1
|
Li L, Cheng M, Jin J, Zhao Y, Bai W, Zhang D, Zhang S, Bai Y, Xu J. The m6A reader YTHDF2 protects vascular smooth muscle cells against the osteogenic differentiation through targeting Runx2. Ren Fail 2025; 47:2488876. [PMID: 40230077 PMCID: PMC12001846 DOI: 10.1080/0886022x.2025.2488876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 03/12/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Vascular calcification (VC) is an important pathological development progress in chronic kidney disease (CKD) and may increase mortality but lacks effective treatments. N6-methyladenosine (m6A) has been verified to be the most prevalent internal chemical RNA modification in mammalian mRNAs. The M6A-modified mRNA degradation process is mediated by the reader YTHDF2 in an m6A-dependent manner. Nevertheless, the exact role and molecular mechanism of YTHDF2 in VC remain unclear. This study aimed to investigate the potential role of YTHDF2 in the osteogenic differentiation of vascular smooth muscle cells (VSMCs). It was found that YTHDF2 was markedly downregulated in both in vivo and in vitro calcified models. Functionally, YTHDF2 plays a protective role in VC. The overexpression of YTHDF2 inhibited the transdifferentiation of VSMCs from a contractile to an osteogenic phenotype, thus decreasing the expression of mineralization regulatory proteins and calcium deposition. Conversely, YTHDF2 deficiency aggravated this process. At the mechanistic level, YTHDF2 suppressed osteogenic transdifferentiation of VSMCs by regulating the Runt-related transcription factor 2 (Runx2). RNA immunoprecipitation-qPCR (RIP-qPCR) confirmed the binding of YTHDF2 to Runx2, and luciferase reporter assays confirmed the presence of the m6A site in Runx2. In addition, an actinomycin D assay showed that the half-life of Runx2 mRNA was dramatically shortened in VSMCs overexpressing YTHDF2. These results suggest that YTHDF2 directly binds to the m6A modification site of Runx2 to mediate the mRNA degradation that prevents VC by inhibiting the osteogenic development of VSMCs. Therefore, YTHDF2 can be considered a potential therapeutic target for managing VC.
Collapse
Affiliation(s)
- Lanmei Li
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Jingjing Jin
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Yunfeng Zhao
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Weiwei Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Dongxue Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| |
Collapse
|
2
|
Cai Z, Xu S, Xiao X, Liu C, Zu L. Mib2 Regulates Lipid Metabolism in Heart Failure With Preserved Ejection Fraction via the Runx2-Hmgcs2 Axis. J Cell Mol Med 2025; 29:e70514. [PMID: 40159625 PMCID: PMC11955417 DOI: 10.1111/jcmm.70514] [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: 11/11/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
Obesity and the mismanagement of lipids significantly contribute to the development of heart failure with preserved ejection fraction (HFpEF). However, the underlying molecular mechanisms that regulate the metabolic changes and disruptions in lipid balance within HFpEF remain to be fully understood. Transcriptome data for HFpEF were sourced from the National Center for Biotechnology Information (NCBI) database. A mouse model for HFpEF was developed utilising leptin-deficient (ob/ob) mice. The cardiac-specific mind bomb E3 ubiquitin protein ligase 2 (Mib2) overexpression in ob/ob mice was achieved by tail vein injection of a recombinant adeno-associated virus serotype 9 vector carrying Mib2 with a cTNT promoter (AAV9-cTNT-Mib2). In vitro, neonatal rat ventricular myocytes were exposed to fatty acid to induce lipotoxicity. The molecular mechanisms were investigated through proteomic analysis, dual luciferase reporter gene assay, and immunoprecipitation assays. GO and KEGG enrichment analyses indicated that the differentially expressed proteins (DEPs) in HFpEF were prominently enriched in pathways related to the fatty acid metabolic process. The transcriptomic and proteomic analyses of heart tissues from HFpEF mice presented a notable elevation in the expression of 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2). Immunoprecipitation assays revealed that mind bomb 2 (Mib2) directly interacted with runt-related transcription factor 2 (Runx2), ubiquitinating and degrading Runx2 to inhibit Hmgcs2 transcription, impeding the fatty acid metabolic process. Mice with cardiac-specific overexpression of Mib2 displayed a more pronounced progression of cardiac dysfunction and an accumulation of lipids compared to the control group. Our research uncovers a mechanism by which Mib2 modulates cardiac lipid metabolic homeostasis in HFpEF, implicating the Runx2-Hmgcs2 axis.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology and Institute of Vascular MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijingChina
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesPeking UniversityBeijingChina
- Beijing Key Laboratory of Cardiovascular Receptors ResearchBeijingChina
| | - Shunyao Xu
- Department of Critical Care MedicineShenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and TechnologyShenzhenChina
| | - Xiaohua Xiao
- Department of GeriatricsThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Chen Liu
- Department of GeriatricsThe First Affiliated Hospital of Shenzhen University, Shenzhen Second People's HospitalShenzhenChina
| | - Lingyun Zu
- Department of Cardiology and Institute of Vascular MedicinePeking University Third HospitalBeijingChina
- State Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijingChina
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesPeking UniversityBeijingChina
- Beijing Key Laboratory of Cardiovascular Receptors ResearchBeijingChina
| |
Collapse
|
3
|
Cuevas RA, Hortells L, Chu CC, Wong R, Crane A, Boufford C, Regan C, Moorhead WJ, Bashline MJ, Parwal A, Parise AM, Behzadi P, Brown MJ, Gurkar A, Bruemmer D, Sembrat J, Sultan I, Gleason TG, Billaud M, St. Hilaire C. Non-Canonical TERT Activity Initiates Osteogenesis in Calcific Aortic Valve Disease. Circ Res 2025; 136:403-421. [PMID: 39835393 PMCID: PMC11825275 DOI: 10.1161/circresaha.122.321889] [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: 08/23/2022] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Calcific aortic valve disease is the pathological remodeling of valve leaflets. The initial steps in valve leaflet osteogenic reprogramming are not fully understood. As TERT (telomerase reverse transcriptase) overexpression primes mesenchymal stem cells to differentiate into osteoblasts, we investigated whether TERT contributes to the osteogenic reprogramming of valve interstitial cells. METHODS Human control and calcific aortic valve disease aortic valve leaflets and patient-specific human aortic valve interstitial cells were used in in vivo and in vitro calcification assays. Loss of function experiments in human aortic valve interstitial cells and cells isolated from Tert-/- and Terc-/- mice were used for mechanistic studies. Calcification was assessed in Tert+/+ and Tert-/- mice ex vivo and in vivo. In silico modeling, proximity ligation, and coimmunoprecipitation assays defined novel TERT interacting partners. Chromatin immunoprecipitation and cleavage under targets and tagmentation sequencing defined protein-DNA interactions. RESULTS TERT protein was highly expressed in calcified valve leaflets without changes in telomere length, DNA damage, or senescence markers, and these features were retained in isolated primary human aortic valve interstitial cells. TERT expression increased with osteogenic or inflammatory stimuli, and knockdown or genetic deletion of TERT prevented calcification in vitro and in vivo. Mechanistically, TERT was upregulated via NF-κB (nuclear factor-kappa B) and required to initiate osteogenic reprogramming, independent of its canonical reverse transcriptase activity and the long noncoding RNA TERC. TERT exerts non-canonical osteogenic functions via binding with STAT5 (signal transducer and activator of transcription 5). Depletion or inhibition of STAT5 prevented calcification. STAT5 was found to bind the promoter region of RUNX2 (runt-related transcription factor 2), the master regulator of osteogenic reprogramming. Finally, we demonstrate that TERT and STAT5 are upregulated and colocalized in calcific aortic valve disease tissue compared with control tissue. CONCLUSIONS TERT's non-canonical activity is required to initiate calcification. TERT is upregulated via inflammatory signaling pathways and partners with STAT5 to bind the RUNX2 gene promoter. These data identify a novel mechanism and potential therapeutic target to decrease vascular calcification.
Collapse
Affiliation(s)
- Rolando A. Cuevas
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Luis Hortells
- Cardiovascular Research Group, Department of Medical Biology, Faculty of Health Science, UiT-The Arctic University of Norway, 9019 Tromsø, Norway
| | - Claire C. Chu
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ryan Wong
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alex Crane
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Camille Boufford
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cailyn Regan
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - William J. Moorhead
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J. Bashline
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aneesha Parwal
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Angelina M. Parise
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Parya Behzadi
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark J. Brown
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aditi Gurkar
- Aging Institute, Division of Geriatrics, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dennis Bruemmer
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ibrahim Sultan
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Thomas G. Gleason
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Marie Billaud
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Cynthia St. Hilaire
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Zeng Z, Zhao Z, Yuan Q, Yang S, Wang Z, Wang Z, Zeng S, Li A, Chen Q, Zhu G, Xiao X, Luo G, Luo H, Li J, Zu X, Xie H, Liu J. Hepatic Steatosis Aggravates Vascular Calcification via Extracellular Vesicle-Mediated Osteochondrogenic Switch of Vascular Smooth Muscle Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408660. [PMID: 39680681 PMCID: PMC11791995 DOI: 10.1002/advs.202408660] [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: 07/26/2024] [Revised: 10/17/2024] [Indexed: 12/18/2024]
Abstract
The global incidence of metabolic dysfunction-associated fatty liver disease (MAFLD) has risen sharply. This condition is strongly associated with the risk of cardiovascular disease (CVD), but how MAFLD affects the development and progression of CVD, particularly concerning vascular calcification, remains unclear. Herein, extracellular vesicles (EVs) are identified from steatotic hepatocytes as a trigger that accelerated the progression of both vascular intimal and medial calcification. Steatotic hepatocytes are found to release more EVs, which are able to reach the vascular tissue, be taken up by vascular smooth muscle cells (VSMCs), and promote their osteogenic differentiation. Within these toxic vesicles, a protein cargo is identified called lectin galactoside-binding soluble 3 binding protein (Lgals3bp) that acted as a potent inducer of osteochondrogenic transformation in VSMCs. Both the inhibition of EV release and the liver-specific knockdown of Lgals3bp profoundly attenuated vascular calcification. This work partially explains the reason for the high incidence of vascular calcification in MAFLD and unveils a novel mechanism that may be used to prevent or treat cardiovascular complications in patients with MAFLD.
Collapse
Affiliation(s)
- Zhao‐Lin Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Department of Cardiovascular MedicineThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400016P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhi‐Bo Zhao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qing Yuan
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Qi Yang
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Zhen‐Xing Wang
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Zuo Wang
- Institute of Cardiovascular DiseaseKey Lab for Arteriosclerology of Hunan ProvinceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Shi‐Yu Zeng
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - An‐Qi Li
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Qian Chen
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guo‐Qiang Zhu
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Xin‐Hua Xiao
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Guang‐Hua Luo
- Department of RadiologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hai‐Yan Luo
- Department of GastroenterologyThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Jiao‐Yang Li
- Department of Occupational and Environmental HealthSchool of Public HealthWuhan UniversityWuhan430071P. R. China
| | - Xu‐Yu Zu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| | - Hui Xie
- Department of OrthopedicsMovement System Injury and Repair Research CenterNational Clinical Research Center for Geriatric DisordersHunan Key Laboratory of AngmedicineXiangya HospitalCentral South UniversityChangshaHunan410008P. R. China
| | - Jiang‐Hua Liu
- Department of Metabolism and EndocrinologyThe First Affiliated Hospital, Hengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
- Diabetes Clinical Medical Research Center of Hunan ProvincialHengyangHunan421001P. R. China
- Department of Clinical Laboratory MedicineThe First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001P. R. China
| |
Collapse
|
5
|
Li Z, Wu H, Yao F, Li Y, Li Y, Xie SA, Yu F, Fu Y, Wang L, Zhou J, Kong W. Runx2-NLRP3 axis orchestrates matrix stiffness-evoked vascular smooth muscle cell inflammation. Am J Physiol Cell Physiol 2025; 328:C467-C482. [PMID: 39761974 DOI: 10.1152/ajpcell.00448.2024] [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: 07/02/2024] [Revised: 12/22/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
Arterial stiffening is a hallmark of chronic kidney disease (CKD)-related cardiovascular events and is primarily attributed to the elevated matrix stiffness. Stiffened arteries are accompanied by low-grade inflammation, but the causal effects of matrix stiffness on inflammation remain unknown. For analysis of the relationship between arterial stiffness and vascular inflammation, pulse-wave velocity (PWV) and aortic inflammatory markers were analyzed in an adenine-induced mouse model of CKD in chronological order. Compared with their control littermates, mice with CKD showed elevated arterial stiffness at the early stage of disease progression, which preceded the onset of vascular inflammation. Correspondingly, the increase of matrix stiffness induced vascular smooth muscle cells (VSMCs) to transdifferentiate into an inflammatory phenotype, as indicated by the increased expression and secretion of MCP-1, IL-6, IL-1β, and IL-18. RNA-sequencing analysis of stiff matrix-cultured VSMCs and bioinformatics analysis with the ChIP-Atlas database revealed the potential involvement of the transcription factor Runx2. The expression and the nuclear localization of Runx2 were significantly increased in stiff matrix-cultured VSMCs. High-throughput ChIP-sequencing and promoter luciferase assays further revealed that NLRP3 was directly transcriptionally regulated by Runx2. The inhibition of Runx2 or NLRP3 inflammasome abrogated the proinflammatory effect of matrix stiffening on VSMCs. Together, these data revealed that arterial stiffness precedes vascular inflammatory responses in CKD mice and that the Runx2-NLRP3 axis orchestrates matrix stiffness and the VSMC inflammatory phenotype, which may contribute to the pathogenic role in arterial stiffness-related vascular inflammation and CKD-related cardiovascular complications.NEW & NOTEWORTHY As a hallmark of chronic kidney disease (CKD), arterial stiffening is related to increased vascular inflammation and cardiovascular morbidity, whereas the underlying mechanism is unclear. The study demonstrates that increased arterial stiffness precedes the onset of vascular inflammation, and matrix stiffness stimulates the transdifferentiation of vascular smooth muscle cells (VSMCs) to an inflammatory phenotype via activating Runx2-NLRP3 signaling, which provides novel insights into CKD-related cardiovascular disorder treatment.
Collapse
MESH Headings
- Animals
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Vascular Stiffness/physiology
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Inflammation/metabolism
- Inflammation/pathology
- Male
- Mice, Inbred C57BL
- Signal Transduction
- Cells, Cultured
- Disease Models, Animal
- Aorta/metabolism
- Aorta/pathology
Collapse
Affiliation(s)
- Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Fang Yao
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
| | - Yiran Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanjie Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Wang
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Pluripotent Stem Cells in Cardiac Repair and Regeneration, Chinese Academy of Medical Sciences, Beijing, 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
|
6
|
Hengtrakul N, Furrow E, Borofsky M, Toth F, Lulich JP. Expression of osteogenic proteins in kidneys of cats with nephrocalcinosis. J Vet Intern Med 2025; 39:e17278. [PMID: 39757788 PMCID: PMC11702495 DOI: 10.1111/jvim.17278] [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: 05/26/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Nephrocalcinosis is a common pathological finding in cats with chronic kidney disease and nephrolithiasis. Understanding its pathogenesis may identify future therapeutic targets. HYPOTHESIS Nephrocalcinosis is associated with expression of an osteogenic phenotype. ANIMALS Kidneys with medullary mineralization were obtained from 18 cats (10 with and 8 without nephroliths) undergoing necropsy. METHODS Cross-sectional study. Microradiography and histopathology (modified von Kossa stain) were used to confirm parenchymal mineralization. Immunohistochemistry for 5 osteogenic markers was performed to determine their co-localization with nephrocalcinosis. The proportion of kidneys with stronger immunointensity in mineralized versus non-mineralized regions was analyzed using 1-tailed sign tests. The proportion of kidneys with co-localization of nephrocalcinosis and each marker was compared between kidneys with and without nephroliths using Fisher's exact tests. RESULTS Nephrocalcinosis co-localized with osteopontin immunoreactivity in all 18 cats (100%) and with osteocalcin in 12 cats (67%). Both osteogenic markers had stronger immunointensity in mineralized regions compared with non-mineralized regions. Limited co-localization was observed with other markers: bone morphogenic protein-2 in 2 kidneys (both with nephroliths) and tissue non-specific alkaline phosphatase in 1 kidney (without nephroliths); runt-related transcription factor-2 was undetected. No statistically significant differences were found in the co-localization of nephrocalcinosis with osteogenic proteins between kidneys with and without nephroliths. CONCLUSIONS AND CLINICAL IMPORTANCE Expression of osteogenic proteins in areas of nephrocalcinosis indicates that nephrocalcinosis is associated with the development of an osteogenic phenotype. Targeting these processes could offer a novel approach to prevent nephrolithiasis at its origin.
Collapse
Affiliation(s)
- Nuttha Hengtrakul
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Eva Furrow
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Michael Borofsky
- Department of Urology, Medical SchoolUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Ferenc Toth
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| | - Jody P. Lulich
- Department of Veterinary Clinical Sciences, College of Veterinary MedicineUniversity of MinnesotaSt PaulMinnesotaUSA
| |
Collapse
|
7
|
Xu L, Liu B, Ma H, Qi E, Ma J, Chang T, Zhang J, Zhang W, Chen W, Cao X, Xiong X. O-GlcNAc transferase promotes vascular smooth muscle calcification through modulating Wnt/β-catenin signaling. FASEB J 2024; 38:e70271. [PMID: 39704274 DOI: 10.1096/fj.202401649rr] [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: 07/17/2024] [Revised: 11/21/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Vascular calcification (VC), associated with high cardiovascular mortality in patients with chronic kidney disease (CKD), involves osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). O-GlcNAcylation, a dynamic post-translational modification, is closely linked to cardiovascular diseases, including VC. However, the exact role and molecular mechanism of O-GlcNAc signaling in abnormal mineral metabolism-induced VC remain unclear. In the current study, we found that the levels of O-GlcNAc transferase (OGT) and global protein O-GlcNAcylation were significantly upregulated in the artery tissues of mouse calcification models and CKD patients with VC. To further delineate the in vivo role of OGT in VC, we generated Ogt smooth muscle cell-specific knockout mice and challenged them with 5/6 nephrectomy (5/6 Nx) or high-dose vitamin D3 to induce VC. Deletion of Ogt in VSMCs led to alleviated VC in response to 5/6 Nx or VD3. Moreover, elevated O-GlcNAcylation, induced by Thiamet-G, facilitated osteogenic transdifferentiation in VSMCs in response to phosphate, whereas OSMI-1, which reduces O-GlcNAcylation, exhibited an opposite phenotypic effect. Mechanistically, O-GlcNAc signaling enhanced the osteogenic conversion of VSMCs through regulation of canonical Wnt/β-catenin pathway. Indeed, β-catenin was O-GlcNAcylated by OGT and further increased its transcriptional activity in VSMCs. Furthermore, pharmacological activation of Wnt/β-catenin signaling largely reversed the diminished aortic calcification caused by Ogt ablation. Our findings demonstrate that smooth muscle O-GlcNAc signaling plays an important role in regulating hyperphosphatemia-induced VC and reveal that O-GlcNAcylation of β-catenin protein modulates its content and activity in VSMCs.
Collapse
MESH Headings
- Animals
- N-Acetylglucosaminyltransferases/metabolism
- N-Acetylglucosaminyltransferases/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Wnt Signaling Pathway
- Mice
- Mice, Knockout
- Humans
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Male
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- beta Catenin/metabolism
- Cells, Cultured
- Osteogenesis
Collapse
Affiliation(s)
- Lin Xu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Boao Liu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Honghui Ma
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Enbo Qi
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jie Ma
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Tingmin Chang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Jinghong Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Wencheng Zhang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Weiqian Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xuan Cao
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, Zhejiang, China
| | - Xiwen Xiong
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
8
|
Hemmati F, Akinpelu A, Nweze DC, Mistriotis P. 3D confinement alters smooth muscle cell responses to chemical and mechanical cues. APL Bioeng 2024; 8:046103. [PMID: 39464377 PMCID: PMC11512639 DOI: 10.1063/5.0225569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Smooth muscle cell (SMC) phenotypic switching is a hallmark of many vascular diseases. Although prior work has established that chemical and mechanical cues contribute to SMC phenotypic switching, the impact of three-dimensional (3D) confinement on this process remains elusive. Yet, in vivo, arterial SMCs reside within confined environments. In this study, we designed a microfluidic assay to investigate the interplay between 3D confinement and different environmental stimuli in SMC function. Our results show that tightly, but not moderately, confined SMCs acquire a contractile phenotype when exposed to collagen I. Elevated compressive forces induced by hydrostatic pressure abolish this upregulation of the contractile phenotype and compromise SMC survival, particularly in tightly confined spaces. Transforming growth factor beta 1, which promotes the contractile state in moderate confinement, fails to enhance the contractility of tightly confined cells. Fibronectin and engagement of cadherin 2 suppress the contractile phenotype of SMCs regardless of the degree of confinement. In contrast, homophilic engagement of cadherin 11 upregulates SMC-specific genes and enhances contractility in both moderately and tightly confined cells. Overall, our work introduces 3D confinement as a regulator of SMC phenotypic responses to chemical and mechanical signals.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Daniel Chinedu Nweze
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | | |
Collapse
|
9
|
Lin A, Miano JM, Fisher EA, Misra A. Chronic inflammation and vascular cell plasticity in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1408-1423. [PMID: 39653823 DOI: 10.1038/s44161-024-00569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cells, endothelial cells and macrophages undergo phenotypic conversions throughout atherosclerosis progression, both as a consequence of chronic inflammation and as subsequent drivers of it. The inflammatory hypothesis of atherosclerosis has been catapulted to the forefront of cardiovascular research as clinical trials have shown that anti-inflammatory therapy reduces adverse cardiovascular events. However, no current therapies have been specifically designed to target the phenotype of plaque cells. Fate mapping has revealed that plaque cells convert to detrimental and beneficial cell phenotypes during atherosclerosis, with cumulative evidence highlighting that vascular cell plasticity is intimately linked with plaque inflammation, ultimately impacting lesion stability. Here we review vascular cell plasticity during atherosclerosis in the context of the chronic inflammatory plaque microenvironment. We highlight the need to better understand how plaque cells behave during therapeutic intervention. We then propose modulating plaque cell phenotype as an unexplored therapeutic paradigm in the clinical setting.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Misra
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
10
|
Wang X, Komasa S, Tahara Y, Inui S, Matsumoto M, Maekawa K. Novel Injectable Collagen/Glycerol/Pullulan Gel Promotes Osteogenic Differentiation of Mesenchymal Stem Cells and the Repair of Rat Cranial Defects. Gels 2024; 10:775. [PMID: 39727533 DOI: 10.3390/gels10120775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Bone tissue engineering is a technique that simulates the bone tissue microenvironment by utilizing cells, tissue scaffolds, and growth factors. The collagen hydrogel is a three-dimensional network bionic material that has properties and structures comparable to those of the extracellular matrix (ECM), making it an ideal scaffold and drug delivery system for tissue engineering. The clinical applications of this material are restricted due to its low mechanical strength. In this investigation, a collagen-based gel (atelocollagen/glycerol/pullulan [Col/Gly/Pul] gel) that is moldable and injectable with high adhesive qualities was created by employing a straightforward technique that involved the introduction of Gly and Pul. This study aimed to characterize the internal morphology and chemical composition of the Col/Gly/Pul gel, as well as to verify its osteogenic properties through in vivo and in vitro experiments. When compared to a standard pure Col hydrogel, this material is more adaptable to the complexity of the local environment of bone defects and the apposition of irregularly shaped flaws due to its greater mechanical strength, injectability, and moldability. Overall, the Col/Gly/Pul gel is an implant that shows great potential for the treatment of complex bone defects and the enhancement of bone regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Satoshi Komasa
- Department of Oral Health Sciences, Osaka Dental University, 1-4-4, Makino-honmachi, Hirakata-shi 573-1144, Osaka, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Shihoko Inui
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Michiaki Matsumoto
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Kenji Maekawa
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| |
Collapse
|
11
|
Liao YR, Tsai YC, Hsieh TH, Tsai MT, Lin FY, Lin SJ, Lin CC, Chiang HY, Chu PH, Li SY. FHL2 in arterial medial calcification in chronic kidney disease. Nephrol Dial Transplant 2024; 39:2025-2039. [PMID: 38664060 PMCID: PMC11596093 DOI: 10.1093/ndt/gfae091] [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: 09/17/2023] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Arterial medial calcification (AMC) is a common complication in individuals with chronic kidney disease (CKD), which can lead to cardiovascular morbidity and mortality. The progression of AMC is controlled by a key transcription factor called runt-related transcription factor 2 (RUNX2), which induces vascular smooth muscle cells (VSMCs) transdifferentiation into an osteogenic phenotype. However, RUNX2 has not been targeted for therapy due to its essential role in bone development. The objective of our study was to discover a RUNX2 coactivator that is highly expressed in arterial VSMCs as a potential therapy for AMC. METHODS We employed transcriptomic analysis of human data and an animal reporter system to pinpoint four and a half LIM domains 2 (FHL2) as a potential target. Subsequently, we investigated the mRNA and protein expression patterns of FHL2 in the aortas of both human and animal subjects with CKD. To examine the role of FHL2 in the RUNX2 transcription machinery, we conducted coimmunoprecipitation and chromatin immunoprecipitation experiments. Next, we manipulated FHL2 expression in cultured VSMCs to examine its impact on high phosphate-induced transdifferentiation. Finally, we employed FHL2-null mice to confirm the role of FHL2 in the development of AMC in vivo. RESULTS Among all the potential RUNX2 cofactors, FHL2 displays selective expression within the cardiovascular system. In the context of CKD subjects, FHL2 undergoes upregulation and translocation from the cytosol to the nucleus of arterial VSMCs. Once in the nucleus, FHL2 interacts structurally and functionally with RUNX2, acting as a coactivator of RUNX2. Notably, the inhibition of FHL2 expression averts transdifferentiation of VSMCs into an osteogenic phenotype and mitigates aortic calcification in uremic animals, without causing any detrimental effects on the skeletal system. CONCLUSION These observations provide evidence that FHL2 is a promising target for treating arterial calcification in patients with CKD.
Collapse
MESH Headings
- Animals
- LIM-Homeodomain Proteins/metabolism
- LIM-Homeodomain Proteins/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/complications
- Humans
- Mice
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Vascular Calcification/genetics
- Male
- Cell Transdifferentiation
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice, Knockout
Collapse
Affiliation(s)
- Yuan-Ru Liao
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yu-Cheng Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Ming-Tsun Tsai
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yen Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Ching Lin
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hou-Yu Chiang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Science, College of Medicine, Chang Guang University, Taoyuan, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital, Taiwan
| | - Szu-Yuan Li
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
12
|
Bessueille L, Briolay A, Guillot N, Mebarek S, Viallon S, Laroche N, Lafage-Proust MH, Magne D. Teriparatide administration is osteoanabolic but does not impact atherosclerotic plaque calcification and progression in a mouse model of menopause. Bone 2024; 190:117316. [PMID: 39491714 DOI: 10.1016/j.bone.2024.117316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Menopause exacerbates osteoporosis and increases the risk of atherosclerotic plaque rupture, leading to cardiovascular mortality. Osteoporotic women are increasingly treated with teriparatide (TPTD, 1-34 parathyroid hormone), one of the few treatments that stimulate bone formation. Despite the fact that atherosclerotic plaque calcification is a hallmark of plaque development, the impact of TPTD administration on plaque calcification remain unclear. In this context, we sought to determine the effects of TPTD administration on atherosclerosis in ovariectomized (OVX) apolipoprotein E deficient mice (ApoE-/-), as a model of postmenopausal osteoporosis. OVX ApoE-/- mice, fed a high fat, high cholesterol diet to induce atherosclerosis, received either vehicle or TPTD daily injections (40 μg/kg/d) for 4 or 10 weeks, at which points plaques are respectively weakly and heavily calcified. After sacrifice, bone remodeling was evaluated by serum markers and bone histomorphometry. Bone architectural parameters were measured by μCT. Aortic plaques were analyzed histologically, and their calcification with von Kossa staining and the calcium tracer Osteosense. Plaque inflammation and calcification markers were measured by RT-qPCR. Intermittent TPTD increased bone volume in OVX mice, due to a higher stimulation of bone formation relatively to bone resorption. These effects were not accompanied by changes in serum levels of cholesterol, triglycerides, glucose or insulin. TPTD neither significantly affected aortic plaque size, inflammation, and calcification, even if it slightly increased vascular smooth muscle cell transdifferentiation into calcifying cells. In conclusion, TPTD exhibits osteoanabolic effects in OVX ApoE-/- mice, without significantly influencing atherosclerotic plaque progression or calcification in the short term.
Collapse
Affiliation(s)
- Laurence Bessueille
- Univ Lyon, Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, F69622 Lyon, France
| | - Anne Briolay
- Univ Lyon, Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, F69622 Lyon, France
| | - Nicolas Guillot
- Université Claude Bernard Lyon 1, Laboratoire Interuniversitaire de Biologie de la Motricité (LiBM) EA7424, Team Vascular Biology and Red Blood Cell, F69622 Villeurbanne, France
| | - Saïda Mebarek
- Univ Lyon, Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, F69622 Lyon, France
| | - Solène Viallon
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F42023 Saint-Étienne, France
| | - Norbert Laroche
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F42023 Saint-Étienne, France
| | - Marie-Hélène Lafage-Proust
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F42023 Saint-Étienne, France
| | - David Magne
- Univ Lyon, Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, F69622 Lyon, France.
| |
Collapse
|
13
|
Wang D, Li Q, Xie C. The role and mechanism of protein post‑translational modification in vascular calcification (Review). Exp Ther Med 2024; 28:419. [PMID: 39301258 PMCID: PMC11411399 DOI: 10.3892/etm.2024.12708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024] Open
Abstract
Vascular calcification is closely associated with morbidity and mortality in patients with chronic kidney disease, atherosclerosis and diabetes. In the past few decades, vascular calcification has been studied extensively and the findings have shown that the mechanism of vascular calcification is not merely a consequence of a high-phosphorus and high-calcium environment but also an active process characterized by abnormal calcium phosphate deposition on blood vessel walls that involves various molecular mechanisms. Recent advances in bioinformatics approaches have led to increasing recognition that aberrant post-translational modifications (PTMs) play important roles in vascular calcification. This review presents the latest progress in clarifying the roles of PTMs, such as ubiquitination, acetylation, carbamylation and glycosylation, as well as signaling pathways, such as the Wnt/β-catenin pathway, in vascular calcification.
Collapse
Affiliation(s)
- Dongyan Wang
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225100, P.R. China
| | - Qin Li
- Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, Jiangsu 225100, P.R. China
| | - Caidie Xie
- Department of Nephrology, Nanjing Second Hospital, Nanjing Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu 210037, P.R. China
| |
Collapse
|
14
|
Liu SF, Kucherenko MM, Sang P, Li Q, Yao J, Nambiar Veetil N, Gransar T, Alesutan I, Voelkl J, Salinas G, Grune J, Simmons S, Knosalla C, Kuebler WM. RUNX2 is stabilised by TAZ and drives pulmonary artery calcification and lung vascular remodelling in pulmonary hypertension due to left heart disease. Eur Respir J 2024; 64:2300844. [PMID: 39542509 DOI: 10.1183/13993003.00844-2023] [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: 05/19/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Calcification is common in chronic vascular disease, yet its role in pulmonary hypertension due to left heart disease is unknown. Here, we probed for the role of runt-related transcription factor-2 (RUNX2), a master transcription factor in osteogenesis, and its regulation by the HIPPO pathway transcriptional coactivator with PDZ-binding motif (TAZ) in the osteogenic reprogramming of pulmonary artery smooth muscle cells and vascular calcification in patients with pulmonary hypertension due to left heart disease. We similarly examined its role using an established rat model of pulmonary hypertension due to left heart disease induced by supracoronary aortic banding. METHODS Pulmonary artery samples were collected from patients and rats with pulmonary hypertension due to left heart disease. Genome-wide RNA sequencing was performed, and pulmonary artery calcification assessed. Osteogenic signalling via TAZ and RUNX2 was delineated by protein biochemistry. In vivo, the therapeutic potential of RUNX2 or TAZ inhibition by CADD522 or verteporfin was tested in the rat model. RESULTS Gene ontology term analysis identified significant enrichment in ossification and osteoblast differentiation genes, including RUNX2, in pulmonary arteries of patients and lungs of rats with pulmonary hypertension due to left heart disease. Pulmonary artery calcification was evident in both patients and rats. Both TAZ and RUNX2 were upregulated and activated in pulmonary artery smooth muscle cells of patients and rats. Co-immunoprecipitation revealed a direct interaction of RUNX2 with TAZ in pulmonary artery smooth muscle cells. TAZ inhibition or knockdown decreased RUNX2 abundance due to accelerated RUNX2 protein degradation rather than reduced de novo synthesis. Inhibition of either TAZ or RUNX2 attenuated pulmonary artery calcification, distal lung vascular remodelling and pulmonary hypertension development in the rat model. CONCLUSION Pulmonary hypertension due to left heart disease is associated with pulmonary artery calcification that is driven by TAZ-dependent stabilisation of RUNX2, causing osteogenic reprogramming of pulmonary artery smooth muscle cells. The TAZ-RUNX2 axis may present a therapeutic target in pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Mariya M Kucherenko
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Pengchao Sang
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Qiuhua Li
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Juquan Yao
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Netra Nambiar Veetil
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Tara Gransar
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Ioana Alesutan
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Salinas
- NGS - Integrative Genomics Core Unit (NIG), Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Grune
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Christoph Knosalla
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors share the last authorship
| | - Wolfgang M Kuebler
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- DZL (German Centre for Lung Research), partner site Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Departements of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
- These authors share the last authorship
| |
Collapse
|
15
|
Docshin P, Panshin D, Malashicheva A. Molecular Interplay in Cardiac Fibrosis: Exploring the Functions of RUNX2, BMP2, and Notch. Rev Cardiovasc Med 2024; 25:368. [PMID: 39484128 PMCID: PMC11522771 DOI: 10.31083/j.rcm2510368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac fibrosis, characterized by the excessive deposition of extracellular matrix proteins, significantly contributes to the morbidity and mortality associated with cardiovascular diseases. This article explores the complex interplay between Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein 2 (BMP2), and Notch signaling pathways in the pathogenesis of cardiac fibrosis. Each of these pathways plays a crucial role in the regulation of cellular functions and interactions that underpin fibrotic processes in the heart. Through a detailed review of current research, we highlight how the crosstalk among RUNX2, BMP2, and Notch not only facilitates our understanding of the fibrotic mechanisms but also points to potential biomolecular targets for intervention. This article delves into the regulatory networks, identifies key molecular mediators, and discusses the implications of these signaling pathways in cardiac structural remodeling. By synthesizing findings from recent studies, we provide insights into the cellular and molecular mechanisms that could guide future research directions, aiming to uncover new therapeutic strategies to manage and treat cardiac fibrosis effectively.
Collapse
Affiliation(s)
- Pavel Docshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Daniil Panshin
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology Russian Academy of Science, 194064 St. Petersburg, Russia
| |
Collapse
|
16
|
Platko K, Gyulay G, Lebeau PF, MacDonald ME, Lynn EG, Byun JH, Igdoura SA, Holden RM, Roubtsova A, Seidah NG, Krepinsky JC, Austin RC. GDF10 is a negative regulator of vascular calcification. J Biol Chem 2024; 300:107805. [PMID: 39307303 PMCID: PMC11541827 DOI: 10.1016/j.jbc.2024.107805] [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: 04/03/2024] [Revised: 08/23/2024] [Accepted: 09/11/2024] [Indexed: 10/27/2024] Open
Abstract
Cardiovascular mortality is particularly high and increasing in patients with chronic kidney disease, with vascular calcification (VC) as a major pathophysiologic feature. VC is a highly regulated biological process similar to bone formation involving osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs). We have previously demonstrated that loss of T-cell death-associated gene 51 (TDAG51) expression leads to an attenuation of medial VC. We now show a significant induction of circulating levels of growth differentiation factor 10 (GDF10) in TDAG51-/- mice, which was of interest due to its established role as an inhibitor of osteoblast differentiation. The objective of this study was to examine the role of GDF10 in the osteogenic transdifferentiation of VSMCs. Using primary mouse and human VSMCs, as well as ex vivo aortic ring cultures, we demonstrated that treatment with recombinant human (rh) GDF10 mitigated phosphate-mediated hydroxyapatite (HA) mineral deposition. Furthermore, ex vivo aortic rings from GDF10-/- mice exhibited increased HA deposition compared to C57BL/6J controls. To explain our observations, we identified that rhGDF10 treatment reduced protein expression of runt-related transcription factor 2, a key driver of osteogenic transdifferentiation of VSMCs and VC. In support of these findings, in vivo treatment with rhGDF10 attenuated VD3-induced VC. Furthermore, we demonstrated an increase in circulating GDF10 in patients with chronic kidney disease with clinically defined severe VC, as assessed by coronary artery calcium score. Thus, our studies identify GDF10 as a novel inhibitor of mineral deposition and as such, may represent a potential novel biomarker and therapeutic target for the detection and management of VC.
Collapse
Affiliation(s)
- Khrystyna Platko
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Gabriel Gyulay
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Paul F Lebeau
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Melissa E MacDonald
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Edward G Lynn
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Jae Hyun Byun
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada
| | - Suleiman A Igdoura
- Department of Biology, McMaster University Medical Centre, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rachel M Holden
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Anna Roubtsova
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Nabil G Seidah
- The Institut de Recherches Cliniques de Montréal (IRCM), Affiliated with Université de Montréal, Montréal, Quebec, Canada
| | - Joan C Krepinsky
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| | - Richard C Austin
- Department of Medicine, Division of Nephrology, McMaster University, and The Research Institute of St Joe's Hamilton, Hamilton, Ontario, Canada.
| |
Collapse
|
17
|
Liang X, Li Y, Wang P, Liu H. Key regulators of vascular calcification in chronic kidney disease: Hyperphosphatemia, BMP2, and RUNX2. PeerJ 2024; 12:e18063. [PMID: 39308809 PMCID: PMC11416758 DOI: 10.7717/peerj.18063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Vascular calcification is quite common in patients with end-stage chronic kidney disease and is a major trigger for cardiovascular complications in these patients. These complications significantly impact the survival rate and long-term prognosis of individuals with chronic kidney disease. Numerous studies have demonstrated that the development of vascular calcification involves various pathophysiological mechanisms, with the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) being of utmost importance. High phosphate levels, bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (RUNX2) play crucial roles in the osteogenic transdifferentiation process of VSMCs. This article primarily reviews the molecular mechanisms by which high phosphate, BMP2, and RUNX2 regulate vascular calcification secondary to chronic kidney disease, and discusses the complex interactions among these factors and their impact on the progression of vascular calcification. The insights provided here aim to offer new perspectives for future research on the phenotypic switching and osteogenic transdifferentiation of VSMCs, as well as to aid in optimizing clinical treatment strategies for this condition, bearing significant clinical and scientific implications.
Collapse
Affiliation(s)
- Xinhua Liang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Yankun Li
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Peng Wang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| | - Huafeng Liu
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| |
Collapse
|
18
|
Ballester-Servera C, Alonso J, Taurón M, Rotllán N, Rodríguez C, Martínez-González J. Lysyl oxidase expression in smooth muscle cells determines the level of intima calcification in hypercholesterolemia-induced atherosclerosis. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2024; 36:286-298. [PMID: 38402026 DOI: 10.1016/j.arteri.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/17/2024] [Accepted: 01/24/2024] [Indexed: 02/26/2024]
Abstract
INTRODUCTION Cardiovascular calcification is an important public health issue with an unmeet therapeutic need. We had previously shown that lysyl oxidase (LOX) activity critically influences vascular wall smooth muscle cells (VSMCs) and valvular interstitial cells (VICs) calcification by affecting extracellular matrix remodeling. We have delved into the participation of LOX in atherosclerosis and vascular calcification, as well as in the mineralization of the aortic valve. METHODS Immunohistochemical and expression studies were carried out in human atherosclerotic lesions and experimental models, valves from patients with aortic stenosis, VICs, and in a genetically modified mouse model that overexpresses LOX in CMLV (TgLOXCMLV). Hyperlipemia and atherosclerosis was induced in mice through the administration of adeno-associated viruses encoding a PCSK9 mutated form (AAV-PCSK9D374Y) combined with an atherogenic diet. RESULTS LOX expression is increased in the neointimal layer of atherosclerotic lesions from human coronary arteries and in VSMC-rich regions of atheromas developed both in the brachiocephalic artery of control (C57BL/6J) animals transduced with PCSK9D374Y and in the aortic root of ApoE-/- mice. In TgLOXCMLV mice, PCSK9D374Y transduction did not significantly alter the enhanced aortic expression of genes involved in matrix remodeling, inflammation, oxidative stress and osteoblastic differentiation. Likewise, LOX transgenesis did not alter the size or lipid content of atherosclerotic lesions in the aortic arch, brachiocephalic artery and aortic root, but exacerbated calcification. Among lysyl oxidase isoenzymes, LOX is the most expressed member of this family in highly calcified human valves, colocalizing with RUNX2 in VICs. The lower calcium deposition and decreased RUNX2 levels triggered by the overexpression of the nuclear receptor NOR-1 in VICs was associated with a reduction in LOX. CONCLUSIONS Our results show that LOX expression is increased in atherosclerotic lesions, and that overexpression of this enzyme in VSMC does not affect the size of the atheroma or its lipid content, but it does affect its degree of calcification. Further, these data suggest that the decrease in calcification driven by NOR-1 in VICs would involve a reduction in LOX. These evidences support the interest of LOX as a therapeutic target in cardiovascular calcification.
Collapse
MESH Headings
- Animals
- Humans
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Protein-Lysine 6-Oxidase/metabolism
- Protein-Lysine 6-Oxidase/genetics
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Vascular Calcification/pathology
- Vascular Calcification/genetics
- Vascular Calcification/etiology
- Vascular Calcification/metabolism
- Disease Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Hypercholesterolemia/complications
- Mice, Inbred C57BL
- Aortic Valve Stenosis/pathology
- Aortic Valve Stenosis/metabolism
- Aortic Valve Stenosis/genetics
- Aortic Valve/pathology
- Aortic Valve/metabolism
- Male
- Proprotein Convertase 9/genetics
- Proprotein Convertase 9/metabolism
- Mice, Transgenic
- Tunica Intima/pathology
- Tunica Intima/metabolism
- Diet, Atherogenic/adverse effects
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, España
| | - Noemí Rotllán
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud CarlosIII, Madrid, España
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, España; CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud CarlosIII, Madrid, España; Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, España.
| |
Collapse
|
19
|
Zheng J, He J, Li H. FAM19A5 in vascular aging and osteoporosis: Mechanisms and the "calcification paradox". Ageing Res Rev 2024; 99:102361. [PMID: 38821416 DOI: 10.1016/j.arr.2024.102361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/05/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Aging induces a progressive decline in the vasculature's structure and function. Vascular aging is a determinant factor for vascular ailments in the elderly. FAM19A5, a recently identified adipokine, has demonstrated involvement in multiple vascular aging-related pathologies, including atherosclerosis, cardio-cerebral vascular diseases and cognitive deficits. This review summarizes the current understanding of FAM19A5' role and explores its putative regulatory mechanisms in various aging-related disorders, including cardiovascular diseases (CVDs), metabolic diseases, neurodegenerative diseases and malignancies. Importantly, we provide novel insights into the underlying therapeutic value of FAM19A5 in osteoporosis. Finally, we outline future perspectives on the diagnostic and therapeutic potential of FAM19A5 in vascular aging-related diseases.
Collapse
Affiliation(s)
- Jin Zheng
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Jieyu He
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Huahua Li
- Department of Geriatrics, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China.
| |
Collapse
|
20
|
Baba I, Matoba T, Katsuki S, Koga JI, Kawahara T, Kimura M, Akita H, Tsutsui H. EVs-miR-17-5p attenuates the osteogenic differentiation of vascular smooth muscle cells potentially via inhibition of TGF-β signaling under high glucose conditions. Sci Rep 2024; 14:16323. [PMID: 39009669 PMCID: PMC11251274 DOI: 10.1038/s41598-024-67006-9] [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: 07/12/2023] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Vascular calcification, which is a major complication of diabetes mellitus, is an independent risk factor for cardiovascular disease. Osteogenic differentiation of vascular smooth muscle cells (VSMCs) is one of the key mechanisms underlying vascular calcification. Emerging evidence suggests that macrophage-derived extracellular vesicles (EVs) may be involved in calcification within atherosclerotic plaques in patients with diabetes mellitus. However, the role of macrophage-derived EVs in the progression of vascular calcification is largely unknown. In this study, we investigated whether macrophage-derived EVs contribute to the osteogenic differentiation of VSMCs under high glucose conditions. We isolated EVs that were secreted by murine peritoneal macrophages under normal glucose (EVs-NG) or high glucose (EVs-HG) conditions. miRNA array analysis in EVs from murine macrophages showed that miR-17-5p was significantly increased in EVs-HG compared with EVs-NG. Prediction analysis with miRbase identified transforming growth factor β receptor type II (TGF-β RII) as a potential target of miR-17-5p. EVs-HG as well as miR-17-5p overexpression with lipid nanoparticles inhibited the gene expression of Runx2, and TGF-β RII. Furthermore, we demonstrated that VSMCs transfected with miR-17-5p mimic inhibited calcium deposition. Our findings reveal a novel role of macrophage-derived EVs in the negative regulation of osteogenic differentiation in VSMCs under high glucose conditions.
Collapse
Affiliation(s)
- Isashi Baba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Jun-Ichiro Koga
- Second Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takuro Kawahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsukuni Kimura
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hidetaka Akita
- Laboratory of Drug Design and Drug Disposition, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
21
|
Wu S, Luo X, Chen Y, Wang Z, Liu X, Sun N, Zhao J, Luo W, Zhang J, Tong X, Huang L, Liu C, Qin Z. Sodium-glucose cotransporter 2 inhibitors attenuate vascular calcification by suppressing endoplasmic reticulum protein thioredoxin domain containing 5 dependent osteogenic reprogramming. Redox Biol 2024; 73:103183. [PMID: 38759418 PMCID: PMC11127605 DOI: 10.1016/j.redox.2024.103183] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/23/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
AIMS Vascular calcification is strongly linked to the development of major adverse cardiovascular events, but effective treatments are lacking. Sodium-glucose cotransporter 2 (SGLT2) inhibitors are an emerging category of oral hypoglycemic drugs that have displayed marked effects on metabolic and cardiovascular diseases, including recently reported vascular medial calcification. However, the roles and underlying mechanisms of SGLT2 inhibitors in vascular calcification have not been fully elucidated. Thus, we aimed to further determine whether SGLT2 inhibitors protect against vascular calcification and to investigate the mechanisms involved. METHODS AND RESULTS A computed tomography angiography investigation of coronary arteries from 1554 patients with type 2 diabetes revealed that SGLT2 inhibitor use was correlated with a lower Agatston calcification score. In the vitamin D3 overdose, 5/6 nephrectomy chronic kidney disease-induced medial calcification and Western diet-induced atherosclerotic intimal calcification models, dapagliflozin (DAPA) substantially alleviated vascular calcification in the aorta. Furthermore, we showed that DAPA reduced vascular calcification via Runx2-dependent osteogenic transdifferentiation in vascular smooth muscle cells (VSMCs). Transcriptome profiling revealed that thioredoxin domain containing 5 (TXNDC5) was involved in the attenuation of vascular calcification by DAPA. Rescue experiments showed that DAPA-induced TXNDC5 downregulation in VSMCs blocked the protective effect on vascular calcification. Furthermore, TXNDC5 downregulation disrupted protein folding-dependent Runx2 stability and promoted subsequent proteasomal degradation. Moreover, DAPA downregulated TXNDC5 expression via amelioration of oxidative stress and ATF6-dependent endoplasmic reticulum stress. Consistently, the class effects of SGLT2 inhibitors on vascular calcification were validated with empagliflozin in intimal and medial calcification models. CONCLUSIONS SGLT2 inhibitors ameliorate vascular calcification through blocking endoplasmic reticulum stress-dependent TXNDC5 upregulation and promoting subsequent Runx2 proteasomal degradation, suggesting that SGLT2 inhibitors are potentially beneficial for vascular calcification treatment and prevention.
Collapse
MESH Headings
- Vascular Calcification/metabolism
- Vascular Calcification/drug therapy
- Vascular Calcification/pathology
- Vascular Calcification/etiology
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Animals
- Humans
- Osteogenesis/drug effects
- Mice
- Glucosides/pharmacology
- Male
- Thioredoxins/metabolism
- Thioredoxins/genetics
- Benzhydryl Compounds/pharmacology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Endoplasmic Reticulum/metabolism
- Endoplasmic Reticulum/drug effects
- Rats
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Disease Models, Animal
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Female
Collapse
Affiliation(s)
- Shaofa Wu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China; Department of Nephrology, Youyang Hospital, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 409800, China
| | - Xiaolin Luo
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Yang Chen
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Zelan Wang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xi Liu
- Department of Nephrology, Youyang Hospital, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 409800, China
| | - Ning Sun
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Junyong Zhao
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Wenjian Luo
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jiawen Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Xiaoyong Tong
- Innovative Drug Research Centre, Chongqing University, Chongqing, 401331, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China.
| |
Collapse
|
22
|
Chen C, Ding Y, Huang Q, Zhang C, Zhao Z, Zhou H, Li D, Zhou G. Relationship between arginine methylation and vascular calcification. Cell Signal 2024; 119:111189. [PMID: 38670475 DOI: 10.1016/j.cellsig.2024.111189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
In patients on maintenance hemodialysis (MHD), vascular calcification (VC) is an independent predictor of cardiovascular disease (CVD), which is the primary cause of death in chronic kidney disease (CKD). The main component of VC in CKD is the vascular smooth muscle cells (VSMCs). VC is an ordered, dynamic activity. Under the stresses of oxidative stress and calcium-‑phosphorus imbalance, VSMCs undergo osteogenic phenotypic transdifferentiation, which promotes the formation of VC. In addition to traditional epigenetics like RNA and DNA control, post-translational modifications have been discovered to be involved in the regulation of VC in recent years. It has been reported that the process of osteoblast differentiation is impacted by catalytic histone or non-histone arginine methylation. Its function in the osteogenic process is comparable to that of VC. Thus, we propose that arginine methylation regulates VC via many signaling pathways, including as NF-B, WNT, AKT/PI3K, TGF-/BMP/SMAD, and IL-6/STAT3. It might also regulate the VC-related calcification regulatory factors, oxidative stress, and endoplasmic reticulum stress. Consequently, we propose that arginine methylation regulates the calcification of the arteries and outline the regulatory mechanisms involved.
Collapse
Affiliation(s)
- Chen Chen
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Yuanyuan Ding
- Department of Pain Management, Shengjing Hospital, China Medical University, China
| | - Qun Huang
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Chen Zhang
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Zixia Zhao
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Detian Li
- Department of Nephrology, Shengjing Hospital, China Medical University, China
| | - Guangyu Zhou
- Department of Nephrology, Shengjing Hospital, China Medical University, China.
| |
Collapse
|
23
|
Ababneh H, Tóth A, Lente G, Balogh E, Csiki DM, Nagy B, Szöőr Á, Jeney V. High phosphate and calcium induce osteoblastic phenotype switching and calcification of corneal epithelial cells in a Runx2-dependent and synergistic manner; a possible mechanism of chronic kidney disease-associated corneal calcification. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167171. [PMID: 38631411 DOI: 10.1016/j.bbadis.2024.167171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Patients with advanced chronic kidney disease (CKD) have elevated circulating calcium × phosphate product levels and exhibit soft tissue calcification. Besides the cardiovascular system, calcification is commonly observed in the cornea in CKD patients on hemodialysis. Cardiovascular calcification is a cell-mediated, highly regulated process, and we hypothesized that a similar regulatory mechanism is implicated in corneal calcification with the involvement of corneal epithelial cells (CECs). We established a mouse model of CKD-associated corneal calcification by inducing CKD in DBA/2J mice with an adenine and high phosphate diet. CKD was associated with aorta and corneal calcification as detected by OsteoSense staining and corneal Ca measurement (1.67-fold elevation, p < 0.001). In vitro, excess phosphate and Ca induced human CEC calcification in a dose-dependent and synergistic manner, without any influence on cell viability. High phosphate and Ca-containing osteogenic medium (OM; 2.5 mmol/L excess phosphate and 0.6 mmol/L excess Ca over control) increased the protein expression of Runx2 and induced its nuclear translocation. OM increased the expression of the bone-specific Ca-binding protein osteocalcin (130-fold increase, p < 0.001). Silencing of Runx2 attenuated OM-induced CEC calcification. Immunohistology revealed upregulation of Runx2 and overlapping between the Runx2 and the Alizarin red positive areas of calcification in the cornea of CKD mice. This work sheds light on the mechanism of CKD-induced corneal calcification and provides tools to test calcification inhibitors for the prevention of this detrimental process.
Collapse
Affiliation(s)
- Haneen Ababneh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gréta Lente
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Máté Csiki
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Doctoral School of Molecular Cell and Immune Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
24
|
Ding R, Huang L, Yan K, Sun Z, Duan J. New insight into air pollution-related cardiovascular disease: an adverse outcome pathway framework of PM2.5-associated vascular calcification. Cardiovasc Res 2024; 120:699-707. [PMID: 38636937 DOI: 10.1093/cvr/cvae082] [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: 11/17/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| |
Collapse
|
25
|
Zhai X, Cao S, Wang J, Qiao B, Liu X, Hua R, Zhao M, Sun S, Han Y, Wu S, Pang J, Yuan Q, Wang B, Xu F, Wei S, Chen Y. Carbonylation of Runx2 at K176 by 4-Hydroxynonenal Accelerates Vascular Calcification. Circulation 2024; 149:1752-1769. [PMID: 38348663 DOI: 10.1161/circulationaha.123.065830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/19/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Vascular calcification, which is characterized by calcium deposition in arterial walls and the osteochondrogenic differentiation of vascular smooth muscle cells, is an actively regulated process that involves complex mechanisms. Vascular calcification is associated with increased cardiovascular adverse events. The role of 4-hydroxynonenal (4-HNE), which is the most abundant stable product of lipid peroxidation, in vascular calcification has been poorly investigated. METHODS Serum was collected from patients with chronic kidney disease and controls, and the levels of 4-HNE and 8-iso-prostaglandin F2α were measured. Sections of coronary atherosclerotic plaques from donors were immunostained to analyze calcium deposition and 4-HNE. A total of 658 patients with coronary artery disease who received coronary computed tomography angiography were recruited to analyze the relationship between coronary calcification and the rs671 mutation in aldehyde dehydrogenase 2 (ALDH2). ALDH2 knockout (ALDH2-/-) mice, smooth muscle cell-specific ALDH2 knockout mice, ALDH2 transgenic mice, and their controls were used to establish vascular calcification models. Primary mouse aortic smooth muscle cells and human aortic smooth muscle cells were exposed to medium containing β-glycerophosphate and CaCl2 to investigate cell calcification and the underlying molecular mechanisms. RESULTS Elevated 4-HNE levels were observed in the serum of patients with chronic kidney disease and model mice and were detected in calcified artery sections by immunostaining. ALDH2 knockout or smooth muscle cell-specific ALDH2 knockout accelerated the development of vascular calcification in model mice, whereas overexpression or activation prevented mouse vascular calcification and the osteochondrogenic differentiation of vascular smooth muscle cells. In patients with coronary artery disease, patients with ALDH2 rs671 gene mutation developed more severe coronary calcification. 4-HNE promoted calcification of both mouse aortic smooth muscle cells and human aortic smooth muscle cells and their osteochondrogenic differentiation in vitro. 4-HNE increased the level of Runx2 (runt-related transcription factor-2), and the effect of 4-HNE on promoting vascular smooth muscle cell calcification was ablated when Runx2 was knocked down. Mutation of Runx2 at lysine 176 reduced its carbonylation and eliminated the 4-HNE-induced upregulation of Runx2. CONCLUSIONS Our results suggest that 4-HNE increases Runx2 stabilization by directly carbonylating its K176 site and promotes vascular calcification. ALDH2 might be a potential target for the treatment of vascular calcification.
Collapse
MESH Headings
- Animals
- Aldehydes/metabolism
- Vascular Calcification/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/pathology
- Humans
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Aldehyde Dehydrogenase, Mitochondrial/genetics
- Aldehyde Dehydrogenase, Mitochondrial/metabolism
- Mice
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Female
- Middle Aged
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Cells, Cultured
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/pathology
- Aged
Collapse
Affiliation(s)
- Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Shengchuan Cao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Rui Hua
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Menglin Zhao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Shukun Sun
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Yu Han
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Shuo Wu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Jiaojiao Pang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Qiuhuan Yuan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Bailu Wang
- National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, China (B.W.)
| | - Feng Xu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| |
Collapse
|
26
|
Xu L, Wang L, Wang Y, Wang Y, Jiang Y, Du P, Cheng J, Zhang C, Wang R, Jiao T, Xing L, Ma J, Li J. PCSK9 inhibitors ameliorate arterial stiffness in ACS patients: evidences from Mendelian randomization, a retrospective study and basic experiments. Front Med (Lausanne) 2024; 11:1408760. [PMID: 38860206 PMCID: PMC11163136 DOI: 10.3389/fmed.2024.1408760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/09/2024] [Indexed: 06/12/2024] Open
Abstract
Background Current evidences suggest that Proprotein Convertase Subtilisin/kexin Type 9 inhibitors (PCSK9i) exhibit a protective influence on acute coronary syndrome (ACS). Nevertheless, further investigation is required to comprehend the impact and mechanisms of these pharmaceutical agents on inflammatory factors and arterial stiffness (AS) in patients with ACS. Consequently, the objective of this study is to ascertain the influence of PCSK9i on arterial stiffness in ACS patients and elucidate the underlying mechanisms behind their actions. Methods This study employed Mendelian randomization (MR) analysis to examine the association between genetic prediction of PCSK9 inhibition and arterial stiffness. Data of 71 patients with ACS were retrospectively collected, including PCSK9i group (n = 36, PCSK9 inhibitors combined with statins) and control group (n = 35, statins only). Blood lipid levels, inflammatory markers and pulse wave velocity (PWV) data were collected before treatment and at 1 and 6 months after treatment for analysis. Additionally, cell experiments were conducted to investigate the impact of PCSK9i on osteogenesis of vascular smooth muscle cells (VSMCs), utilizing western blot (WB), enzyme-linked immunosorbent assay (ELISA), and calcification index measurements. Results The results of the MR analysis suggest that genetic prediction of PCSK9 inhibition has potential to reduce the PWV. Following treatment of statins combined with PCSK9 inhibitors for 1 and 6 months, the PCSK9i group exhibited significantly lower levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), C-reactive protein (CRP), interleukin-6 (IL-6), fibrinogen (FIB) and procalcitonin (PCT) compared to the control group (p < 0.05). Additionally, PWV in the PCSK9i group demonstrated significant reduction after 6 months of treatment and was found to be associated with the circulating CRP level. In cell experiments, PCSK9i pretreatment ameliorated osteogenesis of VSMCs through reducing the deposition of calcium ions, alkaline phosphatase (ALP) activity, and expression of runt-related transcription factor 2 (RUNX2). Conclusion PCSK9i have potential to enhance arterial stiffness in ACS patients. Specifically, at the clinical level, this impact may be attributed to alterations in circulating CRP levels. At the cellular level, it is associated with the signaling pathway linked to RUNX2.
Collapse
Affiliation(s)
- Linghao Xu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Liang Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanqi Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yiqiong Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanzhen Jiang
- Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Peizhao Du
- Department of Cardiology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Cheng
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunsheng Zhang
- Department of Cardiology, East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Ruijie Wang
- Department of Cardiology, Harbin Medical University First Affiliated Hospital, Harbin, China
| | - Tiantian Jiao
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijian Xing
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiangping Ma
- School of Medicine, Tongji University, Shanghai, China
| | - Jiming Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
27
|
Ababneh H, Balogh E, Csiki DM, Lente G, Fenyvesi F, Tóth A, Jeney V. High glucose promotes osteogenic differentiation of human lens epithelial cells through hypoxia-inducible factor (HIF) activation. J Cell Physiol 2024; 239:e31211. [PMID: 38304971 DOI: 10.1002/jcp.31211] [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: 08/04/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/03/2024]
Abstract
Cataract, a leading cause of blindness, is characterised by lens opacification. Type 2 diabetes is associated with a two- to fivefold higher prevalence of cataracts. The risk of cataract formation increases with the duration of diabetes and the severity of hyperglycaemia. Hydroxyapatite deposition is present in cataractous lenses that could be the consequence of osteogenic differentiation and calcification of lens epithelial cells (LECs). We hypothesised that hyperglycaemia might promote the osteogenic differentiation of human LECs (HuLECs). Osteogenic medium (OM) containing excess phosphate and calcium with normal (1 g/L) or high (4.5 g/L) glucose was used to induce HuLEC calcification. High glucose accelerated and intensified OM-induced calcification of HuLECs, which was accompanied by hyperglycaemia-induced upregulation of the osteogenic markers Runx2, Sox9, alkaline phosphatase and osteocalcin, as well as nuclear translocation of Runx2. High glucose-induced calcification was abolished in Runx2-deficient HuLECs. Additionally, high glucose stabilised the regulatory alpha subunits of hypoxia-inducible factor 1 (HIF-1), triggered nuclear translocation of HIF-1α and increased the expression of HIF-1 target genes. Gene silencing of HIF-1α or HIF-2α attenuated hyperglycaemia-induced calcification of HuLECs, while hypoxia mimetics (desferrioxamine, CoCl2) enhanced calcification of HuLECs under normal glucose conditions. Overall, this study suggests that high glucose promotes HuLEC calcification via Runx2 and the activation of the HIF-1 signalling pathway. These findings may provide new insights into the pathogenesis of diabetic cataracts, shedding light on potential factors for intervention to treat this sight-threatening condition.
Collapse
Affiliation(s)
- Haneen Ababneh
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Enikő Balogh
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dávid Máté Csiki
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Gréta Lente
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, Debrecen, Hungary
| | - Ferenc Fenyvesi
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Andrea Tóth
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória Jeney
- Research Centre for Molecular Medicine, MTA-DE Lendület Vascular Pathophysiology Research Group, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
28
|
Xiong J, Lin W, Yuan C, Bian J, Diao Y, Xu X, Ni B, Zhang H, Shao Y. SIRT6-mediated Runx2 downregulation inhibits osteogenic differentiation of human aortic valve interstitial cells in calcific aortic valve disease. Eur J Pharmacol 2024; 968:176423. [PMID: 38365109 DOI: 10.1016/j.ejphar.2024.176423] [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: 12/30/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Calcific aortic valve disease (CAVD) is a progressive cardiovascular disorder involving multiple pathogenesis. Effective pharmacological therapies are currently unavailable. Sirtuin6 (SIRT6) has been shown to protect against aortic valve calcification in CAVD. The exact regulatory mechanism of SIRT6 in osteoblastic differentiation remains to be determined, although it inhibits osteogenic differentiation of aortic valve interstitial cells. We demonstrated that SIRT6 was markedly downregulated in calcific human aortic valves. Mechanistically, SIRT6 suppressed osteogenic differentiation in human aortic valve interstitial cells (HAVICs), as confirmed by loss- and gain-of-function experiments. SIRT6 directly interacted with Runx2, decreased Runx2 acetylation levels, and facilitated Runx2 nuclear export to inhibit the osteoblastic phenotype transition of HAVICs. In addition, the AKT signaling pathway acted upstream of SIRT6. Together, these findings elucidate that SIRT6-mediated Runx2 downregulation inhibits aortic valve calcification and provide novel insights into therapeutic strategies for CAVD.
Collapse
Affiliation(s)
- Jiaqi Xiong
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Wenfeng Lin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Chunze Yuan
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Jinhui Bian
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Yifei Diao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Xinyang Xu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China
| | - Buqing Ni
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China.
| | - Hui Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China; Lab of Public Platform, Nanjing Medical University, Nanjing, China.
| | - Yongfeng Shao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road, No. 300, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
29
|
Scotti MM, Wilson BK, Bubenik JL, Yu F, Swanson MS, Allen JB. Spaceflight effects on human vascular smooth muscle cell phenotype and function. NPJ Microgravity 2024; 10:41. [PMID: 38548798 PMCID: PMC10979029 DOI: 10.1038/s41526-024-00380-w] [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: 07/26/2023] [Accepted: 03/08/2024] [Indexed: 04/01/2024] Open
Abstract
The cardiovascular system is strongly impacted by the hazards of spaceflight. Astronauts spending steadily increasing lengths of time in microgravity are subject to cardiovascular deconditioning resulting in loss of vascular tone, reduced total blood volume, and diminished cardiac output. Appreciating the mechanisms by which the cells of the vasculature are altered during spaceflight will be integral to understanding and combating these deleterious effects as the human presence in space advances. In this study, we performed RNA-Seq analysis coupled with review by QIAGEN Ingenuity Pathway Analysis software on human aortic smooth muscle cells (HASMCs) cultured for 3 days in microgravity and aboard the International Space Station to assess the transcriptomic changes that occur during spaceflight. The results of our RNA-Seq analysis show that SMCs undergo a wide range of transcriptional alteration while in space, significantly affecting 4422 genes. SMCs largely down-regulate markers of the contractile, synthetic, and osteogenic phenotypes including smooth muscle alpha actin (αSMA), matrix metalloproteinases (MMPs), and bone morphogenic proteins (BMPs). Additionally, components of several cellular signaling pathways were strongly impacted including the STAT3, NFκB, PI3K/AKT, HIF1α, and Endothelin pathways. This study highlights the significant changes in transcriptional behavior SMCs exhibit during spaceflight and puts these changes in context to better understand vascular function in space.
Collapse
Affiliation(s)
- Marina M Scotti
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
| | - Brandon K Wilson
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jodi L Bubenik
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, FL, USA
| | - Fahong Yu
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, USA
| | - Maurice S Swanson
- Department of Molecular Genetics and Microbiology, Center for NeuroGenetics, University of Florida, Gainesville, FL, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
30
|
Turner ME, Beck L, Hill Gallant KM, Chen Y, Moe OW, Kuro-o M, Moe S, Aikawa E. Phosphate in Cardiovascular Disease: From New Insights Into Molecular Mechanisms to Clinical Implications. Arterioscler Thromb Vasc Biol 2024; 44:584-602. [PMID: 38205639 PMCID: PMC10922848 DOI: 10.1161/atvbaha.123.319198] [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: 01/12/2024]
Abstract
Hyperphosphatemia is a common feature in patients with impaired kidney function and is associated with increased risk of cardiovascular disease. This phenomenon extends to the general population, whereby elevations of serum phosphate within the normal range increase risk; however, the mechanism by which this occurs is multifaceted, and many aspects are poorly understood. Less than 1% of total body phosphate is found in the circulation and extracellular space, and its regulation involves multiple organ cross talk and hormones to coordinate absorption from the small intestine and excretion by the kidneys. For phosphate to be regulated, it must be sensed. While mostly enigmatic, various phosphate sensors have been elucidated in recent years. Phosphate in the circulation can be buffered, either through regulated exchange between extracellular and cellular spaces or through chelation by circulating proteins (ie, fetuin-A) to form calciprotein particles, which in themselves serve a function for bulk mineral transport and signaling. Either through direct signaling or through mediators like hormones, calciprotein particles, or calcifying extracellular vesicles, phosphate can induce various cardiovascular disease pathologies: most notably, ectopic cardiovascular calcification but also left ventricular hypertrophy, as well as bone and kidney diseases, which then propagate phosphate dysregulation further. Therapies targeting phosphate have mostly focused on intestinal binding, of which appreciation and understanding of paracellular transport has greatly advanced the field. However, pharmacotherapies that target cardiovascular consequences of phosphate directly, such as vascular calcification, are still an area of great unmet medical need.
Collapse
Affiliation(s)
- Mandy E. Turner
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Laurent Beck
- Nantes Université, CNRS, Inserm, l’institut du thorax, F-44000 Nantes, France
| | - Kathleen M Hill Gallant
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham
- Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Makoto Kuro-o
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Sharon Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
31
|
Faleeva M, Ahmad S, Theofilatos K, Lynham S, Watson G, Whitehead M, Marhuenda E, Iskratsch T, Cox S, Shanahan CM. Sox9 Accelerates Vascular Aging by Regulating Extracellular Matrix Composition and Stiffness. Circ Res 2024; 134:307-324. [PMID: 38179698 PMCID: PMC10826924 DOI: 10.1161/circresaha.123.323365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Vascular calcification and increased extracellular matrix (ECM) stiffness are hallmarks of vascular aging. Sox9 (SRY-box transcription factor 9) has been implicated in vascular smooth muscle cell (VSMC) osteo/chondrogenic conversion; however, its relationship with aging and calcification has not been studied. METHODS Immunohistochemistry was performed on human aortic samples from young and aged patients. Young and senescent primary human VSMCs were induced to produce ECM, and Sox9 expression was manipulated using adenoviral overexpression and depletion. ECM properties were characterized using atomic force microscopy and proteomics, and VSMC phenotype on hydrogels and the ECM were examined using confocal microscopy. RESULTS In vivo, Sox9 was not spatially associated with vascular calcification but correlated with the senescence marker p16 (cyclin-dependent kinase inhibitor 2A). In vitro Sox9 showed mechanosensitive responses with increased expression and nuclear translocation in senescent cells and on stiff matrices. Sox9 was found to regulate ECM stiffness and organization by orchestrating changes in collagen (Col) expression and reducing VSMC contractility, leading to the formation of an ECM that mirrored that of senescent cells. These ECM changes promoted phenotypic modulation of VSMCs, whereby senescent cells plated on ECM synthesized from cells depleted of Sox9 returned to a proliferative state, while proliferating cells on a matrix produced by Sox9 expressing cells showed reduced proliferation and increased DNA damage, reiterating features of senescent cells. LH3 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3) was identified as an Sox9 target and key regulator of ECM stiffness. LH3 is packaged into extracellular vesicles and Sox9 promotes extracellular vesicle secretion, leading to increased LH3 deposition within the ECM. CONCLUSIONS These findings highlight the crucial role of ECM structure and composition in regulating VSMC phenotype. We identify a positive feedback cycle, whereby cellular senescence and increased ECM stiffening promote Sox9 expression, which, in turn, drives further ECM modifications to further accelerate stiffening and senescence.
Collapse
Affiliation(s)
- Maria Faleeva
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Sadia Ahmad
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Konstantinos Theofilatos
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry (S.L.) King’s College London, United Kingdom
| | - Gabriel Watson
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Meredith Whitehead
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Emilie Marhuenda
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Thomas Iskratsch
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Susan Cox
- Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine (S.C.) King’s College London, United Kingdom
| | - Catherine M. Shanahan
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| |
Collapse
|
32
|
Ballester-Servera C, Cañes L, Alonso J, Puertas-Umbert L, Vázquez-Sufuentes P, Taurón M, Roselló-Díez E, Marín F, Rodríguez C, Martínez-González J. Upregulation of NOR-1 in calcified human vascular tissues: impact on osteogenic differentiation and calcification. Transl Res 2024; 264:1-14. [PMID: 37690706 DOI: 10.1016/j.trsl.2023.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
Cardiovascular calcification is a significant public health issue whose pathophysiology is not fully understood. NOR-1 regulates critical processes in cardiovascular remodeling, but its contribution to ectopic calcification is unknown. NOR-1 was overexpressed in human calcific aortic valves and calcified atherosclerotic lesions colocalizing with RUNX2, a factor essential for osteochondrogenic differentiation and calcification. NOR-1 and osteogenic markers were upregulated in calcifying human valvular interstitial cells (VICs) and human vascular smooth muscle cells (VSMCs). Gain- and loss-of-function approaches demonstrated that NOR-1 negatively modulates the expression of osteogenic genes relevant for the osteogenic transdifferentiation (RUNX2, IL-6, BMP2, and ALPL) and calcification of VICs. VSMCs from transgenic mice overexpressing NOR-1 in these cells (TgNOR-1VSMC) expressed lower basal levels of osteogenic genes (IL-6, BMP2, ALPL, OPN) than cells from WT littermates, and their upregulation by a high-phosphate osteogenic medium (OM) was completely prevented by NOR-1 transgenesis. Consistently, this was associated with a dramatic reduction in the calcification of both transgenic VSMCs and aortic rings from TgNOR-1VSMC mice exposed to OM. Atherosclerosis and calcification were induce in mice by the administration of AAV-PCSK9D374Y and a high-fat/high-cholesterol diet. Challenged-TgNOR-1VSMC mice exhibited decreased vascular expression of osteogenic markers, and both less atherosclerotic burden (assessed in whole aorta and lesion size in aortic arch and brachiocephalic artery) and less vascular calcification (assessed either by near-infrared fluorescence imaging or histological analysis) than WT mice. Our data indicate that NOR-1 negatively modulates the expression of genes critically involved in the osteogenic differentiation of VICs and VSMCs, thereby restraining ectopic cardiovascular calcification.
Collapse
Affiliation(s)
- Carme Ballester-Servera
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Laia Cañes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Judith Alonso
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Lidia Puertas-Umbert
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - Paula Vázquez-Sufuentes
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
| | - Manel Taurón
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Elena Roselló-Díez
- Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Departamento de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau-Universitat Autònoma de Barcelona (HSCSP-UAB), Barcelona, Spain
| | - Francisco Marín
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Departamento de Cardiología, Hospital Clínico Universitario Virgen de la Arrixaca-Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau (IRHSCSP), Barcelona, Spain
| | - José Martínez-González
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), Barcelona, Spain; CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.
| |
Collapse
|
33
|
McNeill MC, Li Mow Chee F, Ebrahimighaei R, Sala-Newby GB, Newby AC, Hathway T, Annaiah AS, Joseph S, Carrabba M, Bond M. Substrate stiffness promotes vascular smooth muscle cell calcification by reducing the levels of nuclear actin monomers. J Mol Cell Cardiol 2024; 187:65-79. [PMID: 38181546 DOI: 10.1016/j.yjmcc.2023.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Vascular calcification (VC) is a prevalent independent risk factor for adverse cardiovascular events and is associated with diabetes, hypertension, chronic kidney disease, and atherosclerosis. However, the mechanisms regulating the osteogenic differentiation of vascular smooth muscle cells (VSMC) are not fully understood. METHODS Using hydrogels of tuneable stiffness and lysyl oxidase-mediated stiffening of human saphenous vein ex vivo, we investigated the role of substrate stiffness in the regulation of VSMC calcification. RESULTS We demonstrate that increased substrate stiffness enhances VSMC osteogenic differentiation and VSMC calcification. We show that the effects of substrate stiffness are mediated via a reduction in the level of actin monomer within the nucleus. We show that in cells interacting with soft substrate, elevated levels of nuclear actin monomer repress osteogenic differentiation and calcification by repressing YAP-mediated activation of both TEA Domain transcription factor (TEAD) and RUNX Family Transcription factor 2 (RUNX2). CONCLUSION This work highlights for the first time the role of nuclear actin in mediating substrate stiffness-dependent VSMC calcification and the dual role of YAP-TEAD and YAP-RUNX2 transcriptional complexes.
Collapse
Affiliation(s)
- M C McNeill
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - F Li Mow Chee
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - R Ebrahimighaei
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - G B Sala-Newby
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - A C Newby
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - T Hathway
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - A S Annaiah
- Bristol Heart Institute, University Hospital, Bristol NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| | - S Joseph
- Bristol Heart Institute, University Hospital, Bristol NHS Foundation Trust, Bristol BS2 8HW, United Kingdom
| | - M Carrabba
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom
| | - M Bond
- Department of Translational Health Sciences, Bristol Medical School, Bristol BS2 8HW, United Kingdom.
| |
Collapse
|
34
|
Cai Z, Liu Z, Zhang Y, Ma H, Li R, Guo S, Wu S, Guo X. Associations Between Life's Essential 8 and Abdominal Aortic Calcification Among Middle-Aged and Elderly Populations. J Am Heart Assoc 2023; 12:e031146. [PMID: 38063150 PMCID: PMC10863763 DOI: 10.1161/jaha.123.031146] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Abdominal aortic calcification (AAC) is an independent risk factor for cardiovascular disease. We aim to examine the associations between Life's Essential 8 (LE8), the recently updated measurement of cardiovascular health (CVH), and AAC among participants aged ≥40 years. METHODS AND RESULTS This population-based cross-sectional study used data from the National Health and Nutrition Examination Survey in 2013 to 2014. AAC (AAC score>0) and severe AAC (AAC score>6) were quantified by the Kauppila score system. Multiple linear, multivariable logistic, and restricted cubic spline models were used to assess the associations. A total of 2369 participants were included with a mean AAC score of 1.41 (0.13). Participants in the high-cardiovascular-health group had lower AAC scores, lower prevalence of AAC, and lower prevalence of severe AAC. After the adjustment of potential confounders (age, sex, race and ethnicity, education levels, marital status, poverty income ratio, estimated glomerular filtration rate, serum creatinine, serum uric acid, serum phosphorus, and serum total calcium), higher cardiovascular health was significantly associated with lower risk of AAC. Meanwhile, elevated nicotine exposure score, blood glucose score, and blood pressure score within the LE8 components were significantly associated with lower risk of AAC. Also, nonlinear dose-response relationships were observed. Subgroup analyses (age strata, sex, poverty income ratio, education levels, marital status) indicated the inverse associations of LE8 and AAC were generally similar in different populations. CONCLUSIONS LE8 was negatively and nonlinearly related to the risk of AAC among middle-aged and older populations. Meanwhile, LE8 components should prioritize higher scores for nicotine exposure, blood glucose, and blood pressure evaluations.
Collapse
Affiliation(s)
- Zongao Cai
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| | - Hongxuan Ma
- Department of Kidney TransportationThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Ruihui Li
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shuang Guo
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shiyong Wu
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Xueli Guo
- Department of Vascular SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
35
|
Hashmi S, Shah PW, Aherrahrou Z, Aikawa E, Aherrahrou R. Beyond the Basics: Unraveling the Complexity of Coronary Artery Calcification. Cells 2023; 12:2822. [PMID: 38132141 PMCID: PMC10742130 DOI: 10.3390/cells12242822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
Coronary artery calcification (CAC) is mainly associated with coronary atherosclerosis, which is an indicator of coronary artery disease (CAD). CAC refers to the accumulation of calcium phosphate deposits, classified as micro- or macrocalcifications, that lead to the hardening and narrowing of the coronary arteries. CAC is a strong predictor of future cardiovascular events, such as myocardial infarction and sudden death. Our narrative review focuses on the pathophysiology of CAC, exploring its link to plaque vulnerability, genetic factors, and how race and sex can affect the condition. We also examined the connection between the gut microbiome and CAC, and the impact of genetic variants on the cellular processes involved in vascular calcification and atherogenesis. We aimed to thoroughly analyze the existing literature to improve our understanding of CAC and its potential clinical and therapeutic implications.
Collapse
Affiliation(s)
- Satwat Hashmi
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan;
| | - Pashmina Wiqar Shah
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Rédouane Aherrahrou
- Institute for Cardiogenetics, Universität zu Lübeck, 23562 Lübeck, Germany; (P.W.S.); (Z.A.)
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Heart Centre Lübeck, 23562 Lübeck, Germany
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
36
|
Phadwal K, Tan X, Koo E, Zhu D, MacRae VE. Metformin ameliorates valve interstitial cell calcification by promoting autophagic flux. Sci Rep 2023; 13:21435. [PMID: 38052777 PMCID: PMC10698150 DOI: 10.1038/s41598-023-47774-6] [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: 03/15/2023] [Accepted: 11/18/2023] [Indexed: 12/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is the most common heart disease of the developed world. It has previously been established that metformin administration reduces arterial calcification via autophagy; however, whether metformin directly regulates CAVD has yet to be elucidated. In the present study we investigated whether metformin alleviates valvular calcification through the autophagy-mediated recycling of Runx2. Calcification was reduced in rat valve interstitial cells (RVICs) by metformin treatment (0.5-1.5 mM) (P < 0.01), with a marked decrease in Runx2 protein expression compared to control cells (P < 0.05). Additionally, upregulated expression of Atg3 and Atg7 (key proteins required for autophagosome formation), was observed following metformin treatment (1 mM). Blocking autophagic flux using Bafilomycin-A1 revealed colocalisation of Runx2 with LC3 puncta in metformin treated RVICs (P < 0.001). Comparable Runx2 accumulation was seen in LC3 positive autolysosomes present within cells that had been treated with both metformin and hydroxychloroquine in combination (P < 0.001). Mechanistic studies employing three-way co-immunoprecipitation with Runx2, p62 and LC3 suggested that Runx2 binds to LC3-II upon metformin treatment in VICs. Together these studies suggest that the utilisation of metformin may represent a novel strategy for the treatment of CAVD.
Collapse
Affiliation(s)
- K Phadwal
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK.
| | - X Tan
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
- Guangzhou Institute of Cardiovascular Diseases, Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - E Koo
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| | - D Zhu
- Guangzhou Institute of Cardiovascular Diseases, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - V E MacRae
- The Roslin Institute & R(D)SVS, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, UK
| |
Collapse
|
37
|
de Vries PS, Conomos MP, Singh K, Nicholson CJ, Jain D, Hasbani NR, Jiang W, Lee S, Lino Cardenas CL, Lutz SM, Wong D, Guo X, Yao J, Young EP, Tcheandjieu C, Hilliard AT, Bis JC, Bielak LF, Brown MR, Musharoff S, Clarke SL, Terry JG, Palmer ND, Yanek LR, Xu H, Heard-Costa N, Wessel J, Selvaraj MS, Li RH, Sun X, Turner AW, Stilp AM, Khan A, Newman AB, Rasheed A, Freedman BI, Kral BG, McHugh CP, Hodonsky C, Saleheen D, Herrington DM, Jacobs DR, Nickerson DA, Boerwinkle E, Wang FF, Heiss G, Jun G, Kinney GL, Sigurslid HH, Doddapaneni H, Hall IM, Bensenor IM, Broome J, Crapo JD, Wilson JG, Smith JA, Blangero J, Vargas JD, Mosquera JV, Smith JD, Viaud-Martinez KA, Ryan KA, Young KA, Taylor KD, Lange LA, Emery LS, Bittencourt MS, Budoff MJ, Montasser ME, Yu M, Mahaney MC, Mahamdeh MS, Fornage M, Franceschini N, Lotufo PA, Natarajan P, Wong Q, Mathias RA, Gibbs RA, Do R, Mehran R, Tracy RP, Kim RW, Nelson SC, Damrauer SM, Kardia SL, Rich SS, Fuster V, Napolioni V, Zhao W, Tian W, Yin X, Min YI, Manning AK, Peloso G, Kelly TN, O’Donnell CJ, Morrison AC, Curran JE, Zapol WM, et alde Vries PS, Conomos MP, Singh K, Nicholson CJ, Jain D, Hasbani NR, Jiang W, Lee S, Lino Cardenas CL, Lutz SM, Wong D, Guo X, Yao J, Young EP, Tcheandjieu C, Hilliard AT, Bis JC, Bielak LF, Brown MR, Musharoff S, Clarke SL, Terry JG, Palmer ND, Yanek LR, Xu H, Heard-Costa N, Wessel J, Selvaraj MS, Li RH, Sun X, Turner AW, Stilp AM, Khan A, Newman AB, Rasheed A, Freedman BI, Kral BG, McHugh CP, Hodonsky C, Saleheen D, Herrington DM, Jacobs DR, Nickerson DA, Boerwinkle E, Wang FF, Heiss G, Jun G, Kinney GL, Sigurslid HH, Doddapaneni H, Hall IM, Bensenor IM, Broome J, Crapo JD, Wilson JG, Smith JA, Blangero J, Vargas JD, Mosquera JV, Smith JD, Viaud-Martinez KA, Ryan KA, Young KA, Taylor KD, Lange LA, Emery LS, Bittencourt MS, Budoff MJ, Montasser ME, Yu M, Mahaney MC, Mahamdeh MS, Fornage M, Franceschini N, Lotufo PA, Natarajan P, Wong Q, Mathias RA, Gibbs RA, Do R, Mehran R, Tracy RP, Kim RW, Nelson SC, Damrauer SM, Kardia SL, Rich SS, Fuster V, Napolioni V, Zhao W, Tian W, Yin X, Min YI, Manning AK, Peloso G, Kelly TN, O’Donnell CJ, Morrison AC, Curran JE, Zapol WM, Bowden DW, Becker LC, Correa A, Mitchell BD, Psaty BM, Carr JJ, Pereira AC, Assimes TL, Stitziel NO, Hokanson JE, Laurie CA, Rotter JI, Vasan RS, Post WS, Peyser PA, Miller CL, Malhotra R. Whole-genome sequencing uncovers two loci for coronary artery calcification and identifies ARSE as a regulator of vascular calcification. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1159-1172. [PMID: 38817323 PMCID: PMC11138106 DOI: 10.1038/s44161-023-00375-y] [Show More Authors] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 10/25/2023] [Indexed: 06/01/2024]
Abstract
Coronary artery calcification (CAC) is a measure of atherosclerosis and a well-established predictor of coronary artery disease (CAD) events. Here we describe a genome-wide association study (GWAS) of CAC in 22,400 participants from multiple ancestral groups. We confirmed associations with four known loci and identified two additional loci associated with CAC (ARSE and MMP16), with evidence of significant associations in replication analyses for both novel loci. Functional assays of ARSE and MMP16 in human vascular smooth muscle cells (VSMCs) demonstrate that ARSE is a promoter of VSMC calcification and VSMC phenotype switching from a contractile to a calcifying or osteogenic phenotype. Furthermore, we show that the association of variants near ARSE with reduced CAC is likely explained by reduced ARSE expression with the G allele of enhancer variant rs5982944. Our study highlights ARSE as an important contributor to atherosclerotic vascular calcification, and a potential drug target for vascular calcific disease.
Collapse
Affiliation(s)
- Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew P. Conomos
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Kuldeep Singh
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Christopher J. Nicholson
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Deepti Jain
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Natalie R. Hasbani
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Wanlin Jiang
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Sujin Lee
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Christian L Lino Cardenas
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Sharon M. Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR)
Center, Department of Population Medicine, Harvard Medical School and Harvard
Pilgrim Health Care Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of
Public Health, Boston, MA, USA
| | - Doris Wong
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Erica P. Young
- Cardiovascular Division, Department of Internal Medicine,
Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine Tcheandjieu
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - Austin T. Hilliard
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA,
USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of
Medicine, University of Washington, Seattle, WA, USA
| | - Lawrence F. Bielak
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Michael R. Brown
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Shaila Musharoff
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Genetics, Stanford University School of
Medicine, Stanford, CA, USA
| | - Shoa L. Clarke
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - James G. Terry
- Department of Radiology, Vanderbilt Translational and
Clinical Cardiovascular Research Center, Vanderbilt University Medical Center,
Nashville, TN, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of
Medicine, Winston-Salem, NC, USA
| | - Lisa R. Yanek
- Division of General Internal Medicine, Department of
Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huichun Xu
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Nancy Heard-Costa
- Boston University School of Medicine, Boston, MA,
USA
- Boston University and National Heart, Lung, and Blood
Institute’s Framingham Heart Study, Framingham, MA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Fairbanks School of Public
Health, Indiana University, Indianapolis, IN, USA
- Diabetes Translational Research Center, Indiana
University, Indianapolis, IN, USA
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center and Center for Genomic
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad
Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston,
MA, USA
| | - Rebecca H. Li
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Xiao Sun
- School of Public Health and Tropical Medicine, Department
of Epidemiology, Tulane University, New Orleans, LA, USA
- College of Medicine, Department of Medicine, Division of
Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Adrienne M. Stilp
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Alyna Khan
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Anne B. Newman
- Department of Epidemiology, Graduate School of Public
Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Asif Rasheed
- Center For Non-Communicable Diseases, Karachi,
Pakistan
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine,
Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Brian G. Kral
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Caitlin P. McHugh
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Chani Hodonsky
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Danish Saleheen
- Center For Non-Communicable Diseases, Karachi,
Pakistan
- Department of Medicine, Columbia University Irving
Medical Center, New York, NY, USA
- Department of Cardiology, Columbia University Irving
Medical Center, New York, NY, USA
| | - David M. Herrington
- Department of Internal Medicine, Section of
Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC,
USA
| | - David R. Jacobs
- Division of Epidemiology and Community Health, University
of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Deborah A. Nickerson
- Department of Genome Sciences, University of Washington,
Seattle, WA, USA
- Northwest Genomics Center, University of Washington,
Seattle, WA, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of
Medicine, Houston, TX, USA
| | - Fei Fei Wang
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global
Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Goo Jun
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Greg L. Kinney
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Haakon H. Sigurslid
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | | | - Ira M. Hall
- Yale Center for Genomic Health, Yale School of Medicine,
New Haven, CT, USA
| | - Isabela M. Bensenor
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Jai Broome
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - James D. Crapo
- Department of Medicine, National Jewish Health, Denver,
CO, USA
| | - James G. Wilson
- Division of Cardiology, Beth Israel Deaconess Medical
Center, Boston, MA, USA
| | - Jennifer A. Smith
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research,
University of Michigan, Ann Arbor, MI, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Jose D. Vargas
- Medstar Heart and Vascular Institute, Medstar Georgetown
University Hospital, Washington, DC, USA
| | - Jose Verdezoto Mosquera
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Joshua D. Smith
- Department of Genome Sciences, University of Washington,
Seattle, WA, USA
- Northwest Genomics Center, University of Washington,
Seattle, WA, USA
| | | | - Kathleen A. Ryan
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Kendra A. Young
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Leslie A. Lange
- Department of Medicine, University of Colorado Denver,
Anschutz Medical Campus, Aurora, CO, USA
| | - Leslie S. Emery
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Marcio S. Bittencourt
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Matthew J. Budoff
- Department of Medicine, The Lundquist Institute for
Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - May E. Montasser
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
| | - Miao Yu
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Michael C. Mahaney
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Mohammed S Mahamdeh
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Myriam Fornage
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
- Institute of Molecular Medicine, McGovern Medical School,
The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global
Public health, University of North Carolina, Chapel Hill, NC, USA
| | - Paulo A. Lotufo
- Center for Clinical and Epidemiological Research,
University Hospital, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad
Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston,
MA, USA
| | - Quenna Wong
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Rasika A. Mathias
- Division of General Internal Medicine, Department of
Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Allergy and Clinical Immunology, Department
of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of
Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor
College of Medicine, Houston, TX, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine,
Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School
of Medicine at Mount Sinai, New York, NY, USA
| | - Roxana Mehran
- Icahn School of Medicine at Mount Sinai, New York, NY,
USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Robert
Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Sarah C. Nelson
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Scott M. Damrauer
- Corporal Michael J. Crescenz VA Medical Center,
Philadelphia, PA, USA
- Department of Surgery, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, USA
| | - Sharon L.R. Kardia
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares
Carlos III, Madrid, Spain
- Mount Sinai Heart Center, New York, NY, USA
| | - Valerio Napolioni
- Genomic And Molecular Epidemiology (GAME) Lab, School of
Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Wei Zhao
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Wenjie Tian
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical
Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Yuan-I Min
- Jackson Heart Study, Department of Medicine, University
of Mississippi Medical Center, Jackson, MS, USA
| | - Alisa K. Manning
- Clinical and Translation Epidemiology Unit, Department of
Medicine, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical and Population
Genetics, Broad Institute, Cambridge, MA, USA
| | - Gina Peloso
- Department of Biostatistics, Boston University School of
Public Health, Boston, MA, USA
| | - Tanika N. Kelly
- College of Medicine, Department of Medicine, Division of
Nephrology, University of Illinois Chicago, Chicago, IL, USA
| | - Christopher J. O’Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women’s Hospital,
Boston, MA, USA
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human
Genetics, and Environmental Sciences, School of Public Health, The University of
Texas Health Science Center at Houston, Houston, TX, USA
| | - Joanne E. Curran
- Department of Human Genetics, University of Texas Rio
Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of
Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Warren M. Zapol
- Department of Anesthesia, Critical Care and Pain Medicine
at Massachusetts General Hospital, Boston, MA, USA
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of
Medicine, Winston-Salem, NC, USA
| | - Lewis C. Becker
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University
of Mississippi Medical Center, Jackson, MS, USA
- Department of Population Health Science, University of
Mississippi Medical Center, Jackson, MS, USA
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition,
Department of Medicine, University of Maryland School of Medicine, Baltimore, MD,
USA
- Geriatrics Research and Education Clinical Center,
Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of
Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington,
Seattle, WA, USA
- Department of Health Services, University of Washington,
Seattle, WA, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt Translational and
Clinical Cardiovascular Research Center, Vanderbilt University Medical Center,
Nashville, TN, USA
| | - Alexandre C. Pereira
- Department of Genetics, Harvard Medical School, Boston,
MA, USA
- Laboratory of Genetics and Molecular Cardiology, Heart
Institute, University of São Paulo, São Paulo, Brazil
| | - Themistocles L. Assimes
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of
Medicine, Stanford, CA, USA
| | - Nathan O. Stitziel
- Cardiovascular Division, Department of Internal Medicine,
Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of
Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University School
of Medicine, St. Louis, MO, USA
| | - John E. Hokanson
- Department of Epidemiology, Colorado School of Public
Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cecelia A. Laurie
- Genetic Analysis Center, Department of Biostatistics,
School of Public Health, University of Washington, Seattle, WA, USA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population
Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical
Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ramachandran S. Vasan
- Boston University and National Heart, Lung, and Blood
Institute’s Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of
Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of
Public Health, Boston, MA, USA
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, Johns
Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patricia A. Peyser
- School of Public Health, Department of Epidemiology,
University of Michigan, Ann Arbor, MI, USA
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia
School of Medicine, Charlottesville, VA, USA
| | - Rajeev Malhotra
- Cardiovascular Research Center, Division of Cardiology,
Department of Medicine, Massachusetts General Hospital, Harvard Medical School,
Boston, MA, USA
| |
Collapse
|
38
|
Mizrak D, Zhao Y, Feng H, Macaulay J, Tang Y, Sultan Z, Zhao G, Guo Y, Zhang J, Yang B, Eugene Chen Y. Single-Molecule Spatial Transcriptomics of Human Thoracic Aortic Aneurysms Uncovers Calcification-Related CARTPT-Expressing Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2023; 43:2285-2297. [PMID: 37823268 PMCID: PMC10842613 DOI: 10.1161/atvbaha.123.319329] [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] [Received: 03/21/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Although single-cell RNA-sequencing is commonly applied to dissect the heterogeneity in human tissues, it involves the preparation of single-cell suspensions via cell dissociation, causing loss of spatial information. In this study, we employed high-resolution single-cell transcriptome imaging to reveal rare smooth muscle cell (SMC) types in human thoracic aortic aneurysm (TAA) tissue samples. METHODS Single-molecule spatial distribution of transcripts from 140 genes was analyzed in fresh-frozen human TAA samples with region and sex-matched controls. In vitro studies and tissue staining were performed to examine human CART prepropeptide (CARTPT) regulation and function. RESULTS We captured thousands of cells per sample including a spatially distinct CARTPT-expressing SMC subtype enriched in male TAA samples. Immunoassays confirmed human CART (cocaine- and amphetamine-regulated transcript) protein enrichment in male TAA tissue and truncated CARTPT secretion into cell culture medium. Oxidized low-density lipoprotein, a cardiovascular risk factor, induced CARTPT expression, whereas CARTPT overexpression in human aortic SMCs increased the expression of key osteochondrogenic transcription factors and reduced contractile gene expression. Recombinant human CART treatment of human SMCs further confirmed this phenotype. Alizarin red staining revealed calcium deposition in male TAA samples showing similar localization with human CART staining. CONCLUSIONS Here, we demonstrate the feasibility of single-molecule imaging in uncovering rare SMC subtypes in the diseased human aorta, a difficult tissue to dissociate. We identified a spatially distinct CARTPT-expressing SMC subtype enriched in male human TAA samples. Our functional studies suggest that human CART promotes osteochondrogenic switch of aortic SMCs, potentially leading to medial calcification of the thoracic aorta.
Collapse
Affiliation(s)
- Dogukan Mizrak
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yang Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Hao Feng
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jane Macaulay
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Ying Tang
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Zain Sultan
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Guizhen Zhao
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Yanhong Guo
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Bo Yang
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
| | - Y. Eugene Chen
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Blümm C, Bonaterra GA, Schwarzbach H, Eiden LE, Weihe E, Kinscherf R. PAC1 deficiency reduces chondrogenesis in atherosclerotic lesions of hypercholesterolemic ApoE-deficient mice. BMC Cardiovasc Disord 2023; 23:566. [PMID: 37980508 PMCID: PMC10657554 DOI: 10.1186/s12872-023-03600-5] [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: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Induction of chondrogenesis is associated with progressive atherosclerosis. Deficiency of the ADCYAP1 gene encoding pituitary adenylate cyclase-activating peptide (PACAP) aggravates atherosclerosis in ApoE deficient (ApoE-/-) mice. PACAP signaling regulates chondrogenesis and osteogenesis during cartilage and bone development. Therefore, this study aimed to decipher whether PACAP signaling is related to atherogenesis-related chondrogenesis in the ApoE-/- mouse model of atherosclerosis and under the influence of a high-fat diet. METHODS For this purpose, PACAP-/-/ApoE-/-, PAC1-/-/ApoE-/-, and ApoE-/- mice, as well as wildtype (WT) mice, were studied under standard chow (SC) or cholesterol-enriched diet (CED) for 20 weeks. The amount of cartilage matrix in atherosclerotic lesions of the brachiocephalic trunk (BT) with maximal lumen stenosis was monitored by alcian blue and collagen II staining on deparaffinized cross sections. The chondrogenic RUNX family transcription factor 2 (RUNX2), macrophages [(MΦ), Iba1+], and smooth muscle cells (SMC, sm-α-actin) were immunohistochemically analyzed and quantified. RESULTS ApoE-/- mice fed either SC or CED revealed an increase of alcian blue-positive areas within the media compared to WT mice. PAC1-/-/ApoE-/- mice under CED showed a reduction in the alcian blue-positive plaque area in the BT compared to ApoE-/- mice. In contrast, PACAP deficiency in ApoE-/- mice did not affect the chondrogenic signature under either diet. CONCLUSIONS Our data show that PAC1 deficiency reduces chondrogenesis in atherosclerotic plaques exclusively under conditions of CED-induced hypercholesterolemia. We conclude that CED-related chondrogenesis occurs in atherosclerotic plaques via transdifferentiation of SMCs and MΦ, partly depending on PACAP signaling through PAC1. Thus, PAC1 antagonists or PACAP agonists may offer therapeutic potential against pathological chondrogenesis in atherosclerotic lesions generated under hypercholesterolemic conditions, especially in familial hypercholesterolemia. This discovery opens therapeutic perspectives to be used in the treatment against the progression of atherosclerosis.
Collapse
Affiliation(s)
- C Blümm
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - G A Bonaterra
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany.
| | - H Schwarzbach
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - L E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health Intramural Research Program, Bethesda, MD, 20814, USA
| | - E Weihe
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| | - R Kinscherf
- Institute for Anatomy and Cell Biology, Department of Medical Cell Biology, University of Marburg, 35032, Marburg, Germany
| |
Collapse
|
40
|
Elshamy AM, Hafez YM, Safa MAE, Ibrahim HA, Khalfallah M, Rizk FH, Eltabaa EF, Ghafar MTA, Atef MM. The role of miR-433-3p in vascular calcification in type 2 diabetic patients: targeting WNT/β-Catenin and RANKL/RANK/OPG signaling pathways. Mol Biol Rep 2023; 50:9073-9083. [PMID: 37728820 PMCID: PMC10635945 DOI: 10.1007/s11033-023-08792-9] [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: 05/24/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Vascular calcification (VC) is a major predictor of cardiovascular diseases that represent the principal cause of mortality among type-2 diabetic patients. Accumulating data suggest the vital role of some microRNAs on vascular calcification as an epigenetic regulator. Thus, we assessed herein, the role of serum miR-433-3p in vascular calcification in type-2 diabetic patients. METHODS Twenty healthy subjects (control group) and forty diabetic patients (20 without VC and 20 with VC) were involved in the study. miR-433-3p gene expression was measured. Runx2, Dickkopf-1 (DKK1), β-catenin, Receptor activator of nuclear factor kappa-B ligand (RANKL), and osteoprotegerin (OPG) levels in serum were assessed by ELISA technique. RESULTS Diabetes patients had significantly lower levels of miR-433-3p expression in comparison to the control group, with the lowest levels being found in diabetic patients with VC. Furthermore, Runx2, β-catenin, and RANKL levels were significantly increased with concomitant lower DKK1 and OPG levels detected in the two diabetic groups especially those with VC. CONCLUSION Collectively, the study documented that down-regulation of miR-433-3p may contribute to the development of VC through activating WNT/β-Catenin and RANKL/RANK/OPG signaling pathways.
Collapse
Affiliation(s)
- Amira M Elshamy
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, 31511, Egypt.
| | - Yasser Mostafa Hafez
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed A E Safa
- Internal Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hoda A Ibrahim
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, 31511, Egypt
| | - Mohamed Khalfallah
- Cardiovascular Medicine Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Fatma H Rizk
- Medical Physiology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman F Eltabaa
- Medical Physiology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Marwa Mohamed Atef
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El Geesh Street, Tanta, 31511, Egypt
| |
Collapse
|
41
|
Mohanraj N, Prasanth S, Rajapriya P, Vinothkumar G, Vinodhini VM, Janardhanan R, Venkataraman P. Bisphenol A accelerates the vascular complications in patients with Type 2 diabetes mellitus through vascular calcification-a molecular approach. Int Arch Occup Environ Health 2023; 96:1291-1299. [PMID: 37698613 DOI: 10.1007/s00420-023-02007-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
PURPOSE Environmental pollutant Bisphenol A (BPA) strongly interacts with insulin resistance, which leads to type 2 diabetes mellitus (T2DM). Uncontrolled glucose levels in both blood and urine develops vascular complications in T2DM patients. However, glucose-controlled diabetic patients are also affected by vascular complications due to vascular calcification, and there is a lack of clinically relevant data on BPA levels available in patients with T2DM-associated vascular complications due to vascular calcification. Therefore, we measured BPA levels in T2DM-associated vascular complications and correlated systemic BPA levels with vascular calcification-related gene expression. METHODS This study included 120 participants with T2DM and its associated vascular complications. Serum and urinary BPA were estimated using an ELISA kit, and gene expression of the study participants in peripheral blood mononuclear cells (PBMCs) was studied with quantitative real-time PCR. RESULTS Serum and urinary BPA levels were higher in T2DM and its associated vascular complications with CVD and DN patients compared to control. Both Serum and urinary BPA had higher significance with Sirt1 (p < 0.001, p < 0.001), Runx2 (p < 0.01, p < 0.001) and IL-1beta (p < 0.001, p < 0.02) gene expression in the study groups, but, TNF-alpha significant with Serum BPA (p < 0.04), not urinary BPA (p < 0.31). CONCLUSION BPA levels were positively correlated with lower Sirt1 and increased Runx2 in T2DM-associated vascular complications patients. Also, higher expression of IL-1beta and TNF-alpha was observed in T2DM-associated vascular complications patients. Our study is the first to associate BPA levels with vascular calcification in patients with T2DM and its associated vascular complications.
Collapse
Affiliation(s)
- N Mohanraj
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chennai, India
| | - S Prasanth
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chennai, India
| | - P Rajapriya
- Department of Transfusion Medicine, HLA and Immunology, Dr Rela Institute and Medical Centre, Chennai, India
| | - G Vinothkumar
- Department of Clinical Research, Dr V Balaji Dr V Seshiah Diabetes Care and Research Institute, Chennai, India
| | - V M Vinodhini
- Department of Biochemistry, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chennai, India
| | - Rajiv Janardhanan
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chennai, India
| | - P Venkataraman
- Department of Medical Research, SRM Medical College Hospital and Research Centre, SRM Institute of Science and Technology, Chennai, India.
| |
Collapse
|
42
|
Kamei Y, Okumura Y, Adachi Y, Mori Y, Sakai M, Ohnishi K, Ohminami H, Masuda M, Yamanaka-Okumura H, Taketani Y. Humoral and cellular factors inhibit phosphate-induced vascular calcification during the growth period. J Clin Biochem Nutr 2023; 73:198-204. [PMID: 37970550 PMCID: PMC10636584 DOI: 10.3164/jcbn.23-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 02/25/2023] [Indexed: 11/17/2023] Open
Abstract
Hyperphosphatemia is an independent and non-classical risk factor of cardiovascular disease and mortality in patients with chronic kidney disease (CKD). Increased levels of extracellular inorganic phosphate (Pi) are known to directly induce vascular calcification, but the detailed underlying mechanism has not been clarified. Although serum Pi levels during the growth period are as high as those observed in hyperphosphatemia in adult CKD, vascular calcification does not usually occur during growth. Here, we have examined whether the defence system against Pi-induced vascular calcification can exist during the growth period using mice model. We found that calcification propensity of young serum (aged 3 weeks) was significantly lower than that of adult serum (10 months), possibly due to high fetuin-A levels. In addition, when the aorta was cultured in high Pi medium in vitro, obvious calcification was observed in the adult aorta but not in the young aorta. Furthermore, culture in high Pi medium increased the mRNA level of tissue-nonspecific alkaline phosphatase (TNAP), which degrades pyrophosphate, only in the adult aorta. Collectively, our findings indicate that the aorta in growing mouse may be resistant to Pi-induced vascular calcification via a mechanism in which high serum fetuin-A levels and suppressed TNAP expression.
Collapse
Affiliation(s)
- Yuki Kamei
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food and Nutrition, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yosuke Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuichiro Adachi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yuki Mori
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Maiko Sakai
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Kohta Ohnishi
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hirokazu Ohminami
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Masashi Masuda
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hisami Yamanaka-Okumura
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
- Department of Food Science and Nutrition, Doshisha Women’s College of Liberal Arts, Teramachi Nishi-iru, Imadegawa-dori, Kamigyo-ku, Kyoto 602-0893, Japan
| | - Yutaka Taketani
- Department of Clinical Nutrition and Food Management, Tokushima University Graduate School of Medical Nutrition, 3-18-15 Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
43
|
Feng W, Guan Z, Ying WZ, Xing D, Ying KE, Sanders PW. Matrix metalloproteinase-9 regulates afferent arteriolar remodeling and function in hypertension-induced kidney disease. Kidney Int 2023; 104:740-753. [PMID: 37423509 PMCID: PMC10854403 DOI: 10.1016/j.kint.2023.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 06/01/2023] [Accepted: 06/22/2023] [Indexed: 07/11/2023]
Abstract
This study tested if matrix metalloproteinase (MMP)-9 promoted microvascular pathology that initiates hypertensive (HT) kidney disease in salt-sensitive (SS) Dahl rats. SS rats lacking Mmp9 (Mmp9-/-) and littermate control SS rats were studied after one week on a normotensive 0.3% sodium chloride (Pre-HT SS and Pre-HT Mmp9-/-) or a hypertension-inducing diet containing 4.0% sodium chloride (HT SS and HT Mmp9-/-). Telemetry-monitored blood pressure of both the HT SS and HT Mmp9-/- rats increased and did not differ. Kidney microvessel transforming growth factor-beta 1 (Tgfb1) mRNA did not differ between Pre-HT SS and Pre-HT Mmp9-/- rats, but with hypertension and expression of Mmp9 and Tgfb1 increased in HT SS rats, along with phospho-Smad2 labeling of nuclei of vascular smooth muscle cells, and with peri-arteriolar fibronectin deposition. Loss of MMP-9 prevented hypertension-induced phenotypic transformation of microvascular smooth muscle cells and the expected increased microvascular expression of pro-inflammatory molecules. Loss of MMP-9 in vascular smooth muscle cells in vitro prevented cyclic strain-induced production of active TGF-β1 and phospho-Smad2/3 stimulation. Afferent arteriolar autoregulation was impaired in HT SS rats but not in HT Mmp9-/- rats or the HT SS rats treated with doxycycline, an MMP inhibitor. HT SS but not HT Mmp9-/- rats showed decreased glomerular Wilms Tumor 1 protein-positive cells (a marker of podocytes) along with increased urinary podocin and nephrin mRNA excretion, all indicative of glomerular damage. Thus, our findings support an active role for MMP-9 in a hypertension-induced kidney microvascular remodeling process that promotes glomerular epithelial cell injury in SS rats.
Collapse
Affiliation(s)
- Wenguang Feng
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Zhengrong Guan
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wei-Zhong Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dongqi Xing
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kai Er Ying
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Paul W Sanders
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA; Birmingham Veterans Affairs Health Care System, Birmingham, Alabama, USA.
| |
Collapse
|
44
|
Pineda-Castillo SA, Acar H, Detamore MS, Holzapfel GA, Lee CH. Modulation of Smooth Muscle Cell Phenotype for Translation of Tissue-Engineered Vascular Grafts. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:574-588. [PMID: 37166394 PMCID: PMC10618830 DOI: 10.1089/ten.teb.2023.0006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/12/2023]
Abstract
Translation of small-diameter tissue-engineered vascular grafts (TEVGs) for the treatment of coronary artery disease (CAD) remains an unfulfilled promise. This is largely due to the limited integration of TEVGs into the native vascular wall-a process hampered by the insufficient smooth muscle cell (SMC) infiltration and extracellular matrix deposition, and low vasoactivity. These processes can be promoted through the judicious modulation of the SMC toward a synthetic phenotype to promote remodeling and vascular integration; however, the expression of synthetic markers is often accompanied by a decrease in the expression of contractile proteins. Therefore, techniques that can precisely modulate the SMC phenotypical behavior could have the potential to advance the translation of TEVGs. In this review, we describe the phenotypic diversity of SMCs and the different environmental cues that allow the modulation of SMC gene expression. Furthermore, we describe the emerging biomaterial approaches to modulate the SMC phenotype in TEVG design and discuss the limitations of current techniques. In addition, we found that current studies in tissue engineering limit the analysis of the SMC phenotype to a few markers, which are often the characteristic of early differentiation only. This limited scope has reduced the potential of tissue engineering to modulate the SMC toward specific behaviors and applications. Therefore, we recommend using the techniques presented in this review, in addition to modern single-cell proteomics analysis techniques to comprehensively characterize the phenotypic modulation of SMCs. Expanding the holistic potential of SMC modulation presents a great opportunity to advance the translation of living conduits for CAD therapeutics.
Collapse
Affiliation(s)
- Sergio A. Pineda-Castillo
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Michael S. Detamore
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Chung-Hao Lee
- Biomechanics and Biomaterials Design Laboratory, School of Aerospace and Mechanical Engineering, The University of Oklahoma, Norman, Oklahoma, USA
- Institute for Biomedical Engineering, Science and Technology, The University of Oklahoma, Norman, Oklahoma, USA
| |
Collapse
|
45
|
Li XZ, Xiong ZC, Zhang SL, Hao QY, Liu ZY, Zhang HF, Wang JF, Gao JW, Liu PM. Upregulated LncRNA H19 Sponges MiR-106a-5p and Contributes to Aldosterone-Induced Vascular Calcification via Activating the Runx2-Dependent Pathway. Arterioscler Thromb Vasc Biol 2023; 43:1684-1699. [PMID: 37409531 DOI: 10.1161/atvbaha.123.319308] [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: 12/20/2022] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Excess aldosterone is implicated in vascular calcification (VC), but the mechanism by which aldosterone-MR (mineralocorticoid receptor) complex promotes VC is unclear. Emerging evidence indicates that long-noncoding RNA H19 (H19) plays a critical role in VC. We examined whether aldosterone-induced osteogenic differentiation of vascular smooth muscle cells (VSMCs) through H19 epigenetic modification of Runx2 (runt-related transcription factor-2) in a MR-dependent manner. METHODS We induced in vivo rat model of chronic kidney disease using a high adenine and phosphate diet to explore the relationship among aldosterone, MR, H19, and VC. We also cultured human aortic VSMCs to explore the roles of H19 in aldosterone-MR complex-induced osteogenic differentiation and calcification of VSMCs. RESULTS H19 and Runx2 were significantly increased in aldosterone-induced VSMC osteogenic differentiation and VC, both in vitro and in vivo, which were significantly blocked by the MR antagonist spironolactone. Mechanistically, our findings reveal that the aldosterone-activated MR bound to H19 promoter and increased its transcriptional activity, as determined by chromatin immunoprecipitation, electrophoretic mobility shift assay, and luciferase reporter assay. Silencing H19 increased microRNA-106a-5p (miR-106a-5p) expression, which subsequently inhibited aldosterone-induced Runx2 expression at the posttranscriptional level. Importantly, we observed a direct interaction between H19 and miR-106a-5p, and downregulation of miR-106a-5p efficiently reversed the suppression of Runx2 induced by H19 silencing. CONCLUSIONS Our study clarifies a novel mechanism by which upregulation of H19 contributes to aldosterone-MR complex-promoted Runx2-dependent VSMC osteogenic differentiation and VC through sponging miR-106a-5p. These findings highlight a potential therapeutic target for aldosterone-induced VC.
Collapse
Affiliation(s)
- Xiong-Zhi Li
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Now with Cardiovascular Department, the First Affiliated Hospital of Shaoyang University, Hunan, China (X.-Z.L.)
| | - Zhuo-Chao Xiong
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shao-Ling Zhang
- Department of Endocrinology (S.-L.Z.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Yun Hao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhao-Yu Liu
- Medical Research Center (Z.-Y.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hai-Feng Zhang
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Wei Gao
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pin-Ming Liu
- Department of Cardiology, Guangzhou Key Laboratory on the Molecular Mechanisms of Major Cardiovascular Disease, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology (X.-Z.L., Z.-C.X., Q.-Y.H., H.-F.Z., J.-F.W., J.-W.G., P.-M.L.), Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
46
|
Meng Z, Zhang S, Li W, Wang Y, Wang M, Liu X, Liu CL, Liao S, Liu T, Yang C, Lindholt JS, Rasmussen LM, Obel LM, Stubbe J, Diederichsen AC, Sun Y, Chen Y, Yu PB, Libby P, Shi GP, Guo J. Cationic proteins from eosinophils bind bone morphogenetic protein receptors promoting vascular calcification and atherogenesis. Eur Heart J 2023; 44:2763-2783. [PMID: 37279475 PMCID: PMC10393071 DOI: 10.1093/eurheartj/ehad262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 06/08/2023] Open
Abstract
AIMS Blood eosinophil count and eosinophil cationic protein (ECP) concentration are risk factors of cardiovascular diseases. This study tested whether and how eosinophils and ECP contribute to vascular calcification and atherogenesis. METHODS AND RESULTS Immunostaining revealed eosinophil accumulation in human and mouse atherosclerotic lesions. Eosinophil deficiency in ΔdblGATA mice slowed atherogenesis with increased lesion smooth muscle cell (SMC) content and reduced calcification. This protection in ΔdblGATA mice was muted when mice received donor eosinophils from wild-type (WT), Il4-/-, and Il13-/- mice or mouse eosinophil-associated-ribonuclease-1 (mEar1), a murine homologue of ECP. Eosinophils or mEar1 but not interleukin (IL) 4 or IL13 increased the calcification of SMC from WT mice but not those from Runt-related transcription factor-2 (Runx2) knockout mice. Immunoblot analyses showed that eosinophils and mEar1 activated Smad-1/5/8 but did not affect Smad-2/3 activation or expression of bone morphogenetic protein receptors (BMPR-1A/1B/2) or transforming growth factor (TGF)-β receptors (TGFBR1/2) in SMC from WT and Runx2 knockout mice. Immunoprecipitation showed that mEar1 formed immune complexes with BMPR-1A/1B but not TGFBR1/2. Immunofluorescence double-staining, ligand binding, and Scatchard plot analysis demonstrated that mEar1 bound to BMPR-1A and BMPR-1B with similar affinity. Likewise, human ECP and eosinophil-derived neurotoxin (EDN) also bound to BMPR-1A/1B on human vascular SMC and promoted SMC osteogenic differentiation. In a cohort of 5864 men from the Danish Cardiovascular Screening trial and its subpopulation of 394 participants, blood eosinophil counts and ECP levels correlated with the calcification scores of different arterial segments from coronary arteries to iliac arteries. CONCLUSION Eosinophils release cationic proteins that can promote SMC calcification and atherogenesis using the BMPR-1A/1B-Smad-1/5/8-Runx2 signalling pathway.
Collapse
Affiliation(s)
- Zhaojie Meng
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Shuya Zhang
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou 571199, Hainan, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, Jilin, China
| | - Yunzhe Wang
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Minjie Wang
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Xin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Cong-Lin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sha Liao
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Tianxiao Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Chongzhe Yang
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
- Department of Geriatrics, National Key Clinical Specialty, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou 510000, Guangdong, China
| | - Jes S Lindholt
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
- Elite Research Centre of Individualized Treatment for Arterial Disease, University Hospital, Odense, Denmark
| | - Lars M Rasmussen
- Elite Research Centre of Individualized Treatment for Arterial Disease, University Hospital, Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Lasse M Obel
- Elite Research Centre of Individualized Treatment for Arterial Disease, University Hospital, Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Jane Stubbe
- Cardiovascular and Renal Research unit, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Axel C Diederichsen
- Elite Research Centre of Individualized Treatment for Arterial Disease, University Hospital, Odense, Denmark
- Department of Cardiology, Odense University Hospital, Odense, Denmark
| | - Yong Sun
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Birmingham VA Medical Center, Research Department, Birmingham, AL 35294, USA
| | - Yabing Chen
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Birmingham VA Medical Center, Research Department, Birmingham, AL 35294, USA
| | - Paul B Yu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Peter Libby
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, NRB-7, Boston, MA 02115, USA
| | - Junli Guo
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou 571199, Hainan, China
| |
Collapse
|
47
|
Zhang W, Sun Y, Yang Y, Chen Y. Impaired intracellular calcium homeostasis enhances protein O-GlcNAcylation and promotes vascular calcification and stiffness in diabetes. Redox Biol 2023; 63:102720. [PMID: 37230005 PMCID: PMC10225928 DOI: 10.1016/j.redox.2023.102720] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/13/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Vascular calcification is accelerated in patients with diabetes mellitus and increases risk of cardiovascular events and mortality. Vascular smooth muscle cells (VSMC) play a key role in regulating vascular tone and contribute significantly to the development of diabetic vasculopathy. In this study, the function of stromal interaction molecule 1 (STIM1), an important regulator for intracellular calcium homeostasis, in diabetic vascular calcification was investigated, and the underlying molecular mechanisms were uncovered. A SMC-specific STIM1 deletion mouse model (STIM1Δ/Δ) was generated by breeding the STIM1 floxed mice (STIM1f/f) with SM22α-Cre transgenic mice. Using aortic arteries from the STIM1Δ/Δ mice and their STIM1f/f littermates, we found that SMC-specific STIM1 deletion induced calcification of aortic arteries cultured in osteogenic media ex vivo. Furthermore, STIM1 deficiency promoted osteogenic differentiation and calcification of VSMC from the STIM1Δ/Δ mice. In the low-dose streptozotocin (STZ)-induced mouse model of diabetes, SMC-specific STIM1 deletion markedly enhanced STZ-induced vascular calcification and stiffness in the STIM1Δ/Δ mice. The diabetic mice with SMC-specific STIM1 ablation also exhibited increased aortic expression of the key osteogenic transcription factor, Runx2, and protein O-GlcNAcylation, an important post-translational modulation that we have reported to promote vascular calcification and stiffness in diabetes. Consistently, elevation of O-GlcNAcylation was demonstrated in aortic arteries and VSMC from the STIM1Δ/Δ mice. Inhibition of O-GlcNAcylation with a pharmacological inhibitor abolished STIM1 deficiency-induced VSMC calcification, supporting a critical role of O-GlcNAcylation in mediating STIM1 deficiency-induced VSMC calcification. Mechanistically, we identified that STIM1 deficiency resulted in impaired calcium homeostasis, which activated calcium signaling and increased endoplasmic reticulum (ER) stress in VSMC, while inhibition of ER stress attenuated STIM1-induced elevation of protein O-GlcNAcylation. In conclusion, the study has demonstrated a causative role of SMC-expressed STIM1 in regulating vascular calcification and stiffness in diabetes. We have further identified a novel mechanisms underlying STIM1 deficiency-induced impairment of calcium homeostasis and ER stress in upregulation of protein O-GlcNAcylation in VSMC, which promotes VSMC osteogenic differentiation and calcification in diabetes.
Collapse
Affiliation(s)
- Weiping Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Cardiology, First Affiliated Hospital of Xi'An JiaoTong University, Xi'An, PR China
| | - Yong Sun
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Youfeng Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
48
|
Ibragimova AG, Stanishevskiy YM, Plakkhin AM, Zubko AV, Darvish NA, Koassary AK, Shindyapina AV. Comparative analysis of calcified soft tissues revealed shared deregulated pathways. Front Aging Neurosci 2023; 15:1131548. [PMID: 37441678 PMCID: PMC10335799 DOI: 10.3389/fnagi.2023.1131548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction Calcification of soft tissues is a common age-related pathology that primarily occurs within vascular tissue. The mechanisms underlying pathological calcification in humans and tissue specificity of the process is still poorly understood. Previous studies examined calcified tissues on one to one basis, thus preventing comparison of deregulated pathways across tissues. Purpose This study aimed to establish common and tissue-specific changes associated with calcification in aorta, artery tibial, coronary artery and pituitary gland in subjects from the Genotype-Tissue Expression (GTEx) dataset using its RNA sequencing and histological data. Methods We used publicly available data from the GTEx database https://gtexportal.org/home/aboutGTEx. All GTEx tissue samples were derived by the GTEx consorcium from deceased donors, with age from 20 to 79, both men and women. GTEx study authorization was obtained via next-of-kin consent for the collection and banking of de-identified tissue samples for scientific research. Hematoxylin and eosin (H&E) staining of arteries were manually graded based on the presence of calcification on a scale from zero to four, where zero designates absence of calcification and four designates severe calcification. Samples with fat contamination and mislabeled tissues were excluded, which left 430 aorta, 595 artery tibial, 124 coronary artery, and 283 pituitary samples for downstream gene expression analysis. Transcript levels of protein-coding genes were associated with calcification grade using sex, age bracket and cause of death as covariates, and tested for pathway enrichment using gene set enrichment analysis. Results We identified calcification deposits in 28 (6.5%) aortas, 121 (20%), artery tibials, 54 (43%), coronary arteries, and 24 (8%) pituitary glands of GTEx subjects. We observed an age-dependent increase in incidence of calcification in all vascular tissues, but not in pituitary. Subjects with calcification in the artery tibial were significantly more likely to have calcification in the coronary artery (OR = 2.56, p = 6.3e-07). Markers of calcification previously established in preclinical and in vitro studies, e.g., BMP2 and RUNX2, were deregulated in the calcified tibial and coronary arteries, confirming the relevance of these genes to human pathology. Differentially expressed genes associated with calcification poorly overlapped across tissues suggesting tissue-specific nuances in mechanisms of calcification. Nevertheless, calcified arteries unanimously down-regulated pathways of intracellular transport and up-regulated inflammatory pathways suggesting these as universal targets for pathological calcification. In particular, PD-1 and PD-L1 genes were up-regulated in calcified tissues but not in the blood of the same subjects, suggesting that localized inflammation contributes to pathological calcification. Conclusion Pathological calcification is a prevalent disease of aging that shares little changes in expression in individual genes across tissues. However, our analysis suggests that it potentially can be targeted by alleviating local inflammation of soft tissues.
Collapse
Affiliation(s)
| | | | | | | | - Nidal Akhmedovich Darvish
- Bakoulev National Medical Research Center for Cardiovascular Surgery, Russian Federation, Moscow, Russia
| | - Anton Karenovich Koassary
- Bakoulev National Medical Research Center for Cardiovascular Surgery, Russian Federation, Moscow, Russia
| | - Anastasia V. Shindyapina
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Retro Biosciences Inc., Redwood City, CA, United States
| |
Collapse
|
49
|
Franceschi RT, Hallett SA, Ge C. Discoidin domain receptors; an ancient family of collagen receptors has major roles in bone development, regeneration and metabolism. FRONTIERS IN DENTAL MEDICINE 2023; 4:1181817. [PMID: 38222874 PMCID: PMC10785288 DOI: 10.3389/fdmed.2023.1181817] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
The extracellular matrix (ECM) niche plays a critical role in determining cellular behavior during bone development including the differentiation and lineage allocation of skeletal progenitor cells to chondrocytes, osteoblasts, or marrow adipocytes. As the major ECM component in mineralized tissues, collagen has instructive as well as structural roles during bone development and is required for bone cell differentiation. Cells sense their extracellular environment using specific cell surface receptors. For many years, specific β1 integrins were considered the main collagen receptors in bone, but, more recently, the important role of a second, more primordial collagen receptor family, the discoidin domain receptors, has become apparent. This review will specifically focus on the roles of discoidin domain receptors in mineralized tissue development as well as related functions in abnormal bone formation, regeneration and metabolism.
Collapse
Affiliation(s)
- Renny T. Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Shawn A. Hallett
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| | - Chunxi Ge
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, United States
| |
Collapse
|
50
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|