1
|
Tang X, Estau D, Huang X, Li Z. Edaravone targets PDGFRβ to attenuate VSMC phenotypic transition. Life Sci 2025; 370:123568. [PMID: 40113075 DOI: 10.1016/j.lfs.2025.123568] [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: 12/24/2024] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
AIMS PDGFRβ-driven phenotypic transition of vascular smooth muscle cells (VSMCs) is a pathological hallmark in various cardiovascular diseases, yet effective interventions are lacking. Here, we explored a promising drug targeting PDGFRβ against VSMC phenotypic transition. MATERIALS AND METHODS Connectivity map (CMAP) analysis was employed to identify the promising drug targeting PDGFRβ against VSMC phenotypic transition. A cell model stimulated with PDGF-BB and a mouse model of femoral artery injury were used to study the effects of edaravone (EDA) on VSMC phenotypic transition and PDGFRβ signaling. Molecular docking, drug affinity responsive target stability (DRATS) and cellular thermal shift assay (CETSA) were used to investigate whether EDA targeted PDGFRβ, which was further validated by a titration experiment. KEY FINDINGS Our study revealed that an approved drug EDA might target PDGFRβ against VSMC phenotypic transition. CMAP analysis unraveled EDA as a potential drug related to PDGFRβ. EDA markedly suppressed PDGFRβ-mediated VSMC transition from a contractile to a dedifferentiated phenotype, and reduced neointimal formation in wire-injured arteries. Mechanistically, molecular docking studies showed that EDA interacted with PDGFRβ, which was further confirmed by DRATS and CETSA. Consequently, EDA significantly suppressed PDGFRβ downstream signaling, including AKT and ERK1/2. Furthermore, EDA inhibited VSMC phenotypic transition in a PDGFRβ-dependent manner. SIGNIFICANCE Our work identifies EDA as a repurposed drug targeting PDGFRβ to attenuate VSMC phenotypic transition and provide new intervention measures for cardiovascular diseases associated with VSMC phenotypic transition.
Collapse
MESH Headings
- Animals
- Receptor, Platelet-Derived Growth Factor beta/metabolism
- Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Mice
- Edaravone/pharmacology
- Molecular Docking Simulation
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Male
- Mice, Inbred C57BL
- Phenotype
- Humans
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Dannya Estau
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China
| | - Xiaoru Huang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Beijing Key Laboratory of Cardiovascular Receptors Research; Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China; Department of Pharmaceutical Management and Clinical Pharmacy, College of Pharmacy, Peking University, Beijing, China.
| |
Collapse
|
2
|
Zhang L, Zhou J, Kong W. Extracellular matrix in vascular homeostasis and disease. Nat Rev Cardiol 2025; 22:333-353. [PMID: 39743560 DOI: 10.1038/s41569-024-01103-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 01/04/2025]
Abstract
The extracellular matrix is an essential component and constitutes a dynamic microenvironment of the vessel wall with an indispensable role in vascular homeostasis and disease. From early development through to ageing, the vascular extracellular matrix undergoes various biochemical and biomechanical alterations in response to diverse environmental cues and exerts precise regulatory control over vessel remodelling. Advances in novel technologies that enable the comprehensive evaluation of extracellular matrix components and cell-matrix interactions have led to the emergence of therapeutic strategies that specifically target this fine-tuned network. In this Review, we explore various aspects of extracellular matrix biology in vascular development, disorders and ageing, emphasizing the effect of the extracellular matrix on disease initiation and progression. Additionally, we provide an overview of the potential therapeutic implications of targeting the extracellular matrix microenvironment in vascular diseases.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| |
Collapse
|
3
|
Tu L, Zou Z, Yang Y, Wang S, Xing B, Feng J, Jin Y, Cheng M. Targeted drug delivery systems for atherosclerosis. J Nanobiotechnology 2025; 23:306. [PMID: 40269931 PMCID: PMC12016489 DOI: 10.1186/s12951-025-03384-0] [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: 01/19/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Atherosclerosis is a complex cardiovascular disease driven by multiple factors, including aging, inflammation, oxidative stress, and plaque rupture. The progression of this disease is often covert, emphasizing the need for early biomarkers and effective intervention measures. In recent years, advancements in therapeutic strategies have highlighted the potential of targeting specific processes in atherosclerosis, such as plaque localization, macrophage activity, and key enzymes. Based on this, this review discusses the potential role of targeted drugs in the treatment of atherosclerosis. It also focuses on their clinical efficacy in anti-atherosclerosis treatment and their ability to provide more precise therapeutic approaches. The findings underscore that future research can concentrate on exploring newer drug delivery systems and biomarkers to further refine clinical treatment strategies and enhance the long-term dynamic management of atherosclerosis.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Ye Yang
- Wenzhou Yining Geriatric Hospital, Wenzhou, 325041, P.R. China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China
- Guangxi University of Chinese Medicine, Nanning, 530200, P.R. China
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang, 330006, P.R. China.
| |
Collapse
|
4
|
Zaman S, Wasfy JH, Kapil V, Ziaeian B, Parsonage WA, Sriswasdi S, Chico TJA, Capodanno D, Colleran R, Sutton NR, Song L, Karam N, Sofat R, Fraccaro C, Chamié D, Alasnag M, Warisawa T, Gonzalo N, Jomaa W, Mehta SR, Cook EES, Sundström J, Nicholls SJ, Shaw LJ, Patel MR, Al-Lamee RK. The Lancet Commission on rethinking coronary artery disease: moving from ischaemia to atheroma. Lancet 2025; 405:1264-1312. [PMID: 40179933 DOI: 10.1016/s0140-6736(25)00055-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/01/2025] [Accepted: 01/09/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Sarah Zaman
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia
| | - Jason H Wasfy
- Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Vikas Kapil
- William Harvey Research Institute, Centre for Cardiovascular Medicine and Devices, NIHR Barts Biomedical Research Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Boback Ziaeian
- Division of Cardiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - William A Parsonage
- Australian Centre for Health Services Innovation, Queensland University of Technology, Brisbane, QLD, Australia; Department of Cardiology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Pathum Wan, Bangkok, Thailand; Faculty of Medicine, Chulalongkorn University, Pathum Wan, Bangkok, Thailand
| | - Timothy J A Chico
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK; British Heart Foundation Data Science Centre, Health Data Research UK, London, UK
| | - Davide Capodanno
- Division of Cardiology, Azienda Ospedaliero Universitaria Policlinico, University of Catania, Catania, Italy
| | - Róisín Colleran
- Department of Cardiology and Cardiovascular Research Institute, Mater Private Network, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland
| | - Nadia R Sutton
- Department of Internal Medicine, and Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lei Song
- Department of Cardiology, National Clinical Research Centre for Cardiovascular Diseases, Fuwai Hospital, Beijing, China; Peking Union Medical College (Chinese Academy of Medical Sciences), Beijing, China
| | - Nicole Karam
- Cardiology Department, European Hospital Georges Pompidou, Paris City University, Paris, France
| | - Reecha Sofat
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Chiara Fraccaro
- Division of Cardiology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
| | - Daniel Chamié
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mirvat Alasnag
- Cardiac Center, King Fahd Armed Forces Hospital, Jeddah, Saudi Arabia
| | | | - Nieves Gonzalo
- Cardiology Department, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Walid Jomaa
- Cardiology B Department, Fattouma Bourguiba University Hospital, University of Monastir, Monastir, Tunisia
| | - Shamir R Mehta
- Population Health Research Institute, Hamilton Health Sciences, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Elizabeth E S Cook
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Johan Sundström
- Uppsala University, Uppsala, Sweden; The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | | | - Leslee J Shaw
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manesh R Patel
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA; Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Rasha K Al-Lamee
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
5
|
Chen D, Wang X, Zhang S, Huang J, Li M, Wang L, Jiang T. The experimental study of the effect of fluid shear force on the migration rate of human umbilical vein endothelial cells. Biochem Biophys Res Commun 2025; 758:151619. [PMID: 40117976 DOI: 10.1016/j.bbrc.2025.151619] [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: 12/01/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND The vascular endothelium is a continuous monolayer of flattened cells that cover the surface of the lumen of blood vessels. Endothelial cell damage can readily result in thrombus formation and thickening of the intima. Accelerating the migration and repair of peripheral endothelial cells is essential. Shear force is an important hydrodynamic factor affecting endothelial cell function. We aimed to investigate the effect of different shear forces on the migration rate of endothelial cells. METHODS Human umbilical vein endothelial cells (HUVECs) were used instead of endothelial cells to establish a cell scratch model. Plate flow chambers were then used to intervene in HUVECs growth with different shear force magnitudes (4 dyn/cm2, 8 dyn/cm2, and 12 dyn/cm2). The healing rate of the scratches was observed under light microscopy, and finally the expression of RhoA and CDC42 was detected by molecular experiments. The expression of CDC42 factor was inhibited by siRNA interference, and the wound healing ability of HUVECs in the control group and the CDC42 inhibition group under different fluid shear forces was observed under light microscopy. RESULTS High shear forces promote the healing of scratches. In addition, relatively strong shear forces promoted the expression of cytokines RhoA and CDC42. Compared with untransfected HUVECs, HUVECs with inhibition of CDC42 expression by siRNA interference showed weak migration ability in different fluid shear groups. CONCLUSION Increasing fluid shear force in a range (4-12 dyn/cm2) contributes to endothelial cell migration. Inhibition of CDC42 expression weakened the migration ability of HUVECs under different fluid shear forces.
Collapse
Affiliation(s)
- Dong Chen
- Dalian University of Technology, China; Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China; China Medical University, Shenyang, China
| | - Xianwei Wang
- Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China.
| | - Sen Zhang
- Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China; Dalian Medical University, Dalian, China
| | - Jiaming Huang
- Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China
| | - Mei Li
- Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China
| | | | - Tao Jiang
- Department of Neurosurgery, Dalian University of Technology Affiliated Central Hospital, China; China Medical University, Shenyang, China; West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Wang R, Wu J, Lin Y, Xiao Y, Yang B, Yao S, Pan T, Fu Z, Li S, Wang C, Zhu Y. An epitope-directed mRNA vaccine inhibits tumor metastasis through the blockade of MICA/B α1/2 shedding. Cell Rep Med 2025; 6:101981. [PMID: 39999840 PMCID: PMC11970329 DOI: 10.1016/j.xcrm.2025.101981] [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: 05/04/2024] [Revised: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025]
Abstract
Antigenic peptide-based mRNA vaccines have been explored for immunotherapeutic use in various types of cancer because of their advantages in activating durable and specific immune responses. However, their role in modulating tumor metastasis is still unclear. Here, we identify a conserved linear epitope-based peptide, Ma3P, located in the proteolytic region of major histocompatibility complex (MHC) class I-related chain A (MICA) α3 and further design mCM10-L, an mRNA vaccine that encodes the carrier protein CRM197 and 10 tandem repeats of Ma3P. We demonstrate that vaccination with mCM10-L induces the production of specific antibodies that block MICA/B α1/2 shedding, activate CD8+ T cells and natural killer (NK) cells, and significantly inhibit MICA/B+ tumor metastasis in mice. Furthermore, mCM10-L stimulation triggers the production of specific antibodies to promote MICA/B-mediated immune killing in an in-vitro-interacting human organoid model and humanized mice. Our results indicate the potential clinical application prospects of the mCM10-L vaccine.
Collapse
Affiliation(s)
- Rui Wang
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jingni Wu
- Department of International Healthcare Center and General Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifeng Lin
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yufei Xiao
- Department of Clinical Laboratory, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bin Yang
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sheng Yao
- Department of Hepatobiliary and Pancreatic Surgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tianhui Pan
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhixuan Fu
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Shuyu Li
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Caihua Wang
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Yongliang Zhu
- Laboratory of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
7
|
Chen L, Tong X, Wu Y, Liu C, Tang C, Qi X, Kong F, Li M, Jin L, Zeng B. A dataset of single-cell transcriptomic atlas of Bama pig and potential marker genes across seven tissues. BMC Genom Data 2025; 26:16. [PMID: 40075302 PMCID: PMC11899051 DOI: 10.1186/s12863-025-01308-3] [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: 11/26/2024] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
The use of single-cell sequencing technology for single-cell transcriptomics studies in pigs is expanding progressively. However, the comprehensive classification of cell types across different anatomical tissues and organs of pig in multiple datasets remains relatively limited. This study employs single-cell and single-nucleus sequencing technologies in Bama pig to identify unique marker genes and their corresponding transcriptomic profiles across diverse cell types in various anatomical tissues and organs, including subcutaneous fat, visceral fat, psoas major muscle, liver, spleen, lung, and kidney. Through detailed data analyses, we observed widespread cellular diversity across various anatomical tissues and organs of Bama pig. This work contributes a comprehensive dataset that supports physiological studies and aids in the identification and prediction of potential marker genes through single-cell transcriptomics of these tissues. The methodologies and data employed in this study are designed to improve the accuracy of cell type identification and ensure consistent cell type allocation.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xingyan Tong
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yujie Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Can Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Chuang Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xu Qi
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Fanli Kong
- College of Life Science, Sichuan Agricultural University, Ya'an, 625099, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Long Jin
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Bo Zeng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
8
|
Mead TJ, Bhutada S, Peruzzi N, Adegboye J, Seifert DE, Cahill E, Drinko J, Donnellan E, Guggiliam A, Popovic Z, Griffin B, Tran-Lundmark K, Apte SS. ADAMTS7, a target in atherosclerosis, cooperates with its homolog ADAMTS12 to protect against myxomatous valve degeneration. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100288. [PMID: 40115634 PMCID: PMC11925103 DOI: 10.1016/j.jmccpl.2025.100288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
The physiological roles of the metalloprotease-proteoglycan ADAMTS7, a drug target in atherosclerosis and vascular restenosis, and its homolog ADAMTS12, are undefined in the cardiovascular system. The objective of the present work was to investigate their roles in mice with genetic inactivation of both proteases and in relation to the resulting valve defects, to define their proteolytic activities in the matrisome. Here, we demonstrate that Adamts7 and Adamts12 are co-expressed in heart valves and each buffers inactivation of the other by compensatory upregulation. Leaflets of Adamts7 -/-;Adamts12 -/- aortic valves, but not the respective single mutants, were abnormally shaped at birth, with progressively severe disorganization and enlargement occurring thereafter. Doppler echocardiography showed that Adamts7 -/-;Adamts12 -/- mice had stenotic and regurgitant aortic valves. We investigated ADAMTS7 and ADAMTS12 substrates relevant to the valve matrisome in secretome libraries from Adamts7 -/-;Adamts12 -/- cells using the N-terminomics technique Terminal Amine Isotopic Labeling of Substrates (TAILS). Although ADAMTS7 and ADAMTS12 shared several extracellular matrix (ECM) substrates, cleavage sites and sequence preference for each protease were distinct. Adamts7 -/-;Adamts12 -/- valve leaflets showed accumulation of several of the identified ECM substrates, including periostin, a matricellular protein crucial for cardiac valve homeostasis. We conclude that the myxomatous degeneration in Adamts7 -/-;Adamts12 -/- valve leaflets reflects a complex disturbance of ECM proteostasis with accumulation of multiple ADAMTS7 and ADAMTS12 ECM substrates, and perturbation of regulatory pathways with roots in ECM, such as TGFβ signaling, which was increased in the mutant valves.
Collapse
Affiliation(s)
- Timothy J Mead
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- University Hospitals Rainbow Babies and Children's Hospital, Cleveland, OH, USA
| | - Sumit Bhutada
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Niccolò Peruzzi
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Janet Adegboye
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Deborah E Seifert
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elisabeth Cahill
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Jeanne Drinko
- Department of Cardiovascular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Eoin Donnellan
- Department of Cardiovascular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Anu Guggiliam
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Zoran Popovic
- Department of Cardiovascular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Brian Griffin
- Department of Cardiovascular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Karin Tran-Lundmark
- Department of Experimental Medical Science and Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- The Pediatric Heart Center, Skane University Hospital, Lund, Sweden
| | - Suneel S Apte
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| |
Collapse
|
9
|
Wu J, Liu Y, Zong J, Qiu M, Zhou Y, Li Y, Aili T, Zhao X, Hu B. TTK Inhibition Alleviates Postinjury Neointimal Formation and Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409250. [PMID: 39716891 PMCID: PMC11809377 DOI: 10.1002/advs.202409250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 11/19/2024] [Indexed: 12/25/2024]
Abstract
Atherosclerosis and its associated cardio-cerebrovascular complications remain the leading causes of mortality worldwide. Current lipid-lowering therapies reduce only approximately one-third of the cardiovascular risk. Furthermore, vascular restenosis and thrombotic events following surgical interventions for severe vascular stenosis significantly contribute to treatment failure. This highlights the urgent need for novel therapeutic targets to manage atherosclerosis and prevent restenosis and thrombosis after vascular injury. This study identifies TTK protein kinase (TTK) as a key regulator of vascular smooth muscle cell (VSMC) phenotypic switching in the context of postinjury neointimal formation and atherosclerosis. Mechanistically, TTK upregulation in VSMCs phosphorylates p120-catenin, leading to β-catenin nuclear accumulation and dissociation of the myocardin (MYOCD)/serum response factor (SRF) complex. Deletion of TTK specifically in VSMCs reduces postinjury neointimal formation in vascular injury models and attenuates atherosclerotic lesions in ApoE-/- mice. Notably, oral administration of the TTK inhibitor CFI-402257 mitigated neointimal formation without impairing reendothelialization and reduced atherosclerotic lesions in ApoE-/- mice without altering lipid levels. These findings suggest that targeting TTK, through inhibitors or alternative strategies, represents a promising approach to simultaneously prevent postinjury restenosis and treat atherosclerosis.
Collapse
Affiliation(s)
- Jie‐Hong Wu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yu‐Xiao Liu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Jia‐Bin Zong
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Min Qiu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Yi‐Fan Zhou
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Ya‐Nan Li
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Tuersun Aili
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Xin‐Ran Zhao
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| | - Bo Hu
- Department of NeurologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
| |
Collapse
|
10
|
Jia B, Wei R, Yuan C, Cheng T, Shi S, Chu Y, Hu Y. A bibliometric analysis of vaccination against atherosclerosis. Hum Vaccin Immunother 2024; 20:2365500. [PMID: 39693182 DOI: 10.1080/21645515.2024.2365500] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 12/20/2024] Open
Abstract
A growing body of research indicates the promising potential of vaccines in both preventing and treating atherosclerosis (AS). To gain a comprehensive understanding of the current research landscape and emerging trends in this field, this study conducted a bibliometric analysis of publications on AS vaccines using the Web of Science Core Collection (WoSCC) database, based on the "bibliometric" R package and VOSviewer software. From 1991 to 2024, a total of 462 publications were identified in the WoSCC. The United States appeared as the leading contributor in terms of both total publications and citations. The Vaccine journal exhibited the highest publications output. Nilsson J from the Lund University in Sweden was the author with the most published articles, total citations and Hirsch index (H-index). Keywords analysis and thematic maps analysis revealed the passive immunotherapy (AS protective antibodies vaccines) was a hot mature theme, the active immunotherapy (AS antigens vaccines) was an emerging and booming theme, while the efficacy and safety of AS vaccines was a niche and well-developed theme. These findings offered valuable insights into the AS vaccination and provided guidance for future research in this domain.
Collapse
Affiliation(s)
- Bochao Jia
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Rui Wei
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Chenlu Yuan
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tao Cheng
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuai Shi
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuguang Chu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Department of Cardiovascular Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Tsioulos G, Vallianou NG, Skourtis A, Dalamaga M, Kotsi E, Kargioti S, Adamidis N, Karampela I, Mourouzis I, Kounatidis D. Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy? Biomolecules 2024; 14:1637. [PMID: 39766344 PMCID: PMC11727084 DOI: 10.3390/biom14121637] [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: 11/25/2024] [Revised: 12/15/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation.
Collapse
Affiliation(s)
- Georgios Tsioulos
- Fourth Department of Internal Medicine, Medical School, Attikon General University Hospital, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Natalia G. Vallianou
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Alexandros Skourtis
- Department of Internal Medicine, Evangelismos General Hospital, 10676 Athens, Greece;
| | - Maria Dalamaga
- Department of Biological Chemistry, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Evangelia Kotsi
- Second Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Hippokratio General Hospital, 11527 Athens, Greece;
| | - Sofia Kargioti
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Nikolaos Adamidis
- First Department of Internal Medicine, Sismanogleio General Hospital, 15126 Athens, Greece; (S.K.); (N.A.)
| | - Irene Karampela
- Second Department of Critical Care, Medical School, Attikon General University Hospital, University of Athens, 12461 Athens, Greece;
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitris Kounatidis
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 11527 Athens, Greece;
| |
Collapse
|
12
|
Vasilieva MI, Shatalova RO, Matveeva KS, Shindyapin VV, Minskaia E, Ivanov RA, Shevyrev DV. Senolytic Vaccines from the Central and Peripheral Tolerance Perspective. Vaccines (Basel) 2024; 12:1389. [PMID: 39772050 PMCID: PMC11680330 DOI: 10.3390/vaccines12121389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Preventive medicine has proven its long-term effectiveness and economic feasibility. Over the last century, vaccination has saved more lives than any other medical technology. At present, preventative measures against most infectious diseases are successfully used worldwide; in addition, vaccination platforms against oncological and even autoimmune diseases are being actively developed. At the same time, the development of medicine led to an increase in both life expectancy and the proportion of age-associated diseases, which pose a heavy socio-economic burden. In this context, the development of vaccine-based approaches for the prevention or treatment of age-related diseases opens up broad prospects for extending the period of active longevity and has high economic potential. It is well known that the development of age-related diseases is associated with the accumulation of senescent cells in various organs and tissues. It has been demonstrated that the elimination of such cells leads to the restoration of functions, rejuvenation, and extension of the lives of experimental animals. However, the development of vaccines against senescent cells is complicated by their antigenic heterogeneity and the lack of a unique marker. In addition, senescent cells are the body's own cells, which may be the reason for their low immunogenicity. This mini-review discusses the mechanisms of central and peripheral tolerance that may influence the formation of an anti-senescent immune response and be responsible for the accumulation of senescent cells with age.
Collapse
Affiliation(s)
- Mariia I. Vasilieva
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Rimma O. Shatalova
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Kseniia S. Matveeva
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia;
| | - Vadim V. Shindyapin
- Research Center for Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia;
| | - Ekaterina Minskaia
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Roman A. Ivanov
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| | - Daniil V. Shevyrev
- Research Center for Translational Medicine, Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Sirius 354349, Russia
| |
Collapse
|
13
|
Kitada K. Hypertension research 2024 update and perspectives: basic research. Hypertens Res 2024; 47:3304-3309. [PMID: 39251854 DOI: 10.1038/s41440-024-01878-2] [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: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
This review article introduces some basic studies that were recently published in this journal, as a part of Hypertension Research 2024 Update and Perspectives. Including recent basic research trends in other scientific journals, we would like to summarize basic research on keywords such as hypertension and its associated organ damage, new treatments, and others. It is expected that the accumulation of basic studies will lead to breakthroughs in hypertension treatment in the future and lead to the definitive treatment of hypertension beyond blood pressure control with anti-hypertensive drugs.
Collapse
Affiliation(s)
- Kento Kitada
- Department of Pharmacology, Faculty of Medicine, Kagawa University, Kagawa, 7610793, Japan.
| |
Collapse
|
14
|
Cheng Y, Zhang X, Wu X, Ding Y, Zhu L, Pan J, Zhao Y, Zhou M. Multifunctional Microneedle Patches for Perivascular Gene Delivery and Treatment of Vascular Intimal Hyperplasia. ACS NANO 2024; 18:32578-32588. [PMID: 39545708 DOI: 10.1021/acsnano.4c09527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Gene therapy has emerged as a promising approach to address challenging cardiovascular diseases. Extensive efforts have been focused on developing highly efficient gene vectors with precise delivery techniques to enhance its effectiveness. In this study, we present multifunctional dopamine-gelatin microneedle patches with gene therapy capabilities to achieve perivascular gene delivery for intimal hyperplasia treatment. These patches that were fabricated through freeze-drying of gelatin are with recombinant adeno-associated virus (rAAVs)-carrying tips and dopamine coating backing layers. The lyophilized gelatin could not only effectively preserve the therapeutic activity of rAAVs but could also demonstrate the capability to penetrate the adventitia for efficient delivery. The incorporation of dopamine facilitated patch adhesion and extended the release duration. Based on these advantages, we have demonstrated that the rAAVs-loaded microneedle patches (AMNPs) behave satisfactorily in perivascular gene delivery to inhibit carotid artery restenosis in rats. These features indicate that the AMNPs are clinically valuable in the treatment of vascular intimal hyperplasia diseases.
Collapse
Affiliation(s)
- Yi Cheng
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xiaoxuan Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiangyi Wu
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Youjun Ding
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Linxi Zhu
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jun Pan
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuanjin Zhao
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Min Zhou
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
15
|
Aherrahrou R, Reinberger T, Hashmi S, Erdmann J. GWAS breakthroughs: mapping the journey from one locus to 393 significant coronary artery disease associations. Cardiovasc Res 2024; 120:1508-1530. [PMID: 39073758 DOI: 10.1093/cvr/cvae161] [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: 01/27/2024] [Revised: 03/20/2024] [Accepted: 06/12/2024] [Indexed: 07/30/2024] Open
Abstract
Coronary artery disease (CAD) poses a substantial threat to global health, leading to significant morbidity and mortality worldwide. It has a significant genetic component that has been studied through genome-wide association studies (GWAS) over the past 17 years. These studies have made progress with larger sample sizes, diverse ancestral backgrounds, and the discovery of multiple genomic regions related to CAD risk. In this review, we provide a comprehensive overview of CAD GWAS, including information about the genetic makeup of the disease and the importance of ethnic diversity in these studies. We also discuss challenges of identifying causal genes and variants within GWAS loci with a focus on non-coding regions. Additionally, we highlight tissues and cell types relevant to CAD, and discuss clinical implications of GWAS findings including polygenic risk scores, sex-specific differences in CAD genetics, ethnical aspects of personalized interventions, and GWAS guided drug development.
Collapse
Affiliation(s)
- Rédouane Aherrahrou
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Tobias Reinberger
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Satwat Hashmi
- Department of Biological and Biomedical Sciences, Aga Khan University, Stadium Road, 74800 Karachi, Pakistan
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Marie-Curie-Str. Haus 67/BMF, 23562 Lübeck, Germany
- DZHK (German Centre for Cardiovascular Research), Institute for Cardiogenetics, Universität zu Lübeck, Partner Site Hamburg/Kiel/Lübeck, Germany
- University Heart Centre Lübeck, University Hospital Schleswig-Holstein, Ratzeburger Allee 160, 23562 Lübeck, Germany
| |
Collapse
|
16
|
Chung A, Chang HK, Pan H, Bashore AC, Shuck K, Matias CV, Gomez J, Yan H, Li M, Bauer RC. ADAMTS7 Promotes Smooth Muscle Foam Cell Expansion in Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582156. [PMID: 38463994 PMCID: PMC10925101 DOI: 10.1101/2024.02.26.582156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Human genetic studies have repeatedly associated ADAMTS7 with atherosclerotic cardiovascular disease. Subsequent investigations in mice demonstrated that ADAMTS7 is proatherogenic and induced in response to vascular injury and that the proatherogenicity of ADAMTS7, a secreted protein, is due to its catalytic activity. However, the cell-specific mechanisms governing ADAMTS7 proatherogenicity remain unclear. To determine which vascular cell types express ADAMTS7, we interrogated single-cell RNA sequencing of human carotid atherosclerosis and found ADAMTS7 expression in smooth muscle cells (SMCs), endothelial cells (ECs), and fibroblasts. We subsequently created SMC- and EC-specific Adamts7 conditional knockout and transgenic mice. Conditional knockout of Adamts7 in either cell type is insufficient to reduce atherosclerosis, whereas transgenic induction in either cell type increases atherosclerosis. In SMC transgenic mice, this increase coincides with an expansion of lipid-laden SMC foam cells and decreased fibrous cap formation. RNA-sequencing in SMCs revealed an upregulation of lipid uptake genes typically assigned to macrophages. Subsequent experiments demonstrated that ADAMTS7 increases SMC oxLDL uptake through increased CD36 levels. Furthermore, Cd36 expression is increased due to increased levels of PU.1, a transcription factor typically associated with myeloid fate determination. In summary, Adamts7 expression in either SMCs or ECs promotes SMC foam cell formation and atherosclerosis. In SMCs, ADAMTS7 promotes oxLDL uptake via increased PU.1 and Cd36 expression, thereby increasing SMC foam cell formation and atherosclerosis.
Collapse
|
17
|
Brennan PG, Mota L, Aridi T, Patel N, Liang P, Ferran C. Advancements in Omics and Breakthrough Gene Therapies: A Glimpse into the Future of Peripheral Artery Disease. Ann Vasc Surg 2024; 107:229-246. [PMID: 38582204 DOI: 10.1016/j.avsg.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 04/08/2024]
Abstract
Peripheral artery disease (PAD), a highly prevalent global disease, associates with significant morbidity and mortality in affected patients. Despite progress in endovascular and open revascularization techniques for advanced PAD, these interventions grapple with elevated rates of arterial restenosis and vein graft failure attributed to intimal hyperplasia (IH). Novel multiomics technologies, coupled with sophisticated analyses tools recently powered by advances in artificial intelligence, have enabled the study of atherosclerosis and IH with unprecedented single-cell and spatial precision. Numerous studies have pinpointed gene hubs regulating pivotal atherogenic and atheroprotective signaling pathways as potential therapeutic candidates. Leveraging advancements in viral and nonviral gene therapy (GT) platforms, gene editing technologies, and cutting-edge biomaterial reservoirs for delivery uniquely positions us to develop safe, efficient, and targeted GTs for PAD-related diseases. Gene therapies appear particularly fitting for ex vivo genetic engineering of IH-resistant vein grafts. This manuscript highlights currently available state-of-the-art multiomics approaches, explores promising GT-based candidates, and details GT delivery modalities employed by our laboratory and others to thwart mid-term vein graft failure caused by IH, as well as other PAD-related conditions. The potential clinical translation of these targeted GTs holds the promise to revolutionize PAD treatment, thereby enhancing patients' quality of life and life expectancy.
Collapse
Affiliation(s)
- Phillip G Brennan
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Lucas Mota
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Tarek Aridi
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Nyah Patel
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Patric Liang
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, and Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Division of Nephrology and the Transplant Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
| |
Collapse
|
18
|
Tang D, Liu Y, Duan R, Lin R, Li Z, Liu X, Huang J, Zhao M. COL6A6 Peptide Vaccine Alleviates Atherosclerosis through Inducing Immune Response and Regulating Lipid Metabolism in Apoe-/- Mice. Cells 2024; 13:1589. [PMID: 39329770 PMCID: PMC11429512 DOI: 10.3390/cells13181589] [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: 07/11/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Atherosclerosis is an autoimmune disease characterized by lipid imbalances and chronic inflammation within blood vessels, with limited preventive and treatment options currently available. In this study, a vaccine prepared with COL6A6 peptide (named the Pep_A6 vaccine) was administered to immunize Apoe-/- mice, and the immune mechanism of the Pep_A6 vaccine against atherosclerosis was first investigated. The results of arterial oil red O staining demonstrated that the Pep_A6 vaccine significantly reduced the atherosclerotic plaque area in Apoe-/- mice fed with a high-fat diet for 20 weeks. A flow cytometry analysis revealed that the Pep_A6 vaccine inhibited Th1 cell differentiation and increased the proportion of Treg cells. Furthermore, there was a significant increase in Ly6Clow monocytes observed in the vaccinated group. The ELISA results showed that the Pep_A6 vaccine induced a significant expression of Pep_A6-specific antibody IgG and IgG1 in mouse serum. Additionally, we found that the Pep_A6 vaccine significantly decreased serum LDL-C content and regulated the expression of genes related to liver lipid metabolism. Together, our findings suggest that the Pep_A6 vaccine alleviates atherosclerosis by inducing a positive immune response and regulating lipid metabolism, providing new insights into potential prevention strategies for atherosclerosis as an innovative vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ming Zhao
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (D.T.)
| |
Collapse
|
19
|
Wu Y, Chen Z, Zheng Z, Li X, Shu J, Mao R, An J, Fan S, Luo R, Guo Y, Xu W, Liang M, Huang K, Wang C. Tudor-SN exacerbates pathological vascular remodeling by promoting the polyubiquitination of PTEN via NEDD4-1. J Biomed Sci 2024; 31:88. [PMID: 39237902 PMCID: PMC11378411 DOI: 10.1186/s12929-024-01076-9] [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: 05/09/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Dysregulation of vascular homeostasis can induce cardiovascular diseases and increase global mortality rates. Although lineage tracing studies have confirmed the pivotal role of modulated vascular smooth muscle cells (VSMCs) in the progression of pathological vascular remodeling, the underlying mechanisms are still unclear. METHODS The expression of Tudor-SN was determined in VSMCs of artery stenosis, PDGF-BB-treated VSMCs and atherosclerotic plaque. Loss- and gain-of-function approaches were used to explore the role of Tudor-SN in the modulation of VSMCs phenotype both in vivo and in vitro. RESULTS In this study, we demonstrate that Tudor-SN expression is significantly elevated in injury-induced arteries, atherosclerotic plaques, and PDGF-BB-stimulated VSMCs. Tudor-SN deficiency attenuates, but overexpression aggravates the synthetic phenotypic switching of VSMCs and pathological vascular remodeling. Loss of Tudor-SN also reduces atherosclerotic plaque formation and increases plaque stability. Mechanistically, PTEN, the major regulator of the MAPK and PI3K-AKT signaling pathways, plays a vital role in Tudor-SN-mediated regulation on proliferation and migration of VSMCs. Tudor-SN facilitates the polyubiquitination and degradation of PTEN via NEDD4-1, thus exacerbating vascular remodeling under pathological conditions. BpV (HOpic), a specific inhibitor of PTEN, not only counteracts the protective effect of Tudor-SN deficiency on proliferation and migration of VSMCs, but also abrogates the negative effect of carotid artery injury-induced vascular remodeling in mice. CONCLUSIONS Our findings reveal that Tudor-SN deficiency significantly ameliorated pathological vascular remodeling by reducing NEDD4-1-dependent PTEN polyubiquitination, suggesting that Tudor-SN may be a novel target for preventing vascular diseases.
Collapse
Affiliation(s)
- Yichen Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Zilong Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhe Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruiqi Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jie An
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Siyuan Fan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruijie Luo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yi Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Wenjing Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| | - Cheng Wang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| |
Collapse
|
20
|
Zhang Y, Zheng BY, Zhang QF, Zhao YN, Yu QM, Liu X, Ding SY, Qian SS, Wu H, Wu QY, Zhang YH, Zheng L, Zhang XH, Zhang HF, Hao YM, Lu JC, Wang L, Wen JK, Zheng B. Nanoparticles targeting OPN loaded with BY1 inhibits vascular restenosis by inducing FTH1-dependent ferroptosis in vascular smooth muscle cells. Biomaterials 2024; 309:122613. [PMID: 38759485 DOI: 10.1016/j.biomaterials.2024.122613] [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: 10/25/2023] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
Vascular restenosis following angioplasty continues to pose a significant challenge. The heterocyclic trioxirane compound [1, 3, 5-tris((oxiran-2-yl)methyl)-1, 3, 5-triazinane-2, 4, 6-trione (TGIC)], known for its anticancer activity, was utilized as the parent ring to conjugate with a non-steroidal anti-inflammatory drug, resulting in the creation of the spliced conjugated compound BY1. We found that BY1 induced ferroptosis in VSMCs as well as in neointima hyperplasia. Furthermore, ferroptosis inducers amplified BY1-induced cell death, while inhibitors mitigated it, indicating the contribution of ferroptosis to BY1-induced cell death. Additionally, we established that ferritin heavy chain1 (FTH1) played a pivotal role in BY1-induced ferroptosis, as evidenced by the fact that FTH1 overexpression abrogated BY1-induced ferroptosis, while FTH1 knockdown exacerbated it. Further study found that BY1 induced ferroptosis by enhancing the NCOA4-FTH1 interaction and increasing the amount of intracellular ferrous. We compared the effectiveness of various administration routes for BY1, including BY1-coated balloons, hydrogel-based BY1 delivery, and nanoparticles targeting OPN loaded with BY1 (TOP@MPDA@BY1) for targeting proliferated VSMCs, for prevention and treatment of the restenosis. Our results indicated that TOP@MPDA@BY1 was the most effective among the three administration routes, positioning BY1 as a highly promising candidate for the development of drug-eluting stents or treatments for restenosis.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Bo-Yang Zheng
- Department of tumor biotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Qian-Fan Zhang
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ya-Nan Zhao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qi-Ming Yu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin Liu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Si-Ying Ding
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuang-Shuang Qian
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Han Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Qian-Yu Wu
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yu-Han Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Lei Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China; Institution of Chinese Integrative Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China
| | - Hao-Feng Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China
| | - Yi-Ming Hao
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jing-Chao Lu
- Department of Cardiovascular Medicine, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lei Wang
- Department of Medicinal Chemistry, School of Pharmacy, Hebei Medical University, Hebei Province Key Laboratory of Innovative Drug Research and Evaluation, Shijiazhuang, 050017, China.
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
21
|
Zhang L, Feng Q, Kong W. ECM Microenvironment in Vascular Homeostasis: New Targets for Atherosclerosis. Physiology (Bethesda) 2024; 39:0. [PMID: 38984789 DOI: 10.1152/physiol.00028.2023] [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: 11/20/2023] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 07/11/2024] Open
Abstract
Alterations in vascular extracellular matrix (ECM) components, interactions, and mechanical properties influence both the formation and stability of atherosclerotic plaques. This review discusses the contribution of the ECM microenvironment in vascular homeostasis and remodeling in atherosclerosis, highlighting Cartilage oligomeric matrix protein (COMP) and its degrading enzyme ADAMTS7 as examples, and proposes potential avenues for future research aimed at identifying novel therapeutic targets for atherosclerosis based on the ECM microenvironment.
Collapse
Affiliation(s)
- Lu Zhang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qianqian Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
22
|
Liu HH, Wei W, Wu FF, Cao L, Yang BJ, Fu JN, Li JX, Liang XY, Dong HY, Heng YY, Zhang PF. Sodium tanshinone IIA sulfonate protects vascular relaxation in ApoE-knockout mice by inhibiting the SYK-NLRP3 inflammasome-MMP2/9 pathway. BMC Cardiovasc Disord 2024; 24:354. [PMID: 38992615 PMCID: PMC11241843 DOI: 10.1186/s12872-024-03990-0] [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: 01/22/2024] [Accepted: 06/19/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Hyperlipidemia damages vascular wall and serves as a foundation for diseases such as atherosclerosis, hypertension and stiffness. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome is implicated in vascular dysfunction associated with hyperlipidemia-induced vascular injury. Sodium tanshinone IIA sulfonate (STS), a well-established cardiovascular protective drug with recognized anti-inflammatory, antioxidant, and vasodilatory properties, is yet to be thoroughly investigated for its impact on vascular relaxant imbalance induced by hyperlipidemia. METHODS In this study, we treated ApoE-knockout (ApoE-/-) mouse with STS and assessed the activation of the NLRP3 inflammasome, expression of MMP2/9, integrity of elastic fibers, and vascular constriction and relaxation. RESULTS Our findings reveal that STS intervention effectively preserves elastic fibers, significantly restores aortic relaxation function in ApoE-/- mice, and reduces their excessive constriction. Furthermore, STS inhibits the phosphorylation of spleen tyrosine kinase (SYK), suppresses NLRP3 inflammasome activation, and reduces MMP2/9 expression. CONCLUSIONS These results demonstrate that STS protects vascular relaxation against hyperlipidemia-induced damage through modulation of the SYK-NLRP3 inflammasome-MMP2/9 pathway. This research provides novel insights into the mechanisms underlying vascular relaxation impairment in a hyperlipidemic environment and uncovers a unique mechanism by which STS preserves vascular relaxation, offering valuable foundational research evidence for its clinical application in promoting vascular health.
Collapse
Affiliation(s)
- Hai-Hua Liu
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China
| | - Wei Wei
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China.
- Department of Pharmacology, Changzhi Medical College, No.161, Jiefang East Street, Changzhi, 046000, Shanxi, China.
- Department of Clinical Center Laboratory, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China.
| | - Fei-Fei Wu
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China
| | - Lu Cao
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China
| | - Bing-Jie Yang
- Department of Stomatology, Changzhi Medical College, No.161, Jiefang East Street, Changzhi, 046000, Shanxi, China
| | - Jia-Ning Fu
- Department of Stomatology, Changzhi Medical College, No.161, Jiefang East Street, Changzhi, 046000, Shanxi, China
| | - Jing-Xia Li
- Department of Anesthesia, Changzhi Medical College, No.161, Jiefang East Street, Changzhi, 046000, Shanxi, China
| | - Xin-Yue Liang
- Department of Medical Imageology, Changzhi Medical College, No.161, Jiefang East Street, Changzhi, 046000, Shanxi, China
| | - Hao-Yu Dong
- Department of Endocrinology, Heping Hospital Affiliated to Changzhi Medical College, No.110, Yan'an South Road, Changzhi, 046000, Shanxi, China
| | - Yan-Yan Heng
- Department of Nephrology Heping Hospital, Changzhi Medical College, No.110, Yanan Road South, Changzhi, 046000, Shanxi, China
| | - Peng-Fei Zhang
- Department of Nephrology Heping Hospital, Changzhi Medical College, No.110, Yanan Road South, Changzhi, 046000, Shanxi, China
| |
Collapse
|
23
|
Khan A, Ley K. Immunotherapy for atherosclerosis by targeting pro-inflammatory T cells. Cell Res 2024; 34:467-468. [PMID: 38565655 PMCID: PMC11217497 DOI: 10.1038/s41422-024-00955-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Affiliation(s)
- Amir Khan
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - Klaus Ley
- Immunology Center of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
24
|
Heng Y, Wei W, Cheng L, Wu F, Dong H, Li J, Fu J, Yang B, Liang X, Liu C, Li H, Liu H, Zhang P. FGF21 overexpression alleviates VSMC senescence in diabetic mice by modulating the SYK-NLRP3 inflammasome-PPARγ-catalase pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:892-904. [PMID: 38733164 PMCID: PMC11214975 DOI: 10.3724/abbs.2024032] [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: 10/29/2023] [Accepted: 01/16/2024] [Indexed: 05/13/2024] Open
Abstract
Diabetes accelerates vascular senescence, which is the basis for atherosclerosis and stiffness. The activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome and oxidative stress are closely associated with progressive senescence in vascular smooth muscle cells (VSMCs). The vascular protective effect of FGF21 has gradually gained increasing attention, but its role in diabetes-induced vascular senescence needs further investigation. In this study, diabetic mice and primary VSMCs are transfected with an FGF21 activation plasmid and treated with a peroxisome proliferator-activated receptor γ (PPARγ) agonist (rosiglitazone), an NLRP3 inhibitor (MCC950), and a spleen tyrosine kinase (SYK)-specific inhibitor, R406, to detect senescence-associated markers. We find that FGF21 overexpression significantly restores the level of catalase (CAT), vascular relaxation, inhibits the intensity of ROSgreen fluorescence and p21 immunofluorescence, and reduces the area of SA-β-gal staining and collagen deposition in the aortas of diabetic mice. FGF21 overexpression restores CAT, inhibits the expression of p21, and limits the area of SA-β-gal staining in VSMCs under high glucose conditions. Mechanistically, FGF21 inhibits SYK phosphorylation, the production of the NLRP3 dimer, the expression of NLRP3, and the colocalization of NLRP3 with PYCARD (ASC), as well as NLRP3 with caspase-1, to reverse the cleavage of PPARγ, preserve CAT levels, suppress ROSgreen density, and reduce the expression of p21 in VSMCs under high glucose conditions. Our results suggest that FGF21 alleviates vascular senescence by regulating the SYK-NLRP3 inflammasome-PPARγ-catalase pathway in diabetic mice.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Syk Kinase/metabolism
- Syk Kinase/genetics
- PPAR gamma/metabolism
- PPAR gamma/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Inflammasomes/metabolism
- Mice
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Cellular Senescence
- Male
- Signal Transduction
- Mice, Inbred C57BL
- Fibroblast Growth Factors/metabolism
- Fibroblast Growth Factors/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
Collapse
Affiliation(s)
- Yanyan Heng
- Department of NephrologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Wei Wei
- Department of PharmacologyChangzhi Medical CollegeChangzhi046000China
- Department of EndocrinologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
- Department of Clinical Central LaboratoryHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Linzhong Cheng
- Department of National Institute for Clinical Trials of Drugs and Phase I Clinical Trial LaboratoryHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Feifei Wu
- Department of EndocrinologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Haoyu Dong
- Department of EndocrinologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Jingxia Li
- Department of AnesthesiaChangzhi Medical CollegeChangzhi046000China
| | - Jianing Fu
- Department of StomatologyChangzhi Medical CollegeChangzhi046000China
| | - Bingjie Yang
- Department of StomatologyChangzhi Medical CollegeChangzhi046000China
| | - Xinyue Liang
- Department of Medical ImageologyChangzhi Medical CollegeChangzhi046000China
| | - Chunyan Liu
- Department of AnesthesiaChangzhi Medical CollegeChangzhi046000China
| | - Haiju Li
- Department of PharmacologyChangzhi Medical CollegeChangzhi046000China
- Department of National Institute for Clinical Trials of Drugs and Phase I Clinical Trial LaboratoryHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Haihua Liu
- Department of EndocrinologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| | - Pengfei Zhang
- Department of NephrologyHeping Hospital Affiliated to Changzhi Medical CollegeChangzhi046000China
| |
Collapse
|
25
|
Xu Y, Ge L, Rui Y, Wang Y, Wang Z, Yang J, Shi Y, Dong Z, Zhang Y, Zhang R, Yang T, Lv L, Xiang F, Chen S, Song A, Li T, Liu M, Guo Y. Suramin inhibits phenotypic transformation of vascular smooth muscle cells and neointima hyperplasia by suppressing transforming growth factor beta receptor 1 /Smad2/3 pathway activation. Eur J Pharmacol 2024; 968:176422. [PMID: 38365108 DOI: 10.1016/j.ejphar.2024.176422] [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: 09/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Vascular smooth muscle cells (VSMCs) contribute to neointimal hyperplasia (NIH) after vascular injury, a common feature of vascular remodelling disorders. Suramin is known to exert antitumour effects by inhibiting the proliferation of various tumour cells; however, its effects and mechanism on VSMCs remain unclear. This study investigated the effects of suramin on human aortic smooth muscle cells (HASMCs), rat aortic smooth muscle cells (RASMCs) and NIH to examine its suitability for the prevention of vascular remodelling disorders. In vitro, suramin administration reduced platelet-derived growth factor type BB (PDGF-BB)-stimulated proliferation, migration, and dedifferentiation of VSMCs through a transforming growth factor beta receptor 1 (TGFBR1)/Smad2/3-dependent pathway. Suramin dramatically inhibited NIH ligation in the left common carotid artery (LCCA) vivo. Therefore, our results indicate that suramin protects against the development of pathological vascular remodelling by attenuating VSMCs proliferation, migration, and phenotypic transformation and may be used as a potential medicine for the treatment of NIH.
Collapse
Affiliation(s)
- Yingjie Xu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Luning Ge
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yanan Rui
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuchen Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zhonghua Wang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiemei Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yuanqi Shi
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Zengxiang Dong
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, China
| | - Yu Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Rongzhen Zhang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tao Yang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Lin Lv
- Experimental Animal Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fei Xiang
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Sixuan Chen
- Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Aoliang Song
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tiankai Li
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Mingyu Liu
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Yuanyuan Guo
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China; Department of Geriatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
26
|
Wu R, Sun F, Zhang W, Ren J, Liu GH. Targeting aging and age-related diseases with vaccines. NATURE AGING 2024; 4:464-482. [PMID: 38622408 DOI: 10.1038/s43587-024-00597-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 02/20/2024] [Indexed: 04/17/2024]
Abstract
Aging is a major risk factor for numerous chronic diseases. Vaccination offers a promising strategy to combat these age-related diseases by targeting specific antigens and inducing immune responses. Here, we provide a comprehensive overview of recent advances in vaccine-based interventions targeting these diseases, including Alzheimer's disease, type II diabetes, hypertension, abdominal aortic aneurysm, atherosclerosis, osteoarthritis, fibrosis and cancer, summarizing current approaches for identifying disease-associated antigens and inducing immune responses against these targets. Further, we reflect on the recent development of vaccines targeting senescent cells, as a strategy for more broadly targeting underlying causes of aging and associated pathologies. In addition to highlighting recent progress in these areas, we discuss important next steps to advance the therapeutic potential of these vaccines, including improving and robustly demonstrating efficacy in human clinical trials, as well as rigorously evaluating the safety and long-term effects of these vaccine strategies.
Collapse
Affiliation(s)
- Ruochen Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fei Sun
- Department of Cell Biology, Duke University Medical Center, Durham, NC, USA
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China.
- Sino-Danish College, School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jie Ren
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China.
- Sino-Danish College, School of Future Technology, University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Key Laboratory of RNA Science and Engineering, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [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] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
28
|
Meibom D, Wasnaire P, Beyer K, Broehl A, Cancho-Grande Y, Elowe N, Henninger K, Johannes S, Jungmann N, Krainz T, Lindner N, Maassen S, MacDonald B, Menshykau D, Mittendorf J, Sanchez G, Schaefer M, Stefan E, Torge A, Xing Y, Zubov D. BAY-9835: Discovery of the First Orally Bioavailable ADAMTS7 Inhibitor. J Med Chem 2024; 67:2907-2940. [PMID: 38348661 PMCID: PMC10895658 DOI: 10.1021/acs.jmedchem.3c02036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/23/2024]
Abstract
The matrix metalloprotease ADAMTS7 has been identified by multiple genome-wide association studies as being involved in the development of coronary artery disease. Subsequent research revealed the proteolytic function of the enzyme to be relevant for atherogenesis and restenosis after vessel injury. Based on a publicly known dual ADAMTS4/ADAMTS5 inhibitor, we have in silico designed an ADAMTS7 inhibitor of the catalytic domain, which served as a starting point for an optimization campaign. Initially our inhibitors suffered from low selectivity vs MMP12. An X-ray cocrystal structure inspired us to exploit amino acid differences in the binding site of MMP12 and ADAMTS7 to improve selectivity. Further optimization composed of employing 5-membered heteroaromatic groups as hydantoin substituents to become more potent on ADAMTS7. Finally, fine-tuning of DMPK properties yielded BAY-9835, the first orally bioavailable ADAMTS7 inhibitor. Further optimization to improve selectivity vs ADAMTS12 seems possible, and a respective starting point could be identified.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eric Stefan
- Broad
Institute, 02142 Cambridge, United States
| | | | - Yi Xing
- Broad
Institute, 02142 Cambridge, United States
| | | |
Collapse
|
29
|
Bai X, Zhang W, Yu T. Integrative bioinformatics analysis identifies APOB as a critical biomarker in coronary in-stent restenosis. Biomark Med 2023; 17:983-998. [PMID: 38223945 DOI: 10.2217/bmm-2023-0507] [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] [Indexed: 01/16/2024] Open
Abstract
Aim: Coronary artery disease (CAD) is a major contributor to the worldwide prevalence of cardiovascular disease. In-stent restenosis (ISR) is a common complication which can lead to stent implantation failure, necessitating repeated intervention and presenting a significant obstacle for CAD management. Methods: To accurately assess and determine the hub genes associated with ISR, CAD databases from the Gene Expression Omnibus were utilized and weighted gene coexpression network analysis was employed to identify key genes in blood samples. Results: APOB was identified as a risk gene for ISR occurrence. Subsequent correlation analysis of APOB demonstrated a positive association with ISR. Clinical validation further confirmed the predictive value of APOB in ISR detection. Conclusion: We have identified APOB as a critical predictive biomarker for ISR in CAD patients.
Collapse
Affiliation(s)
- Xinghua Bai
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| | - Weizong Zhang
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| | - Tao Yu
- Department of Cardiovascular Medicine, The First People's Hospital of Linping District, Hangzhou, 311100, PR China
| |
Collapse
|
30
|
Raposo-Gutiérrez I, Rodríguez-Ronchel A, Ramiro AR. Atherosclerosis antigens as targets for immunotherapy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1129-1147. [PMID: 39196152 DOI: 10.1038/s44161-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/18/2023] [Indexed: 08/29/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arteries that can lead to thrombosis, infarction and stroke, underlying the first cause of mortality worldwide. Adaptive immunity plays critical roles in atherosclerosis, and numerous studies have ascribed both atheroprotective and atherogenic functions to specific subsets of T and B cells. However, less is known on how antigen specificity determines the protective or adverse outcome of such adaptive responses. Understanding antigen triggers in atherosclerosis is crucial to delve deeper into mechanisms of disease initiation and progression and to implement specific immunotherapeutic approaches, including vaccination strategies. Here we review the role of adaptive immunity in atherosclerosis and the insights that single-cell technology has provided into the function of distinct immune cell subsets. We outline the most relevant atherosclerosis antigens and antibodies reported to date and examine their immunotherapeutic potential. Finally, we review the most promising vaccination-based clinical trials targeting the adaptive immune system.
Collapse
Affiliation(s)
- Irene Raposo-Gutiérrez
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Ana Rodríguez-Ronchel
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain
| | - Almudena R Ramiro
- B Lymphocyte Lab, Novel Mechanisms of Atherosclerosis Program, Spanish National Center for Cardiovascular Research, Madrid, Spain.
| |
Collapse
|
31
|
Li Z, Qiao O, Wang Y, Li N, Gong Y. Potential therapeutic targets for trauma management. Trends Pharmacol Sci 2023; 44:S0165-6147(23)00234-1. [PMID: 39492319 DOI: 10.1016/j.tips.2023.10.001] [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: 08/20/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 11/05/2024]
Abstract
Despite advances in medical treatments for severe trauma, it remains a critical condition associated with high mortality. During trauma, the release of endogenous damage-associated molecular patterns (DAMPs) can induce immune dysfunction, leading to sepsis or multiple organ dysfunction syndrome (MODS). Vaccines based on specific pathogen antigens and pathogen-associated molecular patterns (PAMPs) contribute largely to the prevention of communicable diseases through the induction of adaptive immune responses. Vaccines developed based on autologous molecules may also promote recovery from non-communicable diseases (NCDs) by eliciting appropriate immune responses, as recent clinical trials indicate. Developing new vaccines targeting DAMPs may be an effective pre-protective measure for trauma management. We describe the role of DAMPs in post-traumatic immune dysfunction and discuss the potential of harnessing them for trauma vaccine development as well as the risks and challenges.
Collapse
Affiliation(s)
- Zizheng Li
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China; Thinking Biomed (Beijing) Co., Ltd, Beijing Economic and Technological Development Zone, Beijing, 100176, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| |
Collapse
|
32
|
Wang L, Liu T, Zheng Y, Zhou J, Hua H, Kong L, Huang W, Peng X, Wen T. P4HA2-induced prolyl hydroxylation of YAP1 restricts vascular smooth muscle cell proliferation and neointima formation. Life Sci 2023; 330:122002. [PMID: 37549826 DOI: 10.1016/j.lfs.2023.122002] [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: 05/12/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation and neointima formation play significant roles in atherosclerosis development and restenosis following percutaneous coronary intervention. Our team previously discovered that TEA domain transcription factor 1 (TEAD1) promotes vascular smooth muscle differentiation, which is necessary for vascular development. Conversely, aberrant YAP1 activation upregulates the platelet-derived growth factor receptor beta to encourage VSMC proliferation and neointima formation. In this study, we aimed to investigate the molecular mechanisms of YAP1/TEAD signaling during neointima formation. Our research focused on the prolyl 4-hydroxylase alpha 2 (P4HA2) and its downstream target, Yes-associated protein 1 (YAP1), in regulating VSMC differentiation and neointima formation. Our results indicated that P4HA2 reduction leads to VSMC dedifferentiation and promotes neointima formation after injury. Furthermore, we found that P4HA2-induced prolyl hydroxylation of YAP1 restricts its transcriptional activity, which is essential to maintaining VSMC differentiation. These findings suggest that targeting P4HA2-mediated prolyl hydroxylation of YAP1 may be a promising therapeutic approach to prevent injury-induced neointima formation in cardiovascular disease.
Collapse
Affiliation(s)
- Liang Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Ting Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yaofu Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Jiamin Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Hexiang Hua
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Liming Kong
- Department of Outpatient clinic, The First Affiliated Hospital of Nanchang, University, Nanchang, Jiangxi 330006, China
| | - Weilin Huang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China
| | - Tong Wen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Hypertension Research Institute of Jiangxi Province, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
33
|
Sharifi MA, Wierer M, Dang TA, Milic J, Moggio A, Sachs N, von Scheidt M, Hinterdobler J, Müller P, Werner J, Stiller B, Aherrahrou Z, Erdmann J, Zaliani A, Graettinger M, Reinshagen J, Gul S, Gribbon P, Maegdefessel L, Bernhagen J, Sager HB, Mann M, Schunkert H, Kessler T. ADAMTS-7 Modulates Atherosclerotic Plaque Formation by Degradation of TIMP-1. Circ Res 2023; 133:674-686. [PMID: 37675562 PMCID: PMC7615141 DOI: 10.1161/circresaha.123.322737] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/23/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND The ADAMTS7 locus was genome-wide significantly associated with coronary artery disease. Lack of the ECM (extracellular matrix) protease ADAMTS-7 (A disintegrin and metalloproteinase-7) was shown to reduce atherosclerotic plaque formation. Here, we sought to identify molecular mechanisms and downstream targets of ADAMTS-7 mediating the risk of atherosclerosis. METHODS Targets of ADAMTS-7 were identified by high-resolution mass spectrometry of atherosclerotic plaques from Apoe-/- and Apoe-/-Adamts7-/- mice. ECM proteins were identified using solubility profiling. Putative targets were validated using immunofluorescence, in vitro degradation assays, coimmunoprecipitation, and Förster resonance energy transfer-based protein-protein interaction assays. ADAMTS7 expression was measured in fibrous caps of human carotid artery plaques. RESULTS In humans, ADAMTS7 expression was higher in caps of unstable as compared to stable carotid plaques. Compared to Apoe-/- mice, atherosclerotic aortas of Apoe-/- mice lacking Adamts-7 (Apoe-/-Adamts7-/-) contained higher protein levels of Timp-1 (tissue inhibitor of metalloprotease-1). In coimmunoprecipitation experiments, the catalytic domain of ADAMTS-7 bound to TIMP-1, which was degraded in the presence of ADAMTS-7 in vitro. ADAMTS-7 reduced the inhibitory capacity of TIMP-1 at its canonical target MMP-9 (matrix metalloprotease-9). As a downstream mechanism, we investigated collagen content in plaques of Apoe-/- and Apoe-/-Adamts7-/- mice after a Western diet. Picrosirius red staining of the aortic root revealed less collagen as a readout of higher MMP-9 activity in Apoe-/- as compared to Apoe-/- Adamts7-/- mice. To facilitate high-throughput screening for ADAMTS-7 inhibitors with the aim of decreasing TIMP-1 degradation, we designed a Förster resonance energy transfer-based assay targeting the ADAMTS-7 catalytic site. CONCLUSIONS ADAMTS-7, which is induced in unstable atherosclerotic plaques, decreases TIMP-1 stability reducing its inhibitory effect on MMP-9, which is known to promote collagen degradation and is likewise associated with coronary artery disease. Disrupting the interaction of ADAMTS-7 and TIMP-1 might be a strategy to increase collagen content and plaque stability for the reduction of atherosclerosis-related events.
Collapse
Affiliation(s)
- M. Amin Sharifi
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Tan An Dang
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Jelena Milic
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Aldo Moggio
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Nadja Sachs
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Moritz von Scheidt
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Philipp Müller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Julia Werner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Barbara Stiller
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics and University Heart Centre Lübeck, University of Lübeck, Lübeck, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Mira Graettinger
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Jeanette Reinshagen
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Sheraz Gul
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Hamburg, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Hamburg, Germany
| | - Lars Maegdefessel
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Vascular Biology and Experimental Vascular Medicine Unit, Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Jürgen Bernhagen
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
- Division of Vascular Biology, Institute for Stroke and Dementia Research, Ludwig Maximilian University of Munich, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
34
|
Xu R, Li T, Li Z, Kong W, Wang T, Zhang X, Luo J, Li W, Jiao L. Knowledge fields and emerging trends about extracellular matrix in carotid artery disease from 1990 to 2021: analysis of the scientific literature. Eur J Med Res 2023; 28:284. [PMID: 37587506 PMCID: PMC10428572 DOI: 10.1186/s40001-023-01259-4] [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: 12/02/2022] [Accepted: 08/01/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Stroke is a heavy burden in modern society, and carotid artery disease is a major cause. The role of the extracellular matrix (ECM) in the development and progression of carotid artery disease has become a popular research focus. However, there is no published bibliometric analysis to derive the main publication features and trends in this scientific area. We aim to conduct a bibliometric analysis to reveal current status of ECM in carotid artery disease and to predict future hot spots. METHODS We searched and downloaded articles from the Web of Science Core Collection with "Carotid" and "Extracellular Matrix" as subject words from 1990 to 2021. The complete bibliographic data were analyzed by Bibliometrics, BICOMB, gCLUTO and CiteSpace softwares. RESULTS Since 1990, the United States has been the leader in the number of publications in the field of ECM in carotid artery disease, followed by China, Japan and Germany. Among institutions, Institut National De La Sante Et De La Recherche Medicale Inserm, University of Washington Seattle and Harvard University are in the top 3. "Arteriosclerosis Thrombosis and Vascular Biology" is the most popular journal and "Circulation" is the most cited journal. "Clowes AW", "Hedin Ulf" and "Nilsson Jan" are the top three authors of published articles. Finally, we investigated the frontiers through the strongest citation bursts, conducted keyword biclustering analysis, and discovered five clusters of research hotspots. Our research provided a comprehensive analysis of the fundamental data, knowledge organization, and dynamic evolution of research about ECM in carotid artery disease. CONCLUSIONS The field of ECM in carotid artery disease has received increasing attention. We summarized the history of the field and predicted five future hotspots through bibliometric analysis. This study provided a reference for researchers in this fields, and the methodology can be extended to other fields.
Collapse
Affiliation(s)
- Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Tianhua Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
35
|
Chung A, Reilly MP, Bauer RC. ADAMTS7: a Novel Therapeutic Target in Atherosclerosis. Curr Atheroscler Rep 2023; 25:447-455. [PMID: 37354304 PMCID: PMC11457552 DOI: 10.1007/s11883-023-01115-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2023] [Indexed: 06/26/2023]
Abstract
PURPOSE OF REVIEW Genome-wide association studies have repeatedly linked the metalloproteinase ADAMTS7 to coronary artery disease. Here we aim to highlight recent findings surrounding the human genetics of ADAMTS7, novel mouse models that investigate ADAMTS7 function, and potential substrates of ADAMTS7 cleavage. RECENT FINDINGS Recent genome-wide association studies in coronary artery disease have replicated the GWAS signal for ADAMTS7 and shown that the signal holds true even across different ethnic groups. However, the direction of effect in humans remains unclear. A recent novel mouse model revealed that the proatherogenicity of ADAMTS7 is derived from its catalytic functions, while at the translational level, vaccinating mice against ADAMTS7 reduced atherosclerosis. Finally, in vitro proteomics approaches have identified extracellular matrix proteins as candidate substrates that may be causal for the proatherogenicity of ADAMTS7. ADAMTS7 represents an enticing target for therapeutic intervention. The recent studies highlighted here have replicated prior findings, confirming the genetic link between ADAMTS7 and atherosclerosis, while providing further evidence in mice that ADAMTS7 is a targetable proatherogenic enzyme.
Collapse
Affiliation(s)
- Allen Chung
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Muredach P Reilly
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
- Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | - Robert C Bauer
- Cardiometabolic Genomics Program, Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA.
| |
Collapse
|
36
|
Affiliation(s)
- Thorsten Kessler
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, 80636 Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, 80636 Munich, Germany
| | - Heribert Schunkert
- German Heart Centre Munich, Department of Cardiology, Technical University of Munich, 80636 Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), partner site Munich Heart Alliance, 80636 Munich, Germany
| |
Collapse
|
37
|
Petrovič D, Nussdorfer P, Petrovič D. The rs3825807 Polymorphism of ADAMTS7 as a Potential Genetic Marker for Myocardial Infarction in Slovenian Subjects with Type 2 Diabetes Mellitus. Genes (Basel) 2023; 14:508. [PMID: 36833435 PMCID: PMC9957282 DOI: 10.3390/genes14020508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/01/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND A disintegrin and metalloprotease with thrombospondin motif 7 (ADAMTS-7) was reported to play a role in the migration of vascular smooth muscle cells and neointimal formation. The object of the study was to investigate the association between the rs3825807 polymorphism of ADAMTS7 and myocardial infarction among patients with type 2 diabetes mellitus in a Slovenian cohort. METHODS 1590 Slovenian patients with type 2 diabetes mellitus were enrolled in this retrospective cross-sectional case-control study. In total, 463 had a history of recent myocardial infarction, and 1127 of the subjects in the control group had no clinical signs of coronary artery disease. Genetic analysis of an rs3825807 polymorphism of ADAMTS7 was performed with logistic regression. RESULTS Patients with the AA genotype had a higher prevalence of myocardial infarction than those in the control group in recessive [odds ratio (OR) 1.647; confidence interval (CI) 1.120-2.407; p = 0.011] and co-dominant (OR 2.153; CI 1.215-3.968; p = 0.011) genetic models. CONCLUSION We found a statistically significant association between rs3825807 and myocardial infarction in a cohort of Slovenian patients with type 2 diabetes mellitus. We report that the AA genotype might be a genetic risk factor for myocardial infarction.
Collapse
Affiliation(s)
- David Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Petra Nussdorfer
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| |
Collapse
|