1
|
Wang X, Liu Q, Cheng P, Yang T, Zhao T, Liu M, Dai E, Sha W, Yuan J, Rong J, Qu H, Zhou H. LuQi formula ameliorates pressure overload-induced heart failure by regulating macrophages and regulatory T cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156527. [PMID: 40118747 DOI: 10.1016/j.phymed.2025.156527] [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: 09/02/2024] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Inflammatory macrophages in failing myocardium secrete CCL17, which targets CCR4 in immunosuppressive Tregs and inhibits the intracellular second messenger ARRB2-mediated cardiac chemotaxis. Traditional Chinese medicine (TCM) LuQi formula (LQF) is safe and effective in treating heart failure (HF). This study aims to elucidate the cardioprotective mechanism of LQF through its modulation of cardiac macrophages and Tregs. METHODS In vivo, the HF mouse model was established via transverse aortic constriction (TAC), with the superagonistic anti-CD28 monoclonal antibody (CD28-SA)-induced Tregs expansion as a positive control. Proteomics analysis elucidated the core link of LQF in anti-HF. In vitro, bone marrow-derived macrophages (BMDMs) were isolated, and Naive CD4+T cells were sorted and stimulated to differentiate into Tregs. The pharmacological mechanism of LQF was confirmed through histological and molecular biology experiments. RESULTS Proteomics reveals that LQF modulates the immune microenvironment of failing myocardium. We revealed that LQF inhibited cardiac inflammatory macrophage infiltration and NF-κB (p50, p65)/CCL17 axis expression, and promoted cardiac Tregs recruitment against HF, with the comparable efficacy of CD-SA28-induced Tregs expansion. Mechanistically, LQF inhibited the NF-κB activator 1-induced NF-κB (p50, p65)/CCL17 axis overexpression, and JSH-23 (NF-κB Inhibitor) abolished NF-κB (p50, p65)/CCL17 axis expression in inflammatory macrophages. Furthermore, the inhibition of CCL17 expression in inflammatory macrophages by LQF was found to be mediated by NF-κB (p50, p65). LQF concentration-dependently promoted Tregs CD73/Foxp3 axis expression, enhancing Tregs immunosuppressive function. LQF activated CCR4-ARRB2 complex and CCR4/ARRB2 axis expression in Tregs. Although AZD2098 (CCR4 Inhibitor) blocked CCR4 expression and CCR4-ARRB2 complex, LQF promoted ARRB2-mediated Tregs cardiac chemotaxis independent of the CCR4. We revealed that NF-κB p50SEP337-CCL17, NF-κB p65SEP536-CCL17, and CCR4-ARRB2 highly bound subunit interface targets. Molecular docking analysis demonstrated that the LQF's active ingredients exhibit good binding affinity with the NF-κB (p50, p65) /CCL17 axis in macrophages and Foxp3 in Tregs. CONCLUSION LQF has the potential to enhance the cardiac immune microenvironment and effectively prevent and treat HF by modulating both innate and adaptive immune responses. It achieves this by inhibiting the infiltration of inflammatory macrophages, suppressing the NF-κB (p50, p65)/CCL17 axis, and promoting Tregs recruitment. The active ingredients of LQF provide valuable candidate compounds for developing new anti-HF drugs. Furthermore, CD-28SA-induced Tregs expansion showed cardioprotective effects in TAC-induced non-ischemic HF models.
Collapse
Affiliation(s)
- Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200040, China
| | - Tingyao Zhao
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Meng Liu
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Enrui Dai
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanjing Sha
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinfeng Yuan
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jingfeng Rong
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huiyan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Dai Z, Ding H, Zhang Q, Fu L, Tai S. Spatial Insights in Cardiovascular Aging. Aging Dis 2025:AD.2025.0272. [PMID: 40423633 DOI: 10.14336/ad.2025.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Spatial omics provides unprecedented insights into how the cardiovascular system is spatially organized and how cellular phenotypes are distributed. Researchers have been able to clarify the complex spatial architecture of the cardiovascular system and how cellular phenotypes are distributed during the aging process by integrating data from spatial omics. In addition, this new technology has revealed previously hidden patterns of gene expression and cellular communication that were not detected using traditional bulk omics approaches. In this review, we explore the contribution of spatial omics in clarifying the molecular mechanisms that influence cardiovascular aging, highlighting the importance and application of spatial omics in unraveling the spatial heterogeneity within the aging cardiovascular system. This will help us understand the molecular mechanisms implicated in age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Zhongling Dai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Huiqin Ding
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Quan Zhang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Liyao Fu
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shi Tai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
3
|
Zhang Z, Wang Y, Chen X, Wu C, Zhou J, Chen Y, Liu X, Tang X. The aging heart in focus: The advanced understanding of heart failure with preserved ejection fraction. Ageing Res Rev 2024; 101:102542. [PMID: 39396676 DOI: 10.1016/j.arr.2024.102542] [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: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for 50 % of heart failure (HF) cases, making it the most common type of HF, and its prevalence continues to increase in the aging society. HFpEF is a systemic syndrome resulting from many risk factors, such as aging, metabolic syndrome, and hypertension, and its clinical features are highly heterogeneous in different populations. HFpEF syndrome involves the dysfunction of multiple organs, including the heart, lung, muscle, and vascular system. The heart shows dysfunction of various cells, including cardiomyocytes, endothelial cells, fibroblasts, adipocytes, and immune cells. The complex etiology and pathobiology limit experimental research on HFpEF in animal models, delaying a comprehensive understanding of the mechanisms and making treatment difficult. Recently, many scientists and cardiologists have attempted to improve the clinical outcomes of HFpEF. Recent advances in clinically related animal models and systemic pathology studies have improved our understanding of HFpEF, and clinical trials involving sodium-glucose cotransporter 2 inhibitors have significantly enhanced our confidence in treating HFpEF. This review provides an updated comprehensive discussion of the etiology and pathobiology, molecular and cellular mechanisms, preclinical animal models, and therapeutic trials in animals and patients to enhance our understanding of HFpEF and improve clinical outcomes.
Collapse
Affiliation(s)
- Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chuan Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Jingyue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Yan Chen
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China.
| |
Collapse
|
4
|
Longtine AG, Greenberg NT, Gonzalez A, Lindquist A, VanDongen NS, Mahoney SA, Rahman G, Clayton ZS, Ziemba BP, Ludwig KR, Widlansky ME, Knight R, Seals DR, Brunt VE. Oral Supplementation with the Short-Chain Fatty Acid Acetate Ameliorates Age-Related Arterial Dysfunction in Mice. AGING BIOLOGY 2024; 2:20240033. [PMID: 39897133 PMCID: PMC11785404 DOI: 10.59368/agingbio.20240033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Adverse changes in the gut microbiome with aging are an emerging mediator of arterial dysfunction, which contributes to cardiovascular disease (CVD) development. We investigated the therapeutic potential of enhancing the bioavailability of gut-derived short-chain fatty acids (SCFAs; produced from dietary fiber) for improving age-related arterial dysfunction. We performed gut microbial whole-genome sequencing in young (3 months) versus old (24 months) male C57BL/6N mice to explore changes in bacterial taxonomic abundance and functional pathways with aging and relations to arterial function. We then supplemented young and old mice with the SCFA acetate in drinking water versus controls and versus a high-fiber diet for 8-10 weeks to test the effects of these interventions on vascular function and explore potential mechanisms. Of the various differences in the gut microbiomes of old mice, lower SCFA-producing capacity (taxonomic abundance and functional pathways) stood out as a key feature related to worse arterial function after adjusting for age. Acetate supplementation and a high-fiber diet reversed ~30% of the age-related increase in aortic pulse wave velocity (stiffness) and fully restored carotid artery endothelium-dependent dilation (endothelial function) to young levels. Acetate and a high-fiber diet reduced age-related increases in systemic inflammation. We also found that improvements in endothelial function were likely mediated by suppressed early growth response-1 signaling using innovative siRNA-based knockdown in isolated arteries. There were no effects of the interventions in young mice. Acetate supplementation was comparably effective for ameliorating arterial dysfunction with aging as a high-fiber diet and thus shows promise for reducing CVD risk in older adults.
Collapse
Affiliation(s)
- Abigail G. Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Nathan T. Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Alexandra Lindquist
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Nicholas S. VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Sophia A. Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Gibraan Rahman
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Zachary S. Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Brian P. Ziemba
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Katelyn R. Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Michael E. Widlansky
- Departments of Medicine and Pharmacology and the Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, Department of Computer Science and Engineering, and Halıcıoğlu Data Science Institute, and Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Douglas R. Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Vienna E. Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Division of Renal Diseases and Hypertension, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
5
|
Shi L, Li G, Hou N, Tu L, Li J, Luo J, Hu S. APOB and CCL17 as mediators in the protective effect of SGLT2 inhibition against myocardial infarction: Insights from proteome-wide mendelian randomization. Eur J Pharmacol 2024; 976:176619. [PMID: 38679119 DOI: 10.1016/j.ejphar.2024.176619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
AIMS Sodium-glucose cotransporter 2 (SGLT2) inhibitors offer a novel therapeutic avenue for myocardial infarction (MI). However, the exact nature of this relationship and the underlying mechanisms are not fully understood. METHODS Utilizing a two-sample Mendelian Randomization (MR) analysis, we elucidated the causal effects stemming from the inhibition of SGLT2 on MI. Then, The pool of 4907 circulating proteins within the plasma proteome were utilized to explore the mediators of SGLT2 inhibitors on MI. Protein-protein network and enrichment analysis were conducted to clarify the potential mechanism. Finally, employing MR analysis and meta-analysis techniques, we systematically assessed the causal associations between SGLT2 inhibition and coronary heart diseases (CHD). RESULTS SGLT2 inhibition (per 1 SD decrement in HbA1c) was associated with reduced risk of MI (odds ratio [OR] = 0.462, [95% CI 0.222, 0.958], P = 0.038). Among 4907 circulating proteins, we identified APOB and CCL17 which were related to both SGLT2 inhibition and MI. Mediation analysis showed evidence of the indirect effect of SGLT2 inhibition on MI through APOB (β = -0.557, 95%CI [-1.098, -0.155]) with a mediated proportion of 72%, and CCL17 (β = -0.176, 95%CI [-0.332, -0.056]) with a mediated proportion of 17%. The meta-analysis result showed that SGLT2 inhibition was associated with a lower risk of CHD. CONCLUSION Based on proteome-wide mendelian randomization, APOB and CCL17 were seen as mediators in the protective effect of SGLT2 inhibition against myocardial infarction.
Collapse
Affiliation(s)
- Lili Shi
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Gen Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ningxin Hou
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Tu
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China
| | - Jun Li
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinlan Luo
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China.
| | - Shuiqing Hu
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, China; Division of Cardiology and Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
6
|
Nagar N, Naidu G, Panda SK, Gulati K, Singh RP, Poluri KM. Elucidating the role of chemokines in inflammaging associated atherosclerotic cardiovascular diseases. Mech Ageing Dev 2024; 220:111944. [PMID: 38782074 DOI: 10.1016/j.mad.2024.111944] [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: 01/31/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Age-related inflammation or inflammaging is a critical deciding factor of physiological homeostasis during aging. Cardiovascular diseases (CVDs) are exquisitely associated with aging and inflammation and are one of the leading causes of high mortality in the elderly population. Inflammaging comprises dysregulation of crosstalk between the vascular and cardiac tissues that deteriorates the vasculature network leading to development of atherosclerosis and atherosclerotic-associated CVDs in elderly populations. Leukocyte differentiation, migration and recruitment holds a crucial position in both inflammaging and atherosclerotic CVDs through relaying the activity of an intricate network of inflammation-associated protein-protein interactions. Among these interactions, small immunoproteins such as chemokines play a major role in the progression of inflammaging and atherosclerosis. Chemokines are actively involved in lymphocyte migration and severe inflammatory response at the site of injury. They relay their functions via chemokine-G protein-coupled receptors-glycosaminoglycan signaling axis and is a principal part for the detection of age-related atherosclerosis and related CVDs. This review focuses on highlighting the detailed intricacies of the effects of chemokine-receptor interaction and chemokine oligomerization on lymphocyte recruitment and its evident role in clinical manifestations of atherosclerosis and related CVDs. Further, the role of chemokine mediated signaling for formulating next-generation therapeutics against atherosclerosis has also been discussed.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Goutami Naidu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Santosh Kumar Panda
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Khushboo Gulati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India
| | - Ravindra Pal Singh
- Department of Industrial Biotechnology, Gujarat Biotechnology University, Gujarat International Finance Tec-City, Gandhinagar, Gujarat 382355, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India; Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India.
| |
Collapse
|
7
|
Wen Y, Chen H, Wang Y, Sun Y, Dou F, Du X, Liu T, Chen C. Extracellular vesicle-derived TP53BP1, CD34, and PBX1 from human peripheral blood serve as potential biomarkers for the assessment and prediction of vascular aging. Hereditas 2024; 161:3. [PMID: 38173016 PMCID: PMC10763334 DOI: 10.1186/s41065-023-00306-8] [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: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Vascular aging is an important pathophysiological basis for the senescence of various organs and systems in the human body, and it is a common pathogenetic trigger for many chronic diseases in the elderly. METHODS The extracellular vesicles (EVs) from young and aged umbilical vein endothelial cells were isolated and identified by qPCR the differential expression levels of 47 mRNAs of genes closely related to aging in the two groups. RESULTS There were significant differences in the expression levels of 18 genes (we noted upregulation in PLA2G12A, TP53BP1, CD144, PDE11A, FPGT, SERPINB4, POLD1, and PPFIBP2 and downregulation in ATP2C2, ROBO2, RRM2, GUCY1B1, NAT1-14, VEGFR2, WTAPP1, CD146, DMC1, and GRIK2). Subsequent qPCR identification of the above-mentioned genes in PBMCs and plasma-EVs from the various age groups revealed that the trend in expression levels in peripheral blood plasma-EVs of the different age groups was approximately the same as that in PBMCs. Of these mRNAs, the expression of four genes-PLA2G12A, TP53BP1, OPRL1, and KIAA0895-was commensurate with increasing age. In contradistinction, the expression trend of four genes (CREG1, PBX1, CD34, and SLIT2) was inversely proportional to the increase in age. Finally, by taking their intersection, we determined that the expression of TP53BP1 was upregulated with increasing human age and that CD34 and PBX1 were downregulated with increasing age. CONCLUSION Our study indicates that human peripheral blood plasma-EV-derived TP53BP1, CD34, and PBX1 potentially comprise a noninvasive biomarker for assessing and predicting vascular aging.
Collapse
Affiliation(s)
- Yichao Wen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, Shanghai, 200031, China
| | - Haiyang Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, Shanghai, 200031, China
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yiqing Sun
- Eberly College of Science, Penn State University, University Park, PA, USA
| | - Fangfang Dou
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, Shanghai, 200031, China
| | - Xiling Du
- School of Life Science and Technology, Tongji University, Shanghai, 200092, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, Shanghai, 200031, China.
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 365 South Xiangyang Road, Shanghai, 200031, China.
| |
Collapse
|
8
|
Aging Biomarker Consortium, Zhang W, Che Y, Tang X, Chen S, Song M, Wang L, Sun AJ, Chen HZ, Xu M, Wang M, Pu J, Li Z, Xiao J, Cao CM, Zhang Y, Lu Y, Zhao Y, Wang YJ, Zhang C, Shen T, Zhang W, Tao L, Qu J, Tang YD, Liu GH, Pei G, Li J, Cao F. A biomarker framework for cardiac aging: the Aging Biomarker Consortium consensus statement. LIFE MEDICINE 2023; 2:lnad035. [PMID: 39872891 PMCID: PMC11749273 DOI: 10.1093/lifemedi/lnad035] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 09/26/2023] [Indexed: 01/30/2025]
Abstract
Cardiac aging constitutes a significant risk factor for cardiovascular diseases prevalent among the elderly population. Urgent attention is required to prioritize preventive and management strategies for age-related cardiovascular conditions to safeguard the well-being of elderly individuals. In response to this critical challenge, the Aging Biomarker Consortium (ABC) of China has formulated an expert consensus on cardiac aging biomarkers. This consensus draws upon the latest scientific literature and clinical expertise to provide a comprehensive assessment of biomarkers associated with cardiac aging. Furthermore, it presents a standardized methodology for characterizing biomarkers across three dimensions: functional, structural, and humoral. The functional dimension encompasses a broad spectrum of markers that reflect diastolic and systolic functions, sinus node pacing, neuroendocrine secretion, coronary microcirculation, and cardiac metabolism. The structural domain emphasizes imaging markers relevant to concentric cardiac remodeling, coronary artery calcification, and epicardial fat deposition. The humoral aspect underscores various systemic (N) and heart-specific (X) markers, including endocrine hormones, cytokines, and other plasma metabolites. The ABC's primary objective is to establish a robust foundation for assessing cardiac aging, thereby furnishing a dependable reference for clinical applications and future research endeavors. This aims to contribute significantly to the enhancement of cardiovascular health and overall well-being among elderly individuals.
Collapse
Affiliation(s)
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yang Che
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Siqi Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200433, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200433, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai 200433, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hou-Zao Chen
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Junjie Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Chun-Mei Cao
- Laboratory of Cardiovascular Science, Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
- Capital Institute of Pediatrics, Beijing 100020, China
- Graduate School of Peking Union Medical College, Beijing 100730, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing 100191, China
| | - Yao Lu
- Clinical Research Center, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yingxin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing 100029, China
| | - Yan-Jiang Wang
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing 400042, China
- Institute of Brain and Intelligence, Third Military Medical University, Chongqing 400042, China
- Chongqing Key Laboratory of Ageing and Brain Diseases, Chongqing 400016, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi’an 710032, China
| | - Jing Qu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi-Da Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai 200070, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing 100730, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| |
Collapse
|
9
|
Zhang Y, Wang X, Li XK, Lv SJ, Wang HP, Liu Y, Zhou J, Gong H, Chen XF, Ren SC, Zhang H, Dai Y, Cai H, Yan B, Chen HZ, Tang X. Sirtuin 2 deficiency aggravates ageing-induced vascular remodelling in humans and mice. Eur Heart J 2023:ehad381. [PMID: 37377116 PMCID: PMC10393077 DOI: 10.1093/eurheartj/ehad381] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS The mechanisms underlying ageing-induced vascular remodelling remain unclear. This study investigates the role and underlying mechanisms of the cytoplasmic deacetylase sirtuin 2 (SIRT2) in ageing-induced vascular remodelling. METHODS AND RESULTS Transcriptome and quantitative real-time PCR data were used to analyse sirtuin expression. Young and old wild-type and Sirt2 knockout mice were used to explore vascular function and pathological remodelling. RNA-seq, histochemical staining, and biochemical assays were used to evaluate the effects of Sirt2 knockout on the vascular transcriptome and pathological remodelling and explore the underlying biochemical mechanisms. Among the sirtuins, SIRT2 had the highest levels in human and mouse aortas. Sirtuin 2 activity was reduced in aged aortas, and loss of SIRT2 accelerated vascular ageing. In old mice, SIRT2 deficiency aggravated ageing-induced arterial stiffness and constriction-relaxation dysfunction, accompanied by aortic remodelling (thickened vascular medial layers, breakage of elastin fibres, collagen deposition, and inflammation). Transcriptome and biochemical analyses revealed that the ageing-controlling protein p66Shc and metabolism of mitochondrial reactive oxygen species (mROS) contributed to SIRT2 function in vascular ageing. Sirtuin 2 repressed p66Shc activation and mROS production by deacetylating p66Shc at lysine 81. Elimination of reactive oxygen species by MnTBAP repressed the SIRT2 deficiency-mediated aggravation of vascular remodelling and dysfunction in angiotensin II-challenged and aged mice. The SIRT2 coexpression module in aortas was reduced with ageing across species and was a significant predictor of age-related aortic diseases in humans. CONCLUSION The deacetylase SIRT2 is a response to ageing that delays vascular ageing, and the cytoplasm-mitochondria axis (SIRT2-p66Shc-mROS) is important for vascular ageing. Therefore, SIRT2 may serve as a potential therapeutic target for vascular rejuvenation.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoman Wang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xun-Kai Li
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Shuang-Jie Lv
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - He-Ping Wang
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Yang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Division of Vascular Surgery, Department of General Surgery, and Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Jingyue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Hui Gong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| | - Xiao-Feng Chen
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Chengdu, Sichuan 611137, China
| | - Si-Chong Ren
- Department of Nephrology, First Affiliated Hospital of Chengdu Medical College, 783 Xindu Avenue, Chengdu, Sichuan 610500, China
| | - Huina Zhang
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, No. 2 Anzhen Road, Beijing 10029, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, 180 Fenglin Road, Shanghai 200032, China
| | - Hua Cai
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Bo Yan
- Institute of Precision Medicine, Jining Medical University, 133 Hehua Road, Taibaihu New District, Jining, Shandong 272067, China
| | - Hou-Zao Chen
- Department of Biochemistry & Molecular Biology, State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, 5 Dong Dan San Tiao, Beijing 100005, China
- Medical Epigenetics Research Center, Chinese Academy of Medical Sciences, 5 Dong Dan San Tiao, Beijing 100005, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- National Health Commission Key Laboratory of Chronobiology, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
- Development and Related Diseases of Women and Children, Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, No.17 People's South Road, Chengdu, Sichuan 610041, China
| |
Collapse
|