological vascular microenvironment. The sexually dimorphic characteristics of PVAT include morphological, immunological, and hormonally mediated differences. Thus, targeting PVAT-mediated mechanisms may be a promising option for the (gender-specific) therapeutic management of cardiovascular pathologies. This review examines the emerging importance of PVAT in vascular health, its potential therapeutic implications for AA, and identifies gaps in the current state of research.
Collapse
Affiliation(s)
- Katja Heller
- Department of Vascular Surgery, RWTH Aachen University Hospital, 52074 Aachen, Germany; (P.D.); (C.U.); (A.G.)
| | | | | | | |
Collapse
|
9
|
Kajuluri LP, Guo YY, Lee S, Christof M, Malhotra R. Epigenetic Regulation of Human Vascular Calcification. Genes (Basel) 2025; 16:506. [PMID: 40428328 PMCID: PMC12111397 DOI: 10.3390/genes16050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Vascular diseases present a significant threat to human health worldwide. Atherosclerosis is the most prevalent vascular disease, accounting for the majority of morbidity and mortality globally. Vascular calcification is a dynamic pathological process underlying the development of atherosclerotic plaques and involves the phenotypic transformation of vascular smooth muscle cells (VSMCs) into osteogenic cells. Specifically, the phenotypic switch in VSMCs often involves modifications in gene expression due to epigenetic changes, including DNA methylation, histone modification, and non-coding RNAs. Understanding the role of these epigenetic changes in regulating the pathophysiology of vascular calcification, along with the proteins and pathways that mediate these changes, will aid in identifying new therapeutic candidates to enhance vascular health. This review discusses a comprehensive range of epigenetic modifications and their implications for vascular health and the development of vascular calcification.
Collapse
Affiliation(s)
- Lova Prasadareddy Kajuluri
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Yugene Young Guo
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Sujin Lee
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| | - Michael Christof
- School of Arts and Sciences, University of Rochester, Rochester, NY 14627, USA;
| | - Rajeev Malhotra
- Cardiovascular Research Center, Heart and Vascular Institute, Mass General Brigham, Boston, MA 02114, USA; (L.P.K.); (Y.Y.G.); (S.L.)
| |
Collapse
|
10
|
Zhai Y, Shang H, Li Y, Zhang N, Zhang J, Wu S. A Systematic Review of risk factors for major adverse cardiovascular events in patients with coronary heart disease who underwent percutaneous coronary intervention. Front Physiol 2025; 16:1514585. [PMID: 40271216 PMCID: PMC12014560 DOI: 10.3389/fphys.2025.1514585] [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: 10/21/2024] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
Objective This study aims to systematically review the risk factors for major adverse cardiovascular events (MACE) in patients with coronary heart disease who have undergone percutaneous coronary intervention (PCI). Design Systematic review and meta-analysis. Data sources The Cochrane Library, PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), Wanfang Database, and VIP Database for Chinese Technical Periodicals (VIP) were screened until December 2024. Eligibility criteria for selecting studies Case-control studies or cohort studies on the risk factors for MACE in patients with coronary heart disease who underwent PCI. Data extraction and synthesis: The literature review, data extraction, and quality evaluation were conducted by two independent researchers, and the meta-analysis was performed using RevMan 5.4 software. Main outcomes The main outcome was that MACE occurred during the follow-up period. Results A total of 40 articles were included. The meta-analysis erevealed that dyslipidemia (OR = 1.50; 95% CI [1.19, 1.89], p = 0.0007), diabetes mellitus (OR = 1.70; 95% CI [1.43, 2.02], p < 0.00001), hypertension (OR = 1.62; 95% CI [1.35, 1.96], p < 0.0001), history of smoking (OR = 2.08; 95% CI [1.51, 2.85], p < 0.0001), poorer ventricular function (OR = 2.39; 95% CI [2.17-2.64], p < 0.0001), impaired left ventricular ejection fraction (LVEF) (OR = 1.86; 95% CI [1.71-2.03], p < 0.0001), door to balloon (D-to-B) time (OR = 0.61; 95% CI [0.42-0.88]; p = 0.009), thrombolysis in myocardial infarction (TIMI) (OR = 1.41; 95% CI [1.17, 1.70], p = 0.0004), renal dysfunction (OR = 1.82; 95% CI [1.37, 2.43], p < 0.0001), and multi-vessel coronary artery disease (OR = 0.41; 95% CI [0.37, 0.46], p < 0.0001) were significantly associated with MACE after PCI. Conclusion The independent risk factors of MACE after PCI are dyslipidemia, hypertension, diabetes mellitus, smoking history, Killip class > II, LVEF ≤40%, D-to-B time >90 min, TIMI flow grade ≤ II, renal insufficiency, and multivessel disease.
Collapse
Affiliation(s)
- You Zhai
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Nan Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jisi Zhang
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Shangwen Wu
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Zhang B, Liu Z, Chang Y, Lv R, Guo H, Qiang P, Shimosawa T, Xu Q, Yang F. Aldosterone-Induced Transformation of Vascular Smooth Muscle Cells into Macrophage-like Cells Participates in Inflammatory Vascular Lesions. Int J Mol Sci 2025; 26:3345. [PMID: 40244230 PMCID: PMC11989480 DOI: 10.3390/ijms26073345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the most abundant cell type in blood vessels, participating in cardiovascular diseases in various ways, among which their transformation into macrophage-like cells has become a research hotspot. In this study, rats were infused with aldosterone for 12 weeks, and VSMCs stimulated with aldosterone in vitro were used to observe aortic injury and the role of VSMC transformation. Vascular changes were detected via small animal ultrasound and H&E staining. Moreover, immunohistochemistry, immunofluorescence, Western blot, and flow cytometry were used to verify that the transformation of VSMCs into macrophage-like cells is regulated by mineralocorticoid receptor (MR) activation and macrophage colony-stimulating factor (M-CSF) and its receptor. Rat vasculature and in vitro cellular experiments revealed that VSMCs transformed into macrophage-like cells and secreted inflammatory factors such as interleukin-1β (IL-1β) and monocyte chemoattractant protein-1 (MCP-1), thereby exacerbating inflammatory vascular lesions, which was inhibited by the MR antagonist esaxerenone. These results reveal that increased levels of aldosterone activate MR, leading to the secretion of M-CSF by VSMCs. This further promotes the transformation of VSMCs into macrophage-like cells, which participate in inflammatory vascular lesions. Therefore, inhibiting the formation of macrophage-like cells can effectively reduce inflammatory vascular lesions.
Collapse
MESH Headings
- Animals
- Aldosterone/pharmacology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/cytology
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/pathology
- Rats
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Receptors, Mineralocorticoid/metabolism
- Male
- Inflammation/metabolism
- Inflammation/pathology
- Rats, Sprague-Dawley
- Cells, Cultured
- Macrophage Colony-Stimulating Factor/metabolism
- Interleukin-1beta/metabolism
- Chemokine CCL2/metabolism
Collapse
Affiliation(s)
- Boya Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ziqian Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yi Chang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Ruyan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Haixia Guo
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Panpan Qiang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, International University of Health and Welfare, Narita 286-8686, Japan
| | - Qingyou Xu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Fan Yang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Institute of Integrative Medicine, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| |
Collapse
|
12
|
Shi Y, Liu L, Gong Y, Zhang C, Yang Y, Wang W, Qin L. Isovaleroylbinankadsurin A ameliorates atherosclerosis and restenosis by promoting LXRα signaling pathway and inhibiting TGF-β1 and FHL1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156451. [PMID: 39914064 DOI: 10.1016/j.phymed.2025.156451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/03/2025] [Accepted: 01/29/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Atherosclerosis is a leading factor in the development of several cardiovascular conditions, including ischemic heart disease, stroke, and peripheral vascular disease. A defining characteristic of atherosclerosis is the formation of macrophages and vascular smooth muscle cells (VSMCs)-derived foam cells in plaques. Angioplasty can effectively remove atherosclerotic plaque, while it may lead to restenosis. A crucial pathological feature of restenosis is neointimal formation, which is driven by the phenotypic change, growth, and migration of VSMCs. Nonetheless, there are only a handful of effective strategies. Kadsura coccinea is a folk Chinese herb mainly used to treat rheumatism, chronic gastritis, bruises, and dysmenorrhea. Isovaleroylbinankadsurin A (ISBA), isolated from Kadsura coccinea roots, is a dibenzocyclooctadiene lignan that has recently been shown to be beneficial for myocardial ischemia-reperfusion injury. However, its protective effects on atherosclerosis and restenosis remain unknown. PURPOSE To investigate the effects and related mechanisms of ISBA on atherosclerosis and restenosis. METHODS Foam cells were induced by ox-LDL in vitro, and a high-fat diet was administered to ApoE-/- mice. HE staining was applied to evaluate the morphology of vascular tissues. Lipid accumulation of plaques and foam cells was measured using BODIPY-cholesterol, DiI-ox-LDL, Oil Red O staining, and cholesterol quantification tests. A mouse model of femoral artery injury and an in vitro VSMC proliferation model were established. The CCK-8, EdU, plate clone formation, and wound-healing assays were used to evaluate cell viability and migration. Western blot analysis and immunohistochemistry were employed to assess the levels of crucial proteins in ISBA mediating atherosclerosis and restenosis. RESULTS We found for the first time that ISBA could significantly alleviate atherosclerosis and restenosis. Mechanistically, ISBA inhibited lipid accumulation and reduced foam cell formation through the activation of LXRα/ABCA1 signaling pathway, which contributed to preventing atherosclerosis. In addition, ISBA could also suppress the phenotypic switch, proliferation, and migration of VSMCs through repressing TGF-β1/ERK1/2/CTGF and FHL1/ERK1/2/CTGF signaling pathways, thereby mitigating neointimal formation and restenosis. CONCLUSION This study offers a groundbreaking and expanded exploration of the pharmacological effects of ISBA. ISBA may be a novel therapeutic drug to prevent atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Yaning Shi
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Leping Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Yongzhen Gong
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Chanjuan Zhang
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Yupei Yang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China
| | - Li Qin
- Laboratory of Stem Cell Regulation with Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Institutional Key Laboratory of Vascular Biology and Translational Medicine in Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China.
| |
Collapse
|
13
|
Guo L, Du Y, Li H, He T, Yao L, Yang G, Yang X. Metabolites-mediated posttranslational modifications in cardiac metabolic remodeling: Implications for disease pathology and therapeutic potential. Metabolism 2025; 165:156144. [PMID: 39864796 DOI: 10.1016/j.metabol.2025.156144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
The nonenergy - producing functions of metabolism are attracting increasing attention, as metabolic changes are involved in discrete pathways modulating enzyme activity and gene expression. Substantial evidence suggests that myocardial metabolic remodeling occurring during diabetic cardiomyopathy, heart failure, and cardiac pathological stress (e.g., myocardial ischemia, pressure overload) contributes to the progression of pathology. Within the rewired metabolic network, metabolic intermediates and end-products can directly alter protein function and/or regulate epigenetic modifications by providing acyl groups for posttranslational modifications, thereby affecting the overall cardiac stress response and providing a direct link between cellular metabolism and cardiac pathology. This review provides a comprehensive overview of the functional diversity and mechanistic roles of several types of metabolite-mediated histone and nonhistone acylation, namely O-GlcNAcylation, lactylation, crotonylation, β-hydroxybutyrylation, and succinylation, as well as fatty acid-mediated modifications, in regulating physiological processes and contributing to the progression of heart disease. Furthermore, it explores the potential of these modifications as therapeutic targets for disease intervention.
Collapse
Affiliation(s)
- Lifei Guo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; Cadet Team 6 of School of Basic Medicine, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Yuting Du
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China; The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Heng Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Ting He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China
| | - Li Yao
- Department of Pathology, Xi' an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi' an 710018, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| | - Xuekang Yang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, Chang-Le Xi Street #127, Xi' an 710032, China.
| |
Collapse
|
14
|
Wang SF, Yang LY, Zhao AQ, Wang ZY, Wang S, Gong M, Zheng MQ, Liu G, Yang SY, Lin JJ, Sun SG. A Novel Hidden Protein p-414aa Encoded by circSETD2(14,15) Inhibits Vascular Remodeling. Circulation 2025. [PMID: 40099364 DOI: 10.1161/circulationaha.124.070243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Phenotypic switching of vascular smooth muscle cells (VSMCs), leading to neointimal hyperplasia, is a fundamental cause of vascular remodeling diseases such as atherosclerosis and hypertension. Novel hidden proteins encoded by circular RNAs play crucial roles in disease progression, yet their involvement in vascular remodeling diseases has not been comprehensively studied. This study identifies a novel protein derived from a circular RNA in VSMCs and demonstrates its potential role in regulating vascular remodeling. METHODS Cell proliferation assays were performed to investigate the effects of circSETD2(14,15) on VSMC proliferation. Techniques such as vector construction, immunoprecipitation-mass spectrometry, and dual-luciferase reporter gene were used to confirm that circSETD2(14,15) encoded a novel protein, p-414aa. The interaction between p-414aa and HuR (human antigen R) was validated with techniques such as coimmunoprecipitation, mass spectrometry, and proximity ligation assay. Through experiments including RNA sequencing and RNA immunoprecipitation, the interaction between HuR and C-FOS (C-Fos proto-oncogene) mRNA was revealed. The role of p-414aa in neointimal hyperplasia was assessed with a carotid artery ligation model in male mice. RESULTS Overexpression of circSETD2(14,15) inhibits VSMC phenotypic switching. The novel protein p-414aa, encoded by circSETD2(14,15), interacts with HuR to reduce C-FOS mRNA stability, thereby suppressing VSMC proliferation and ultimately inhibiting neointimal hyperplasia in male mice. CONCLUSIONS We uncover a novel hidden protein derived from circSETD2(14,15), called p-414aa, that inhibits vascular remodeling. CircSETD2(14,15) and p-414aa may serve as potential therapeutic targets for vascular remodeling diseases.
Collapse
Affiliation(s)
- Si-Fan Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
- Department of Gynecology and Obstetrics, Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, China (S.-F.W.)
| | - Li-Yun Yang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| | - An-Qi Zhao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| | - Zhao-Yi Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| | - Sen Wang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| | - Miao Gong
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| | - Ming-Qi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, China (M.-Q.Z., G.L.)
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, China (M.-Q.Z., G.L.)
| | - Shu-Yan Yang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, China (S.-Y.Y.)
| | - Jia-Jie Lin
- School of Basic Medicine, Jiangxi Provincial Key Laboratory of Tissue Engineering, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China (J.-J.L.)
| | - Shao-Guang Sun
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China (S.-F.W., L.-Y.Y., A.-Q.Z., Z.-Y.W., S.W., M.G., S.-G.S.)
| |
Collapse
|
15
|
Jin Q, Lin B, Lu L. Potential therapeutic value of dietary polysaccharides in cardiovascular disease: Extraction, mechanisms, applications, and challenges. Int J Biol Macromol 2025; 296:139573. [PMID: 39793800 DOI: 10.1016/j.ijbiomac.2025.139573] [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: 08/29/2024] [Revised: 10/22/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025]
Abstract
Dietary polysaccharides, recognised as significant natural bioactive compounds, have demonstrated promising potential for the prevention and treatment of cardiovascular disease (CVD). This review provides an overview of the biological properties and classification of polysaccharides, with particular emphasis on their extraction and purification methods. The paper then explores the diverse mechanisms by which polysaccharides exert their effects in CVD, including their antioxidant activity, protection against ischemia-reperfusion injury, anti-apoptotic properties, protection against diabetic cardiomyopathy, anticoagulant and antithrombotic effects, prevention of ventricular remodeling, and protection against vascular injury. Furthermore, this paper summarises the current status of clinical trials involving polysaccharides in CVD and analyzes the support and challenges posed by these studies for the practical application of polysaccharides. Finally, the major challenges facing the therapeutic use of polysaccharides in CVD are discussed, particularly the issues of low bioavailability and lack of standardized quality control. Through this review, we aimed to provide a reference and guidance for further research on and application of dietary polysaccharides in CVD.
Collapse
Affiliation(s)
- Qiqi Jin
- Department of Cardiology, Wenzhou Central Hospital, Wenzhou 325000, China
| | - Bin Lin
- Department of Cardiology, Wenzhou Central Hospital, Wenzhou 325000, China.
| | - Lingfen Lu
- Department of Cardiology, Wenzhou Central Hospital, Wenzhou 325000, China.
| |
Collapse
|
16
|
Wang Z, Sun W, Zhang K, Ke X, Wang Z. New insights into the relationship of mitochondrial metabolism and atherosclerosis. Cell Signal 2025; 127:111580. [PMID: 39732307 DOI: 10.1016/j.cellsig.2024.111580] [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: 11/13/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are the number one killer of human health. In view of the important role of mitochondria in the formation and evolution of atherosclerosis, our manuscript aims to comprehensively elaborate the relationship between mitochondria and the formation and evolution of atherosclerosis from the aspects of mitochondrial dynamics, mitochondria-organelle interaction (communication), mitochondria and cell death, mitochondria and vascular smooth muscle cell phenotypic switch, etc., which is combined with genome, transcriptome and proteome, in order to provide new ideas for the pathogenesis of atherosclerosis and the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Zexun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China
| | - Wangqing Sun
- Department of Radiology, Yixing Tumor Hospital, Yixing 214200, China
| | - Kai Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Xianjin Ke
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
17
|
Soudant J, González-Blázquez R, Merino A, Ballesteros-Martínez C, Rodrigues-Diez R, Moreno-Carriles R, Nistal JF, Guerra S, Redondo JM, Salaices M, Briones AM, García-Redondo AB. Interferon stimulated gene 15 (ISG15) modulates phenotype of vascular smooth muscle cells and pathological vascular remodeling. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2025:500769. [PMID: 40023687 DOI: 10.1016/j.arteri.2025.500769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Inflammation is a major determinant of abdominal aortic aneurysms (AAA). Interferon stimulated gene 15 (ISG15) has a role in vascular remodelling in AAA. This study investigates the mechanisms whereby ISG15 might affect vascular remodeling and function. METHODS We used vascular smooth muscle cells (VSMC) from wild type (ISG15+/+) o ISG15 knockout (ISG15-/-) mice, aorta from ISG15+/+ and ISG15-/- mice infused with angiotensin II (1.44mg/kg/day, sc, 14 days), and human AAA. We also performed a model of recombinant ISG15 infusion (rISG15, sc, 100 and 500ng/day, 14 days) in mice. RESULTS In VSMC, ISG15 deficiency increased the expression of contractile (Acta2, Tagln) and synthetic (Fn1, Col1a2, Col3, Col4) markers and decreased the expression of the calcification marker Spp1. Ang II infusion changed the expression of phenotype markers differently in aorta from ISG15+/+ or ISG15-/- mice. ISG15 expression showed a negative correlation with expression of contractile markers (ACTA2, CNN1), and with COL3a1, in human samples from patients with AAA or with stenotic aorto-iliac pathology. rISG15 infusion induced hypotrophic vascular remodelling in mesenteric arteries without affecting vascular mechanics. Aorta of ISG15-/- mice contracted more to thromboxane A2 analogue U46619, compared to ISG15-/-mice. Both aorta and mesenteric arteries from rISG15-treated mice showed less contractility than control mice. CONCLUSIONS ISG15 participates in pathological vascular remodeling probably by modulating VSMC phenotype. These changes could also impact in the vascular function.
Collapse
Affiliation(s)
- Julius Soudant
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Raquel González-Blázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Abraham Merino
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Constanza Ballesteros-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Raquel Rodrigues-Diez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Rosa Moreno-Carriles
- Servicio de Angiología y Cirugía Vascular, Hospital Universitario La Princesa, Madrid, España
| | - J Francisco Nistal
- CIBER Cardiovascular, España; Cirugía Cardiovascular, Hospital Universitario Marqués de Valdecilla, IDIVAL, Departamento de Ciencias Médica y Quirúrgicas, Facultad de Medicina, Universidad de Cantabria, Santander, España
| | - Susana Guerra
- Departamento de Medicina Preventiva y Microbiología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España
| | - Juan Miguel Redondo
- CIBER Cardiovascular, España; Grupo de Regulación Génica en Remodelado Cardiovascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Madrid, España; Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas,Universidad Autónoma de Madrid, Madrid, España
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España; CIBER Cardiovascular, España.
| | - Ana B García-Redondo
- CIBER Cardiovascular, España; Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid. Instituto de Investigación Hospital Universitario La Paz (IdiPaz), Madrid, España.
| |
Collapse
|
18
|
Liu Y, Tan X, Wang L, Jin W, He Y, Yan Y, Hu K, Wang H, Xiang C, Hou M, Lai Y. Identification of the CeRNA axis of circ_0000006/miR-483-5p/KDM2B in the progression of aortic aneurysm to aorta dissection. BMC Cardiovasc Disord 2025; 25:141. [PMID: 40021969 PMCID: PMC11869725 DOI: 10.1186/s12872-025-04598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Aortic aneurysm (AA) and aortic dissection (AD) are serious cardiovascular disorders with a high risk of mortality. The molecular mechanisms underlying the progression from AA to AD are not well understood. This study aimed to identify the key circular RNA (circRNA)-microRNA (miRNA)-messenger RNA (mRNA) regulatory axis involved in this disease progression. METHODS CircRNA microarray, miRNA microarray, and mRNA sequencing were performed on plasma samples from healthy controls, AA patients, and AD patients. Bioinformatics analysis integrated the expression profiles to identify dysregulated circRNA-miRNA-mRNA networks. Key molecules were validated in vascular smooth muscle cells (VSMCs) and an AD mouse model. Cell proliferation, migration, and phenotypic transition assays were conducted after modulating the identified circRNA. The impact on AD progression was evaluated in mice upon circRNA knockdown. RESULTS A total of 12 circRNAs were found upregulated in AD compared to AA samples. miR-483-5p was downregulated while its targets KDM2B and circ_0000006 were upregulated in AD. Silencing circ_0000006 in VSMCs inhibited PDGF-induced phenotypic switching, proliferation, and migration by increasing miR-483-5p and decreasing KDM2B levels. In the AD mouse model, knockdown of circ_0000006 alleviated disease progression with similar molecular changes. CONCLUSION The study identified a novel circ_0000006/miR-483-5p/KDM2B axis dysregulated during AD progression. Targeting this axis, especially circ_0000006, could be a potential strategy to mitigate the transition from AA to AD by modulating VSMC phenotype and function.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Animals
- Humans
- Aortic Dissection/genetics
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/metabolism
- Disease Progression
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Cell Movement
- Disease Models, Animal
- Cell Proliferation
- Male
- Cells, Cultured
- Aortic Aneurysm/genetics
- Aortic Aneurysm/pathology
- Aortic Aneurysm/enzymology
- Aortic Aneurysm/metabolism
- Case-Control Studies
- Female
- Middle Aged
- Mice, Inbred C57BL
- Signal Transduction
- Gene Regulatory Networks
- Gene Expression Regulation
- Mice
- Phenotype
- Aged
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Yong Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China.
- , No. 63, Culture Road, Nanchong, Sichuan, 637000, China.
| | - Xiong Tan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Liang Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Weitao Jin
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yangchen He
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yu Yan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Kai Hu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Hao Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Chaohu Xiang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Ming Hou
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Yinglong Lai
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| |
Collapse
|
19
|
Li X, Zhao W, Wang Z, Moura AK, Roudbari K, Zuo R, Hu JZ, Wang YT, Li PL, Zhang Y. Acid Sphingomyelinase Regulates AdipoRon-Induced Differentiation of Arterial Smooth Muscle Cells via TFEB Activation. Int J Mol Sci 2025; 26:2147. [PMID: 40076784 PMCID: PMC11899876 DOI: 10.3390/ijms26052147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
AdipoRon is a selective adiponectin receptor agonist that inhibits vascular remodeling by promoting the differentiation of arterial smooth muscle cells (SMCs). Our recent studies have demonstrated that activation of TFEB and its downstream autophagy-lysosomal signaling contribute to adipoRon-induced differentiation of SMCs. The present study was designed to examine whether acid sphingomyelinase (ASM; gene symbol Smpd1) is involved in mediating adipoRon-induced activation of TFEB-autophagy signaling and inhibition of proliferation/migration in arterial SMCs. Our results showed that adipoRon induced ASM expression and ceramide production in Smpd1+/+ SMCs, which were abolished in Smpd1-/- SMCs. Compared to Smpd1+/+ SMCs, Smpd1-/- SMCs exhibited less TFEB nuclear translocation and activation of autophagy signaling induced by adipoRon stimulation. SMC differentiation was further characterized by retarded wound healing, reduced proliferation, F-actin reorganization, and MMP downregulation. The results showed that Smpd1-/- SMCs were less responsive to adipoRon-induced differentiation than Smpd1+/+ SMCs. Mechanistically, adipoRon increased the expression of protein phosphatases such as calcineurin and PP2A in Smpd1+/+ SMCs. The calcineurin inhibitor FK506/cyclosporin A or PP2A inhibitor okadaic acid significantly attenuated adipoRon-induced activation of TFEB-autophagy signaling. In addition, adipoRon-induced expressions of calcineurin and PP2A were not observed in Smpd1-/- SMCs. However, activation of calcineurin by lysosomal TRPML1-Ca2+ channel agonist ML-SA1 rescued the activation of TFEB-autophagy signaling and the effects of adipoRon on cell differentiation in Smpd1-/- SMCs. Taken together, these data suggested that ASM regulates adipoRon-induced SMC differentiation through TFEB activation. This study provided novel mechanistic insights into the therapeutic effects of adipoRon on TFEB signaling and pathological vascular remodeling.
Collapse
MESH Headings
- Sphingomyelin Phosphodiesterase/metabolism
- Sphingomyelin Phosphodiesterase/genetics
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Cell Differentiation/drug effects
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/cytology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Piperidines/pharmacology
- Signal Transduction/drug effects
- Autophagy/drug effects
- Mice
- Cells, Cultured
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Ceramides/metabolism
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Wei Zhao
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Zhengchao Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Alexandra K. Moura
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Kiana Roudbari
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Rui Zuo
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Jenny Z. Hu
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Yun-Ting Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA; (X.L.); (W.Z.); (A.K.M.); (K.R.); (R.Z.); (J.Z.H.); (Y.-T.W.)
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Yang Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
20
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Harnessing Gasotransmitters to Combat Age-Related Oxidative Stress in Smooth Muscle and Endothelial Cells. Pharmaceuticals (Basel) 2025; 18:344. [PMID: 40143122 PMCID: PMC11946800 DOI: 10.3390/ph18030344] [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: 01/25/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/28/2025] Open
Abstract
Age-related oxidative stress is a critical factor in vascular dysfunction, contributing to hypertension and atherosclerosis. Smooth muscle cells and endothelial cells are particularly susceptible to oxidative damage, which exacerbates vascular aging through cellular senescence, chronic inflammation, and arterial stiffness. Gasotransmitters-hydrogen sulfide (H2S), nitric oxide (NO), and carbon monoxide (CO)-are emerging as promising therapeutic agents for counteracting these processes. This review synthesizes findings from recent studies focusing on the mechanisms by which H2S, NO, and CO influence vascular smooth muscle and endothelial cell function. Therapeutic strategies involving exogenous gasotransmitter delivery systems and combination therapies were analyzed. H2S enhances mitochondrial bioenergetics, scavenges ROS, and activates antioxidant pathways. NO improves endothelial function, promotes vasodilation, and inhibits platelet aggregation. CO exhibits cytoprotective and anti-inflammatory effects by modulating heme oxygenase activity and ROS production. In preclinical studies, gasotransmitter-releasing molecules (e.g., NaHS, SNAP, CORMs) and targeted delivery systems show significant promise. Synergistic effects with lifestyle modifications and antioxidant therapies further enhance their therapeutic potential. In conclusion, gasotransmitters hold significant promise as therapeutic agents to combat age-related oxidative stress in vascular cells. Their multifaceted mechanisms and innovative delivery approaches make them potential candidates for treating vascular dysfunction and promoting healthy vascular aging. Further research is needed to translate these findings into clinical applications.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
21
|
Ji Y, Chen Z, Cai J. Roles and mechanisms of histone methylation in vascular aging and related diseases. Clin Epigenetics 2025; 17:35. [PMID: 39988699 PMCID: PMC11849368 DOI: 10.1186/s13148-025-01842-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
The global aging trend has posed significant challenges, rendering healthcare for older adults a crucial focus in medical research. Among the numerous health concerns related to aging, vascular aging and dysfunction are important risk factors and underlying causes of age-related diseases. Histone methylation and demethylation, which are involved in gene expression and cellular senescence, are closely associated with the occurrence and development of vascular aging. Consequently, this review aimed to identify the role of histone methylation in the pathogenesis of vascular aging and its potential for treating age-related vascular diseases and provided new insights into therapeutic strategies targeting the vascular system.
Collapse
Affiliation(s)
- Yufei Ji
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhenzhen Chen
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jun Cai
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Xu ZJ, Xu J, Lei WJ, Wang X, Zou QL, Lv LC, Liu C, Hu WM, Xiang YJ, Shen JY, Wei TM, Zeng CL. RANBP1 Regulates NOTCH3-Mediated Autophagy in High Glucose-Induced Vascular Smooth Muscle Cells. FRONT BIOSCI-LANDMRK 2025; 30:26850. [PMID: 40018934 DOI: 10.31083/fbl26850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/29/2024] [Accepted: 12/16/2024] [Indexed: 03/01/2025]
Abstract
BACKGROUND Vascular smooth muscle cells(VSMCs) phenotypic switching under hyperglycemic conditions accelerates atherosclerotic progression. Notch receptor 3(NOTCH3), a critical stabilizer of VSMC homeostasis implicated in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) pathogenesis, ensures vascular integrity; however, its interplay with RAN Binding Protein 1(RANBP1) during pathological hyperglycemia remains uncharacterized. We hypothesize that hyperglycemia-induced autophagic dysregulation is mechanistically governed by theNotch receptor 3 (NOTCH3)/RANBP1 axis, proliferative capacity, and apoptotic signaling in high glucose (HG)-stimulated VSMCs. The aim of this study was to elucidate the regulatory mechanisms of autophagy in VSMCs under HG conditions, with a focus on the NOTCH3/RANBP1 axis and its implications for vascular health. METHODS Bioinformatics analysis was performed on NOTCH3 sequencing data, including weighted gene co-expression network analysis (WGCNA), screening of differentially expressed genes (DEGs), and construction of a protein-protein interaction (PPI) network, to identify the key gene, RANBP1. In vitro experiments, including cell counting kit-8 (CCK-8) assays, quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting (WB), and flow cytometry, were conducted to examine the effects of NOTCH3 knockdown combined with RANBP1 overexpression on glucose-induced autophagy marker expression and cell viability in VSMCs. RESULTS NOTCH3 knockdown suppressed VSMC proliferation and induced apoptosis, and the cell cycle was stopped at the S phase. Analysis of VSMC sequencing data revealed 38 overlapping genes between the turquoise module and DEGs, 11 (HPF1, RANBP1, CRNKL1, LGALS3, RDX, ECM1, CXCL5, PA2G4, CENPS, ZNF830, and HIST1H4L) of which were significantly underexpressed in VSMC samples with si-NOTCH3. In a dose-dependent manner, HG therapy altered the expression of autophagy-related markers, upregulated NOTCH3, and downregulated phosphorylated mammalian target of rapamycin (p-mTOR). Downregulation of NOTCH3 aggravated the effects of HG on cell viability and autophagy, whereas overexpression of RANBP1 reversed these effects, suggesting an offsetting effect on HG-induced autophagy. CONCLUSION On the basis of sequencing technology, bioinformatics analysis and cell experiments, we conclude that the RANBP1/NOTCH3 axis is essential for the control of autophagy and survival of VSMCs under hyperglycemic stress and could provide new insight for the clinical treatment of VSMC-related diseases.
Collapse
Affiliation(s)
- Zhong-Jiao Xu
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Jian Xu
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Wen-Jing Lei
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Xiang Wang
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Qi-Lin Zou
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Lin-Chun Lv
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Chong Liu
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Wu-Ming Hu
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Yi-Jia Xiang
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Jia-Yi Shen
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Tie-Min Wei
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| | - Chun-Lai Zeng
- Department of Cardiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, 323000 Lishui, Zhejiang, China
| |
Collapse
|
23
|
Wang F, Du R, Shang Y. Biological function of d-tryptophan: a bibliometric analysis and review. Front Microbiol 2025; 15:1455540. [PMID: 39872820 PMCID: PMC11770058 DOI: 10.3389/fmicb.2024.1455540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
Background d-Tryptophan is recognised for its unique physiological properties. In this study, we aimed to explore the dynamic trends and emerging topics in d-tryptophan research to offer fresh perspectives for future studies. Methods Employing bibliometric analysis, we examined the literature on d-tryptophan indexed in the Web of Science Core Collection from January 1987 to December 2023. The "Bibliometrix" R package and CiteSpace were utilised for data processing. Results Analyses of 865 publications revealed 2209 keywords, 4068 authors, 2094 institutions, and contributors from 302 regions. The USA was at the forefront of publications concerning d-tryptophan, but the European Journal of Pharmacology, Journal of Biological Chemistry, and Journal of Medicinal Chemistry were notable for their contributions, co-citations, and impact, respectively. This literature review reveals that since 1987, studies have developed from a focus on d-tryptophan metabolism to the exploration of its functions in organic and medicinal chemistry and food science. Recent findings highlight the potential of d-tryptophan as a non-nutritional sweetener and food preservative as well as its role in inhibiting the growth of bacterial biofilms. Additionally, its immunomodulatory properties are being investigated in relation to allergic diseases. Furthermore, d-tryptophan plays a role in the therapy of atherosclerosis, osteoporosis, tuberculosis, and cancer. Conclusion The results of bibliometric analysis highlight that future research should focus on the biological functions of d-tryptophan as a food preservative and its use in immunomodulation and drug development, providing strong guidance for future research.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Runyu Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
24
|
Zhang M, Shi J, Pan H, Zhu J, Wang X, Zhou J, Deng H. F-53B stimulated vascular smooth muscle cell phenotypic switch and vascular remodeling via ferroptosis-related pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176565. [PMID: 39341237 DOI: 10.1016/j.scitotenv.2024.176565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
The compound 6:2 chlorinated polyfluorinated ether sulfonate (F53B), an alternative to perfluorooctane sulfonate (PFOS), has been widely utilized in China. Although the connection between the exposure and toxicity of F53B is established, the role and mechanisms of the compound in promoting vascular remodeling are yet to be elucidated. Thus, the present study investigated the impact of F53B on the function of vascular smooth muscle cells (VSMCs) and vascular remodeling. The data exhibited that F53B stimulates vascular morphological alterations in vivo, and exposure to the compound caused excessive VSMCs ferroptosis and phenotype switching, as determined using phenotype and molecular assays. Moreover, Fer-1 reversed F-53B-induced VSMC dysfunction and vascular remodeling. Furthermore, F53B activated the ferroptosis-related pathway, encompassing ATR expression and LOC101929922/miR-542-3p/ACSL4 pathway. Thus, the current results elaborated on the multifaceted toxicities of F53B that induce vascular remodeling, thereby necessitating the assessment of vasotoxicity risks associated with the compound.
Collapse
Affiliation(s)
- Min Zhang
- Institute of Cardiovascular Diseases, Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China.
| | - Jun Shi
- Shanghai Institute of Pollution Control and Ecological Security, Key Laboratory of Yangtze River Water Environment Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| | - Huichao Pan
- Institute of Cardiovascular Diseases, Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Jie Zhu
- Center for Translational Neurodegeneration and Regenerative Therapy, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Xueting Wang
- Institute of Cardiovascular Diseases, Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Jun Zhou
- Institute of Cardiovascular Diseases, Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 200336, China
| | - Huiping Deng
- Shanghai Institute of Pollution Control and Ecological Security, Key Laboratory of Yangtze River Water Environment Ministry of Education, College of Environmental Science and Engineering, Tongji University, Shanghai, China
| |
Collapse
|
25
|
Song J, Cao C, Wang Z, Li H, Yang L, Kang J, Meng H, Li L, Liu J. Mechanistic insights into the regression of atherosclerotic plaques. Front Physiol 2024; 15:1473709. [PMID: 39628943 PMCID: PMC11611857 DOI: 10.3389/fphys.2024.1473709] [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: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases and mortality globally. The progression of atherosclerotic disease results in the expansion of plaques and the development of necrotic cores. Subsequent plaque rupture can lead to thrombosis, occluding blood vessels, and end-organ ischemia with consequential ischemic injury. Atherosclerotic plaques are formed by the accumulation of lipid particles overloaded in the subendothelial layer of blood vessels. Abnormally elevated blood lipid levels and impaired endothelial function are the initial factors leading to atherosclerosis. The atherosclerosis research has never been interrupted, and the previous view was that the pathogenesis of atherosclerosis is an irreversible and chronic process. However, recent studies have found that the progression of atherosclerosis can be halted when patients' blood lipid levels are reversed to normal or lower. A large number of studies indicates that it can inhibit the progression of atherosclerosis lesions and promote the regression of atherosclerotic plaques and necrotic cores by lowering blood lipid levels, improving the repair ability of vascular endothelial cells, promoting the reverse cholesterol transport in plaque foam cells and enhancing the ability of macrophages to phagocytize and clear the necrotic core of plaque. This article reviews the progress of research on the mechanism of atherosclerotic plaque regression. Our goal is to provide guidance for developing better therapeutic approaches to atherosclerosis by reviewing and analyzing the latest scientific findings.
Collapse
Affiliation(s)
- Jianshu Song
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ce Cao
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Ziyan Wang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Haoran Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Lili Yang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jing Kang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Meng
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Lei Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jianxun Liu
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
26
|
Han X, Xu S, Hu K, Yu Y, Wang X, Qu C, Yang B, Liu X. Early growth response 1 exacerbates thoracic aortic aneurysm and dissection of mice by inducing the phenotypic switching of vascular smooth muscle cell through the activation of Krüppel-like factor 5. Acta Physiol (Oxf) 2024; 240:e14237. [PMID: 39345002 DOI: 10.1111/apha.14237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/15/2024] [Accepted: 09/14/2024] [Indexed: 10/01/2024]
Abstract
AIM Vascular smooth muscle cell (VSMC) phenotypic switching has been reported to regulate vascular function and thoracic aortic aneurysm and dissection (TAAD) progression. Early growth response 1 (Egr1) is associated with the differentiation of VSMCs. However, the mechanisms through which Egr1 participates in the regulation of VSMCs and progression of TAAD remain unknown. This study aimed to investigate the role of Egr1 in the phenotypic switching of VSMCs and the development of TAAD. METHODS Wild-type C57BL/6 and SMC-specific Egr1-knockout mice were used as experimental subjects and fed β-aminopropionitrile for 4 weeks to construct the TAAD model. Ultrasound and aortic staining were performed to examine the pathological features in thoracic aortic tissues. Transwell, wound healing, CCK8, and immunofluorescence assays detected the migration and proliferation of synthetic VSMCs. Egr1 was directly bound to the promoter of Krüppel-like factor 5 (KLF5) and promoted the expression of KLF5, which was validated by JASPAR database and dual-luciferase reporter assay. RESULTS Egr1 expression increased and was partially co-located with VSMCs in aortic tissues of mice with TAAD. SMC-specific Egr1 deficiency alleviated TAAD and inhibited the phenotypic switching of VSMC. Egr1 knockdown prevented the phenotypic switching of VSMCs and subsequently suppressed the migration and proliferation of synthetic VSMCs. The inhibitory effects of Egr1 deficiency on VSMCs were blunted once KLF5 was overexpressed. CONCLUSION Egr1 aggravated the development of TAAD by promoting the phenotypic switching of VSMCs via enhancing the transcriptional activation of KLF5. These results suggest that inhibition of SMC-specific Egr1 expression is a promising therapy for TAAD.
Collapse
MESH Headings
- Animals
- Early Growth Response Protein 1/metabolism
- Early Growth Response Protein 1/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Aortic Dissection/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Male
- Myocytes, Smooth Muscle/metabolism
- Disease Models, Animal
- Cell Proliferation
Collapse
Affiliation(s)
- Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shengnan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Ke Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
27
|
Mansour H, Slika H, Nasser SA, Pintus G, Khachab M, Sahebkar A, Eid AH. Flavonoids, gut microbiota and cardiovascular disease: Dynamics and interplay. Pharmacol Res 2024; 209:107452. [PMID: 39383791 DOI: 10.1016/j.phrs.2024.107452] [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: 01/28/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Cardiovascular disease (CVD) remains the leading cause of global morbidity and mortality. Extensive efforts have been invested to explicate mechanisms implicated in the onset and progression of CVD. Besides the usual suspects as risk factors (obesity, diabetes, and others), the gut microbiome has emerged as a prominent and essential factor in the pathogenesis of CVD. With its endocrine-like effects, the microbiome modulates many physiologic processes. As such, it is not surprising that dysbiosis-by generating metabolites, inciting inflammation, and altering secondary bile acid signaling- could predispose to or aggravate CVD. Nevertheless, various natural and synthetic compounds have been shown to modulate the microbiome. Prime among these molecules are flavonoids, which are natural polyphenols mainly present in fruits and vegetables. Accumulating evidence supports the potential of flavonoids in attenuating the development of CVD. The ascribed mechanisms of these compounds appear to involve mitigation of inflammation, alteration of the microbiome composition, enhancement of barrier integrity, induction of reverse cholesterol transport, and activation of farnesoid X receptor signaling. In this review, we critically appraise the methods by which the gut microbiome, despite being essential to the human body, predisposes to CVD. Moreover, we dissect the mechanisms and pathways underlying the cardioprotective effects of flavonoids.
Collapse
Affiliation(s)
- Hadi Mansour
- Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Gianfranco Pintus
- Department of Biomedical Sciences, University of Sassari, Sassari 07100, Italy
| | - Maha Khachab
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Beirut, Lebanon
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
28
|
Al Zein M, Khazzeka A, El Khoury A, Al Zein J, Zoghaib D, Eid AH. Revisiting high-density lipoprotein cholesterol in cardiovascular disease: Is too much of a good thing always a good thing? Prog Cardiovasc Dis 2024; 87:50-59. [PMID: 39442601 DOI: 10.1016/j.pcad.2024.10.009] [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: 10/19/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Cardiovascular disease (CVD) continues to be a leading cause of global mortality and morbidity. Various established risk factors are linked to CVD, and modifying these risk factors is fundamental in CVD management. Clinical studies underscore the association between dyslipidemia and CVD, and therapeutic interventions that target low-density lipoprotein cholesterol elicit clear benefits. Despite the correlation between low high-density lipoprotein cholesterol (HDLC) and heightened CVD risk, HDL-raising therapies have yet to showcase significant clinical benefits. Furthermore, evidence from epidemiological and genetic studies reveals that not only low HDL-C levels, but also very high levels of HDL-C are linked to increased risk of CVD. In this review, we focus on HDL metabolism and delve into the relationship between HDL and CVD, exploring HDL functions and the observed alterations in its roles in disease. Altogether, the results discussed herein support the conventional wisdom that "too much of a good thing is not always a good thing". Thus, our recommendation is that a careful reconsideration of the impact of high HDL-C levels is warranted, and shall be revisited in future research.
Collapse
Affiliation(s)
- Mohammad Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Alicia Khazzeka
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Dima Zoghaib
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
29
|
Wang M, Mo D, Zhou C, Guo M, Zhang W, Chen R, Xu J, Zhang N, Yu H. Association between Ward's triangle bone mineral density levels and abdominal aortic calcification: Data from the national health and nutrition examination survey 2013-2014. Clin Nutr ESPEN 2024; 64:344-357. [PMID: 39461592 DOI: 10.1016/j.clnesp.2024.10.154] [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: 07/23/2024] [Revised: 09/05/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND & AIMS Despite extensive research into the cardiovascular implications of abdominal aortic calcification (AAC), there is a scarcity of robust studies exploring its association with Ward's triangle bone mineral density (BMD). This study aimed to evaluate this relationship in a nationally representative sample and compare the predictive value with femoral neck BMD and total femur BMD. METHODS We conducted a cross-sectional analysis of 2013-2014 National Health and Nutrition Examination Survey (NHANES) data, utilizing a complex, stratified, multistage, cluster sampling design. BMD measurements at Ward's triangle, femoral neck, and total femur were assessed using DXA scans. AAC severity was defined by a Kauppila score of ≥5. Predictors of AAC-24 scores were identified through correlation and linear regression models. Stratified regression and restricted cubic splines were applied to explore subgroup and dose-response relationships. RESULTS Of the 2965 participants representing 116, 562, 500 individuals in the U.S., 11 % had severe AAC. Ward's triangle BMD showed a significant negative association with AAC-24 scores (β = -1.90, 95 % CI: -2.80 to -1.00, P < 0.002) and a reduced risk of severe AAC (OR = 0.85, 95 % CI: 0.76 to 0.95, P = 0.010). Non-linear associations were observed between Ward's triangle BMD and AAC outcomes. Ward's triangle BMD outperformed femoral neck and total femur BMD in predicting AAC. CONCLUSIONS Higher Ward's triangle BMD is linked to lower odds of severe AAC, highlighting its potential for improved early detection of AAC over femoral neck and total femur BMD. Healthcare providers should consider the implications of reduced Ward's triangle BMD for systemic atherosclerosis and recommend early AAC screening for enhanced cardiovascular risk management.
Collapse
Affiliation(s)
- Mengmeng Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao 266001, Shandong, China.
| | - Degang Mo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Chi Zhou
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Wenqiang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Rui Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jiachao Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Ning Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China; Institute of Cardiovascular Disease, Qingdao University, Qingdao 266001, Shandong, China.
| |
Collapse
|
30
|
Marway PS, Tjahjadi N, Campello Jorge CA, Knauer H, Spahlinger G, Masotti M, Eagle KA, Hofmann Bowman MA, Patel HJ, Burris NS. Baseline Diameter Does Not Predict Growth Rate in a Presurgical Ascending Thoracic Aortic Aneurysm Population. J Am Heart Assoc 2024; 13:e036896. [PMID: 39392167 PMCID: PMC11935589 DOI: 10.1161/jaha.124.036896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Patients with ascending thoracic aortic aneurysm are recommended to undergo routine imaging surveillance. Although maximal diameter is the primary metric of disease severity, recent American College of Cardiology/American Heart Association guidelines emphasize the importance of aortic growth in determining surgical candidacy and risk. As diameter increases, it is assumed that aortic growth rate accelerates because of increased wall tension; however, this relationship is poorly studied. We aim to investigate the relationship between ascending thoracic aortic aneurysm diameter and growth rate using vascular deformation mapping, a validated technique for 3-dimensional growth mapping with submillimeter accuracy. METHODS AND RESULTS We retrospectively identified adult patients with ascending aortic dilation (≥4.0 cm) and serial gated computed tomography angiograms separated by ≥2 years, excluding confirmed heritable thoracic aortic disease. Ascending growth rate was defined as 90th percentile radial wall deformation by vascular deformation mapping. Maximal diameter measurements were derived from the baseline computed tomography angiogram, and aortic length and body size-adjusted indexes were calculated. Among 258 included patients (63.2% men; age of 63 years [interquartile range, 55-69 years]), mean±SD baseline diameter was 46.3±3.6 mm and median growth rate was 0.21 mm/year (interquartile range, 0.13-0.38 mm/year). No correlation was noted between growth rate and baseline diameter (r=0.02, P=0.74) or other aortic size metrics. On multivariate analysis, age was independently predictive of growth rate (β=-0.007, P=0.021), alongside weight (β=0.003, P=0.016) and the presence of moderate or severe aortic valve insufficiency (β=0.146, P=0.049). CONCLUSIONS Maximal aortic diameter is not predictive of aortic growth rate, in this contemporary cohort of patients with sizes under current surgical thresholds (<55 mm).
Collapse
Affiliation(s)
| | | | | | - Heather Knauer
- Department of RadiologyUniversity of MichiganAnn ArborMI
| | | | - Maria Masotti
- Biostatistics, School of Public HealthUniversity of MichiganAnn ArborMI
| | - Kim A. Eagle
- Department of Internal Medicine—CardiologyUniversity of MichiganAnn ArborMI
| | | | | | | |
Collapse
|
31
|
Kane J, Lemieux A, Baranwal G, Misra S. The Role of Cardio-Renal Inflammation in Deciding the Fate of the Arteriovenous Fistula in Haemodialysis Therapy. Cells 2024; 13:1637. [PMID: 39404400 PMCID: PMC11475948 DOI: 10.3390/cells13191637] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/19/2024] Open
Abstract
Vascular access is an indispensable component of haemodialysis therapy for end-stage kidney disease patients. The arteriovenous fistula (AVF) is most common, but importantly, two-year failure rates are greater than fifty percent. AVF failure can occur due to a lack of suitable vascular remodelling, and inappropriate inflammation preventing maturation, or alternatively neointimal hyperplasia and vascular stenosis preventing long-term use. A comprehensive mechanistic understanding of these processes is still lacking, but recent studies highlight an essential role for inflammation from uraemia and the AVF itself. Inflammation affects each cell in the cascade of AVF failure, the endothelium, the infiltrating immune cells, and the vascular smooth muscle cells. This review examines the role of inflammation in each cell step by step and the influence on AVF failure. Inflammation resulting in AVF failure occurs initially via changes in endothelial cell activation, permeability, and vasoprotective chemokine secretion. Resultingly, immune cells can extravasate into the subendothelial space to release inflammatory cytokines and cause other deleterious changes to the microenvironment. Finally, all these changes modify vascular smooth muscle cell function, resulting in excessive and unchecked hyperplasia and proliferation, eventually leading to stenosis and the failure of the AVF. Finally, the emerging therapeutic options based off these findings are discussed, including mesenchymal stem cells, small-molecule inhibitors, and far-infrared therapies. Recent years have clearly demonstrated a vital role for inflammation in deciding the fate of the AVF, and future works must be centred on this to develop therapies for a hitherto unacceptably underserved patient population.
Collapse
Affiliation(s)
| | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA; (J.K.); (A.L.); (G.B.)
| |
Collapse
|
32
|
Matsumoto T, Nagano T, Taguchi K, Kobayashi T, Tanaka-Totoribe N. Toll-like receptor 3 involvement in vascular function. Eur J Pharmacol 2024; 979:176842. [PMID: 39033837 DOI: 10.1016/j.ejphar.2024.176842] [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: 05/12/2024] [Revised: 06/24/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Maintaining endothelial cell (EC) and vascular smooth muscle cell (VSMC) integrity is an important component of human health and disease because both EC and VSMC regulate various functions, including vascular tone control, cellular adhesion, homeostasis and thrombosis regulation, proliferation, and vascular inflammation. Diverse stressors affect functions in both ECs and VSMCs and abnormalities of functions in these cells play a crucial role in cardiovascular disease initiation and progression. Toll-like receptors (TLRs) are important detectors of pathogen-associated molecular patterns derived from various microbes and viruses as well as damage-associated molecular patterns derived from damaged cells and perform innate immune responses. Among TLRs, several studies reveal that TLR3 plays a key role in initiation, development and/or protection of diseases, and an emerging body of evidence indicates that TLR3 presents components of the vasculature, including ECs and VSMCs, and plays a functional role. An agonist of TLR3, polyinosinic-polycytidylic acid [poly (I:C)], affects ECs, including cell death, inflammation, chemoattractant, adhesion, permeability, and hemostasis. Poly (I:C) also affects VSMCs including inflammation, proliferation, and modulation of vascular tone. Moreover, alterations of vascular function induced by certain molecules and/or interventions are exerted through TLR3 signaling. Hence, we present the association between TLR3 and vascular function according to the latest studies.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan.
| | - Takayuki Nagano
- Second Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Naoko Tanaka-Totoribe
- First Department of Pharmacology, School of Pharmaceutical Sciences, Kyushu University of Medical Science, Nobeoka, Miyazaki, 882-8508, Japan
| |
Collapse
|
33
|
Gao H, Zhao L, Cao X, Zhang G. Differential miR-195-5p and its potential role during the development of carotid artery stenosis. Vascular 2024:17085381241273320. [PMID: 39140317 DOI: 10.1177/17085381241273320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
OBJECTIVES Carotid artery stenosis (CAS) is a leading cause of cerebral ischemic events (CIE). Timely detection and risk assessment can aid in managing CAS patients and improving their prognosis. The aim of the current study is to identify a new biomarker for CAS and to further investigate the impact of miR-195-5p on cellular processes in vascular smooth muscle cells (VSMCs). METHODS This study involved 112 CAS patients and 65 healthy individuals. Serum miR-195-5p levels were measured using RT-qPCR. The ROC curve was then plotted to evaluate the diagnostic potential of miR-195-5p for CAS. The Kaplan-Meier curve and Cox regression were employed to determine miR-195-5p's prognostic significance. In vitro, the effects of miR-195-5p mimic or inhibitor on VSMC proliferation and migration were assessed using CCK-8 and Transwell assays. RESULTS In CAS patients, serum miR-195-5p levels were elevated and correlated with the degree of CAS. The ROC curve had an AUC value of 0.897, with sensitivity of 71.4% and specificity of 95.4%. Higher levels of miR-195-5p indicated a higher risk of CIE occurrence and may serve as an independent predictor of CIE. The upregulation of miR-195-5p promoted VSMC proliferation and migration, while downregulation had the opposite effect. CONCLUSIONS miR-195-5p was demonstrated to have diagnostic and prognostic significance in CAS and may serve as a potential biomarker. It may contribute to the progression of CAS by promoting the proliferation and migration of VSMCs.
Collapse
Affiliation(s)
- Haining Gao
- Department of Neurology, Nanjing Qixia District Hospital, Nanjing, China
| | - Liansheng Zhao
- Department of Neurology, Nanjing Qixia District Hospital, Nanjing, China
| | - Xuemei Cao
- Department of Neurology, Nanjing Qixia District Hospital, Nanjing, China
| | - Guoxin Zhang
- Department of Neurology, Nanjing Qixia District Hospital, Nanjing, China
| |
Collapse
|
34
|
Dai Y, Junho CVC, Schieren L, Wollenhaupt J, Sluimer JC, van der Vorst EPC, Noels H. Cellular metabolism changes in atherosclerosis and the impact of comorbidities. Front Cell Dev Biol 2024; 12:1446964. [PMID: 39188527 PMCID: PMC11345199 DOI: 10.3389/fcell.2024.1446964] [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: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/28/2024] Open
Abstract
Cell activation and nutrient dysregulation are common consequences of atherosclerosis and its preceding risk factors, such as hypertension, dyslipidemia, and diabetes. These diseases may also impact cellular metabolism and consequently cell function, and the other way around, altered cellular metabolism can impact disease development and progression through altered cell function. Understanding the contribution of altered cellular metabolism to atherosclerosis and how cellular metabolism may be altered by co-morbidities and atherosclerosis risk factors could support the development of novel strategies to lower the risk of CVD. Therefore, we briefly review disease pathogenesis and the principles of cell metabolic pathways, before detailing changes in cellular metabolism in the context of atherosclerosis and comorbidities. In the hypoxic, inflammatory and hyperlipidemic milieu of the atherosclerotic plaque riddled with oxidative stress, metabolism shifts to increase anaerobic glycolysis, the pentose-phosphate pathway and amino acid use. We elaborate on metabolic changes for macrophages, neutrophils, vascular endothelial cells, vascular smooth muscle cells and lymphocytes in the context of atherosclerosis and its co-morbidities hypertension, dyslipidemia, and diabetes. Since causal relationships of specific key genes in a metabolic pathway can be cell type-specific and comorbidity-dependent, the impact of cell-specific metabolic changes must be thoroughly explored in vivo, with a focus on also systemic effects. When cell-specific treatments become feasible, this information will be crucial for determining the best metabolic intervention to improve atherosclerosis and its interplay with co-morbidities.
Collapse
Affiliation(s)
- Yusang Dai
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Physical Examination Center, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Carolina Victoria Cruz Junho
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Luisa Schieren
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Julia Wollenhaupt
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
| | - Judith C. Sluimer
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for Cardiorenal Disease (AMICARE), RWTH Aachen Campus, Aachen, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands
| |
Collapse
|