1
|
Zuo YB, Wen ZJ, Cheng MD, Jia DD, Zhang YF, Yang HY, Xu HM, Xin H, Zhang YF. The pro-atherogenic effects and the underlying mechanisms of chronic bisphenol S (BPS) exposure in apolipoprotein E-deficient mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117133. [PMID: 39342757 DOI: 10.1016/j.ecoenv.2024.117133] [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: 04/30/2024] [Revised: 07/23/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Atherosclerosis (AS) and its related cardiovascular diseases (CVDs) remain the most frequent cause of morbidity and mortality worldwide. Researches showed that bisphenol A (BPA) exposure might exacerbate AS progression. However, as an analogue of BPA, little is known about the cardiovascular toxicity of bisphenol S (BPS), especially whether BPS exposure has the pro-atherogenic effects in mammals is still unknown. Here, we firstly constructed an apolipoprotein E knockout (ApoE-/-) mouse model and cultured cells to investigate the risk of BPS on AS and explore the underlying mechanisms. Results showed that prolonged exposure to 50 μg/kg body weight (bw)/day BPS indeed aggravated AS lesions both in the en face aortas and aortic sinuses of ApoE-/- mice. Moreover, BPS were found to be implicated in the AS pathological process: 1) stimulates adhesion molecule expression to promote monocyte-endothelial cells (ECs) adhesion with 3.6 times more than the control group in vivo; 2) increases the distribution of vascular smooth muscle cells (VSMCs) with 9.3 times more than the control group in vivo, possibly through the migration of VSMCs; and 3) induces an inflammatory response by increasing the number of macrophages (MACs), with 3.7 times more than the control group in vivo, and the release of inflammatory mediators. Furthermore, we have identified eight significant AS-related genes induced by BPS, including angiopoietin-like protein 7 (Angptl17) and lipocalin-2 (Lcn2) in ECs; matrix metalloproteinase 9 (Mmp13), secreted phosphoprotein 1 (Spp1), and collagen type II alpha 1 (Col2a1) in VSMCs; and kininogen 1 (Kng1), integrin alpha X (Itgax), and MAC-expressed gene 1 (Mpeg1) in MACs. Overall, this study firstly found BPS exposure could exacerbate mammalian AS and might also provide a theoretical basis for elucidating BPS and its analogues induced AS and related CVDs.
Collapse
Affiliation(s)
- Ying-Bing Zuo
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China; Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China
| | - Zeng-Jin Wen
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Meng-Die Cheng
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China; Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China
| | - Dong-Dong Jia
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Yi-Fei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hong-Yu Yang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China
| | - Hai-Ming Xu
- School of Public Health, Ningxia Medical University, Yinchuan, Ningxia 750004, China.
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao University, 16 Jiangsu Road, Qingdao 266000, China.
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Deng Zhou Road 38, Qingdao 266021, China.
| |
Collapse
|
2
|
Wang Z, Gui Z, Zhang L, Wang Z. Advances in the mechanisms of vascular calcification in chronic kidney disease. J Cell Physiol 2024:e31464. [PMID: 39392232 DOI: 10.1002/jcp.31464] [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: 05/15/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Vascular calcification (VC) is common in patients with advanced chronic kidney disease (CKD).A series of factors, such as calcium and phosphorus metabolism disorders, uremic toxin accumulation, inflammation and oxidative stress and cellular senescence, cause osteoblast-like differentiation of vascular smooth muscle cells, secretion of extracellular vesicles, and imbalance of calcium regulatory factors, which together promote the development of VC in CKD. Recent advances in epigenetics have provided better tools for the investigation of VC etiology and new approaches for finding more accurate biomarkers. These advances have not only deepened our understanding of the pathophysiological mechanisms of VC in CKD, but also provided valuable clues for the optimization of clinical predictors and the exploration of potential therapeutic targets. The aim of this article is to provide a comprehensive overview of the pathogenesis of CKD VC, especially the new advances made in recent years, including the various key factors mentioned above. Through the comprehensive analysis, we expect to provide a solid theoretical foundation and research direction for future studies targeting the specific mechanisms of CKD VC, the establishment of clinical predictive indicators and the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Ziyang Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Zebin Gui
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| | - Lirong Zhang
- Department of Radiology, Affliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Institute of Cardiovascular Diseases, Jiangsu University, Zhenjiang, China
| |
Collapse
|
3
|
Elmarasi M, Elmakaty I, Elsayed B, Elsayed A, Zein JA, Boudaka A, Eid AH. Phenotypic switching of vascular smooth muscle cells in atherosclerosis, hypertension, and aortic dissection. J Cell Physiol 2024; 239:e31200. [PMID: 38291732 DOI: 10.1002/jcp.31200] [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: 08/21/2023] [Revised: 12/12/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Vascular smooth muscle cells (VSMCs) play a critical role in regulating vasotone, and their phenotypic plasticity is a key contributor to the pathogenesis of various vascular diseases. Two main VSMC phenotypes have been well described: contractile and synthetic. Contractile VSMCs are typically found in the tunica media of the vessel wall, and are responsible for regulating vascular tone and diameter. Synthetic VSMCs, on the other hand, are typically found in the tunica intima and adventitia, and are involved in vascular repair and remodeling. Switching between contractile and synthetic phenotypes occurs in response to various insults and stimuli, such as injury or inflammation, and this allows VSMCs to adapt to changing environmental cues and regulate vascular tone, growth, and repair. Furthermore, VSMCs can also switch to osteoblast-like and chondrocyte-like cell phenotypes, which may contribute to vascular calcification and other pathological processes like the formation of atherosclerotic plaques. This provides discusses the mechanisms that regulate VSMC phenotypic switching and its role in the development of vascular diseases. A better understanding of these processes is essential for the development of effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mohamed Elmarasi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim Elmakaty
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Basel Elsayed
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdelrahman Elsayed
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
4
|
Shen J, Ju D, Wu S, Zhao J, Pham L, Ponce A, Yang M, Li HJ, Zhang K, Yang Z, Xie Y, Li L. SM22α deficiency: promoting vascular fibrosis via SRF-SMAD3-mediated activation of Col1a2 transcription following arterial injury. RESEARCH SQUARE 2024:rs.3.rs-3941602. [PMID: 38464061 PMCID: PMC10925461 DOI: 10.21203/rs.3.rs-3941602/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Vascular fibrosis, characterized by increased Type I collagen expression, significantly contributes to vascular remodeling. Our previous studies show that disrupting the expression of SM22α (aka SM22, Tagln) induces extensive vascular remodeling following arterial injury, involving oxidative stress, inflammation, and chondrogenesis within the vessel wall. This study aims to investigate the molecular mechanisms underlying the transcription of Col1a2 , a key fibrotic extracellular matrix marker. We observed upregulation of COL1A2 in the arterial wall of Sm22 -/- mice following carotid injury. Bioinformatics and molecular analyses reveal that Col1a2 transcription depends on a CArG box in the promoter, activated synergistically by SRF and SMAD3. Notably, we detected enhanced nuclear translocation of both SRF and SMAD3 in the smooth muscle cells of the injured carotid artery in Sm22 -/- mice. These findings demonstrate that SM22 deficiency regulates vascular fibrosis through the interaction of SRF and the SMAD3-mediated canonical TGF-β1 signal pathway, suggesting SM22α as a potential therapeutic target for preventing vascular fibrosis.
Collapse
|
5
|
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
|
6
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
7
|
Bickel MA, Sherry DM, Bullen EC, Vance ML, Jones KL, Howard EW, Conley SM. Microvascular smooth muscle cells exhibit divergent phenotypic switching responses to platelet-derived growth factor and insulin-like growth factor 1. Microvasc Res 2024; 151:104609. [PMID: 37716411 PMCID: PMC10842624 DOI: 10.1016/j.mvr.2023.104609] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Vascular smooth muscle cell (VSMC) phenotypic switching is critical for normal vessel formation, vascular stability, and healthy brain aging. Phenotypic switching is regulated by mediators including platelet derived growth factor (PDGF)-BB, insulin-like growth factor (IGF-1), as well as transforming growth factor-β (TGF-β) and endothelin-1 (ET-1), but much about the role of these factors in microvascular VSMCs remains unclear. METHODS We used primary rat microvascular VSMCs to explore PDGF-BB- and IGF-1-induced phenotypic switching. RESULTS PDGF-BB induced an early proliferative response, followed by formation of polarized leader cells and rapid, directionally coordinated migration. In contrast, IGF-1 induced cell hypertrophy, and only a small degree of migration by unpolarized cells. TGF-β and ET-1 selectively inhibit PDGF-BB-induced VSMC migration primarily by repressing migratory polarization and formation of leader cells. Contractile genes were downregulated by both growth factors, while other genes were differentially regulated by PDGF-BB and IGF-1. CONCLUSIONS These studies indicate that PDGF-BB and IGF-1 stimulate different types of microvascular VSMC phenotypic switching characterized by different modes of cell migration. Our studies are consistent with a chronic vasoprotective role for IGF-1 in VSMCs in the microvasculature while PDGF is more involved in VSMC proliferation and migration in response to acute activities such as neovascularization. Better understanding of the nuances of the phenotypic switching induced by these growth factors is important for our understanding of a variety of microvascular diseases.
Collapse
Affiliation(s)
- Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - David M Sherry
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Ken L Jones
- Bioinformatic Solutions, LLC, Sheridan, WY 82801, United States of America
| | - Eric W Howard
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States of America.
| |
Collapse
|
8
|
Arévalo Martínez M, Ritsvall O, Bastrup JA, Celik S, Jakobsson G, Daoud F, Winqvist C, Aspberg A, Rippe C, Maegdefessel L, Schiopu A, Jepps TA, Holmberg J, Swärd K, Albinsson S. Vascular smooth muscle-specific YAP/TAZ deletion triggers aneurysm development in mouse aorta. JCI Insight 2023; 8:e170845. [PMID: 37561588 PMCID: PMC10544211 DOI: 10.1172/jci.insight.170845] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Inadequate adaption to mechanical forces, including blood pressure, contributes to development of arterial aneurysms. Recent studies have pointed to a mechanoprotective role of YAP and TAZ in vascular smooth muscle cells (SMCs). Here, we identified reduced expression of YAP1 in human aortic aneurysms. Vascular SMC-specific knockouts (KOs) of YAP/TAZ were thus generated using the integrin α8-Cre (Itga8-Cre) mouse model (i8-YT-KO). i8-YT-KO mice spontaneously developed aneurysms in the abdominal aorta within 2 weeks of KO induction and in smaller arteries at later times. The vascular specificity of Itga8-Cre circumvented gastrointestinal effects. Aortic aneurysms were characterized by elastin disarray, SMC apoptosis, and accumulation of proteoglycans and immune cell populations. RNA sequencing, proteomics, and myography demonstrated decreased contractile differentiation of SMCs and impaired vascular contractility. This associated with partial loss of myocardin expression, reduced blood pressure, and edema. Mediators in the inflammatory cGAS/STING pathway were increased. A sizeable increase in SOX9, along with several direct target genes, including aggrecan (Acan), contributed to proteoglycan accumulation. This was the earliest detectable change, occurring 3 days after KO induction and before the proinflammatory transition. In conclusion, Itga8-Cre deletion of YAP and TAZ represents a rapid and spontaneous aneurysm model that recapitulates features of human abdominal aortic aneurysms.
Collapse
Affiliation(s)
| | - Olivia Ritsvall
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Armstrong Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Selvi Celik
- Molecular Cardiology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Fatima Daoud
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Christopher Winqvist
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Anders Aspberg
- Rheumatology and Molecular Skeletal Biology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden, and
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Alexandru Schiopu
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Internal Medicine, Skåne University Hospital Lund, Lund, Sweden, and
- Nicolae Simionescu Institute of Cellular Biology and Pathology, Bucharest, Romania
| | - Thomas A. Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
9
|
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: 30] [Impact Index Per Article: 30.0] [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
|
10
|
Xiang X, He J, Zhang W, He Q, Liu Y. Coronary artery calcification in patients with advanced chronic kidney disease. BMC Cardiovasc Disord 2022; 22:453. [PMID: 36309659 PMCID: PMC9618197 DOI: 10.1186/s12872-022-02879-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Cardiovascular disease (CVD) is associated with higher morbidity and mortality rates in patients with chronic kidney disease (CKD). Studies have shown that vascular calcification is a major predictor of CVD. Vascular calcification in the CKD population is associated with various risk factors, and changes in bone and mineral metabolism have been linked to an increased risk of atherosclerosis. Therefore, we aimed to investigate the correlation between vascular calcification and bone metabolism, which is necessary to improve the survival and prognosis of patients with CKD. Methods We included 146 patients with CKD who received coronary artery calcification (CAC) scores at our hospital from May 2017 to November 2018. Spearman rank correlation analysis, Mann–Whitney U test, and Kaplan–Meier method were used to analyze laboratory data and all-cause mortality. Results In the 146 patients, chronic glomerulonephritis accounted for the most common cause of CKD, at approximately 39.0%. Spearman rank correlation analysis on the factors influencing vascular calcification in patients with CKD showed that CAC score was significantly and positively correlated with C-reactive protein, N-terminal/midregion osteocalcin (N-MID), N-terminal peptide of type 1 procollagen (P1NP), β-cross-linked C-telopeptide of type 1 collagen (β-CTx), and parathyroid hormone (P = 0.0423, P = 0.0432, P = 0.0235, P = 0.0061, P < 0.0001, respectively). Serum calcium levels were positively correlated with N-MID, P1NP, β-CTx, and iPTH (r = 0.19, r = 0.24, r = 0.21, r = 0.21, respectively), and serum phosphorus levels were positively correlated with N-MID, P1NP, β-CTx, and iPTH (r = 0.50, r = 0.37, r = 0.50, r = 0.55, respectively). However, no difference was found in CVC scores among patients with CKD in different stages and receiving different treatments. In the Kaplan–Meier analysis of all-cause hospitalization and mortality rates, patients with CAC > 400 had a higher risk. Conclusion We found that the primary cause of CKD is glomerulonephritis, and the CAC score is positively correlated with inflammatory and bone metabolism markers, with a higher risk of all-cause mortality and cardiovascular hospitalization when the CAC score is greater than 400.
Collapse
|
11
|
Yu Q, Liu JX, Zheng X, Yan X, Zhao P, Yin C, Li W, Song Z. Sox9 mediates autophagy-dependent vascular smooth muscle cell phenotypic modulation and transplant arteriosclerosis. iScience 2022; 25:105161. [PMID: 36204267 PMCID: PMC9531173 DOI: 10.1016/j.isci.2022.105161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 08/04/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Qihong Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan 430030, China
| | - Jin-Xin Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueke Yan
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peng Zhao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chuanzheng Yin
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Li
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Corresponding author
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Corresponding author
| |
Collapse
|
12
|
Wen Y, Kong Y, Cao G, Xu Y, Zhang C, Zhang J, Xiao P, Wang Y. Di-n-butyl phthalate regulates vascular smooth muscle cells phenotypic switching by MiR-139-5p-MYOCD pathways. Toxicology 2022; 477:153279. [PMID: 35926758 DOI: 10.1016/j.tox.2022.153279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/20/2022] [Accepted: 07/30/2022] [Indexed: 11/17/2022]
Abstract
Di-n-butyl phthalate (DBP) is ubiquitous in environment and has been detected in almost all human bodies. Few data could be found about the effects of DBP on cardiovascular system, though its reproductive toxicities have been studied extensively. This study aimed to explore effects of DBP on phenotypic switching of vascular smooth muscle cells (VSMCs), an essential step during the formation of atherosclerosis (AS). A7r5 cells were employed and exposed to various levels of DBP (10-9, 10-8, 10-7, 10-6, and 10-5 M) or DMSO as control. CCK-8 assay was used to detect the effects of DBP on cell viability. Expressions of mRNA/miRNAs and proteins were measured by qRT-PCR and western blotting, respectively. Bioinformatic analysis and dual-luciferase reporter assay were used to analyze the combination between miR-139-5p and Myocardin (MYOCD). Results revealed that DBP at 10-7 M prompted phenotypic switching from contractile to synthetic of VSMCs by inhibiting contractile VSMCs marker genes via suppressing the expression of MYOCD. Moreover, miR-139c-5p directly targeted MYOCD 3'UTR and modulated MYOCD expression. Besides, DBP inhibited the expression of MYOCD and VSMCs marker genes by upregulating miR-139-5p. Collectively, these data suggested that DBP could promote the phenotypic switching from contractile to synthetic of VSMCs in A7r5 cells through miR-139-5p-MYOCD.
Collapse
Affiliation(s)
- Yun Wen
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yi Kong
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Guofa Cao
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Yuan Xu
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Chengxiang Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Jingshu Zhang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Pingxi Xiao
- The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yubang Wang
- The Key Laboratory of Modern Toxicology, Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China; The Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Safety Assessment and Research Center for Drug, Pesticide and Veterinary Drug of Jiangsu Province, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
13
|
Zhou W, Bai Y, Chen J, Li H, Zhang B, Liu H. Revealing the Critical Regulators of Modulated Smooth Muscle Cells in Atherosclerosis in Mice. Front Genet 2022; 13:900358. [PMID: 35677564 PMCID: PMC9168464 DOI: 10.3389/fgene.2022.900358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/15/2022] [Indexed: 01/23/2023] Open
Abstract
Background: There are still residual risks for atherosclerosis (AS)-associated cardiovascular diseases to be resolved. Considering the vital role of phenotypic switching of smooth muscle cells (SMCs) in AS, especially in calcification, targeting SMC phenotypic modulation holds great promise for clinical implications. Methods: To perform an unbiased and systematic analysis of the molecular regulatory mechanism of phenotypic switching of SMCs during AS in mice, we searched and included several publicly available single-cell datasets from the GEO database, resulting in an inclusion of more than 80,000 cells. Algorithms implemented in the Seurat package were used for cell clustering and cell atlas depiction. The pySCENIC and SCENIC packages were used to identify master regulators of interested cell groups. Monocle2 was used to perform pseudotime analysis. clusterProfiler was used for Gene Ontology enrichment analysis. Results: After dimensionality reduction and clustering, reliable annotation was performed. Comparative analysis between cells from normal artery and AS lesions revealed that three clusters emerged as AS progression, designated as mSMC1, mSMC2, and mSMC3. Transcriptional and functional enrichment analysis established a continuous transitional mode of SMCs’ transdifferentiation to mSMCs, which is further supported by pseudotime analysis. A total of 237 regulons were identified with varying activity scores across cell types. A potential core regulatory network was constructed for SMC and mSMC subtypes. In addition, module analysis revealed a coordinate regulatory mode of regulons for a specific cell type. Intriguingly, consistent with gain of ossification-related transcriptional and functional characteristics, a corresponding small set of regulators contributing to osteochondral reprogramming was identified in mSMC3, including Dlx5, Sox9, and Runx2. Conclusion: Gene regulatory network inference indicates a hierarchical organization of regulatory modules that work together in fine-tuning cellular states. The analysis here provides a valuable resource that can provide guidance for subsequent biological experiments.
Collapse
Affiliation(s)
- Wenli Zhou
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yongyi Bai
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianqiao Chen
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huiying Li
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Baohua Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Health Care, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Hongbin Liu,
| |
Collapse
|
14
|
H3K4 di-methylation governs smooth muscle lineage identity and promotes vascular homeostasis by restraining plasticity. Dev Cell 2021; 56:2765-2782.e10. [PMID: 34582749 PMCID: PMC8567421 DOI: 10.1016/j.devcel.2021.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 07/09/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022]
Abstract
Epigenetic mechanisms contribute to the regulation of cell differentiation and function. Vascular smooth muscle cells (SMCs) are specialized contractile cells that retain phenotypic plasticity even after differentiation. Here, by performing selective demethylation of histone H3 lysine 4 di-methylation (H3K4me2) at SMC-specific genes, we uncovered that H3K4me2 governs SMC lineage identity. Removal of H3K4me2 via selective editing in cultured vascular SMCs and in murine arterial vasculature led to loss of differentiation and reduced contractility due to impaired recruitment of the DNA methylcytosine dioxygenase TET2. H3K4me2 editing altered SMC adaptative capacities during vascular remodeling due to loss of miR-145 expression. Finally, H3K4me2 editing induced a profound alteration of SMC lineage identity by redistributing H3K4me2 toward genes associated with stemness and developmental programs, thus exacerbating plasticity. Our studies identify the H3K4me2-TET2-miR145 axis as a central epigenetic memory mechanism controlling cell identity and function, whose alteration could contribute to various pathophysiological processes.
Collapse
|
15
|
Seime T, Akbulut AC, Liljeqvist ML, Siika A, Jin H, Winski G, van Gorp RH, Karlöf E, Lengquist M, Buckler AJ, Kronqvist M, Waring OJ, Lindeman JHN, Biessen EAL, Maegdefessel L, Razuvaev A, Schurgers LJ, Hedin U, Matic L. Proteoglycan 4 Modulates Osteogenic Smooth Muscle Cell Differentiation during Vascular Remodeling and Intimal Calcification. Cells 2021; 10:1276. [PMID: 34063989 PMCID: PMC8224064 DOI: 10.3390/cells10061276] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 01/02/2023] Open
Abstract
Calcification is a prominent feature of late-stage atherosclerosis, but the mechanisms driving this process are unclear. Using a biobank of carotid endarterectomies, we recently showed that Proteoglycan 4 (PRG4) is a key molecular signature of calcified plaques, expressed in smooth muscle cell (SMC) rich regions. Here, we aimed to unravel the PRG4 role in vascular remodeling and intimal calcification. PRG4 expression in human carotid endarterectomies correlated with calcification assessed by preoperative computed tomographies. PRG4 localized to SMCs in early intimal thickening, while in advanced lesions it was found in the extracellular matrix, surrounding macro-calcifications. In experimental models, Prg4 was upregulated in SMCs from partially ligated ApoE-/- mice and rat carotid intimal hyperplasia, correlating with osteogenic markers and TGFb1. Furthermore, PRG4 was enriched in cells positive for chondrogenic marker SOX9 and around plaque calcifications in ApoE-/- mice on warfarin. In vitro, PRG4 was induced in SMCs by IFNg, TGFb1 and calcifying medium, while SMC markers were repressed under calcifying conditions. Silencing experiments showed that PRG4 expression was driven by transcription factors SMAD3 and SOX9. Functionally, the addition of recombinant human PRG4 increased ectopic SMC calcification, while arresting cell migration and proliferation. Mechanistically, it suppressed endogenous PRG4, SMAD3 and SOX9, and restored SMC markers' expression. PRG4 modulates SMC function and osteogenic phenotype during intimal remodeling and macro-calcification in response to TGFb1 signaling, SMAD3 and SOX9 activation. The effects of PRG4 on SMC phenotype and calcification suggest its role in atherosclerotic plaque stability, warranting further investigations.
Collapse
Affiliation(s)
- Till Seime
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Asim Cengiz Akbulut
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
| | - Moritz Lindquist Liljeqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Antti Siika
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Hong Jin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
| | - Greg Winski
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
| | - Rick H. van Gorp
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
| | - Eva Karlöf
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Mariette Lengquist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Andrew J. Buckler
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Malin Kronqvist
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Olivia J. Waring
- Department of Pathology, CARIM, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands; (O.J.W.); (E.A.L.B.)
| | - Jan H. N. Lindeman
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Erik A. L. Biessen
- Department of Pathology, CARIM, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands; (O.J.W.); (E.A.L.B.)
| | - Lars Maegdefessel
- Department of Medicine, Karolinska Institutet, 17164 Stockholm, Sweden; (G.W.); (L.M.)
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technische Universität München, 81679 Munich, Germany
| | - Anton Razuvaev
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Leon J. Schurgers
- Department of Biochemistry, CARIM, Maastricht University, 6229 ER Maastricht, The Netherlands; (A.C.A.); (R.H.v.G.); (L.J.S.)
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, 52062 Aachen, Germany
| | - Ulf Hedin
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| | - Ljubica Matic
- Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, 17164 Stockholm, Sweden; (T.S.); (M.L.L.); (A.S.); (H.J.); (E.K.); (M.L.); (A.J.B.); (M.K.); (A.R.); (U.H.)
| |
Collapse
|
16
|
Jiang W, Zhang Z, Li Y, Chen C, Yang H, Lin Q, Hu M, Qin X. The Cell Origin and Role of Osteoclastogenesis and Osteoblastogenesis in Vascular Calcification. Front Cardiovasc Med 2021; 8:639740. [PMID: 33969008 PMCID: PMC8102685 DOI: 10.3389/fcvm.2021.639740] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/24/2021] [Indexed: 02/01/2023] Open
Abstract
Arterial calcification refers to the abnormal deposition of calcium salts in the arterial wall, which results in vessel lumen stenosis and vascular remodeling. Studies increasingly show that arterial calcification is a cell mediated, reversible and active regulated process similar to physiological bone mineralization. The osteoblasts and chondrocytes-like cells are present in large numbers in the calcified lesions, and express osteogenic transcription factor and bone matrix proteins that are known to initiate and promote arterial calcification. In addition, osteoclast-like cells have also been detected in calcified arterial walls wherein they possibly inhibit vascular calcification, similar to the catabolic process of bone mineral resorption. Therefore, tilting the balance between osteoblast-like and osteoclast-like cells to the latter maybe a promising therapeutic strategy against vascular calcification. In this review, we have summarized the current findings on the origin and functions of osteoblast-like and osteoclast-like cells in the development and progression of vascular progression, and explored novel therapeutic possibilities.
Collapse
Affiliation(s)
- Wenhong Jiang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhanman Zhang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yaodong Li
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chuanzhen Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Han Yang
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiuning Lin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ming Hu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
17
|
Zheng JP, He X, Liu F, Yin S, Wu S, Yang M, Zhao J, Dai X, Jiang H, Yu L, Yin Q, Ju D, Li C, Lipovich L, Xie Y, Zhang K, Li HJ, Zhou J, Li L. YY1 directly interacts with myocardin to repress the triad myocardin/SRF/CArG box-mediated smooth muscle gene transcription during smooth muscle phenotypic modulation. Sci Rep 2020; 10:21781. [PMID: 33311559 PMCID: PMC7732823 DOI: 10.1038/s41598-020-78544-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Yin Yang 1 (YY1) regulates gene transcription in a variety of biological processes. In this study, we aim to determine the role of YY1 in vascular smooth muscle cell (VSMC) phenotypic modulation both in vivo and in vitro. Here we show that vascular injury in rodent carotid arteries induces YY1 expression along with reduced expression of smooth muscle differentiation markers in the carotids. Consistent with this finding, YY1 expression is induced in differentiated VSMCs in response to serum stimulation. To determine the underlying molecular mechanisms, we found that YY1 suppresses the transcription of CArG box-dependent SMC-specific genes including SM22α, SMα-actin and SMMHC. Interestingly, YY1 suppresses the transcriptional activity of the SM22α promoter by hindering the binding of serum response factor (SRF) to the proximal CArG box. YY1 also suppresses the transcription and the transactivation of myocardin (MYOCD), a master regulator for SMC-specific gene transcription by binding to SRF to form the MYOCD/SRF/CArG box triad (known as the ternary complex). Mechanistically, YY1 directly interacts with MYOCD to competitively displace MYOCD from SRF. This is the first evidence showing that YY1 inhibits SMC differentiation by directly targeting MYOCD. These findings provide new mechanistic insights into the regulatory mechanisms that govern SMC phenotypic modulation in the pathogenesis of vascular diseases.
Collapse
Affiliation(s)
- Jian-Pu Zheng
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Xiangqin He
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
- The Institute of Translational Medicine, Nanchang University, Nanchang, 330031, Jiangxi, China
| | - Fang Liu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Shuping Yin
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Shichao Wu
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Maozhou Yang
- Bone and Joint Center, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Jiawei Zhao
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Xiaohua Dai
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Hong Jiang
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Luyi Yu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Qin Yin
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Donghong Ju
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Claire Li
- Center for Molecular Medicine and Genetics, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Leonard Lipovich
- Center for Molecular Medicine and Genetics, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Youming Xie
- Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA
| | - Hui J Li
- Department of Medicine, University of Massachusetts, Worcester, MA, 01655, USA
| | - Jiliang Zhou
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Li Li
- Department of Internal Medicine, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA.
- Cardiovascular Research Institute, Wayne State University, 421 E. Canfield Ave. #2146, Detroit, MI, 48201, USA.
| |
Collapse
|
18
|
Cai W, Zhou W, Han Z, Lei J, Zhuang J, Zhu P, Wu X, Yuan W. Master regulator genes and their impact on major diseases. PeerJ 2020; 8:e9952. [PMID: 33083114 PMCID: PMC7546222 DOI: 10.7717/peerj.9952] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/25/2020] [Indexed: 01/10/2023] Open
Abstract
Master regulator genes (MRGs) have become a hot topic in recent decades. They not only affect the development of tissue and organ systems but also play a role in other signal pathways by regulating additional MRGs. Because a MRG can regulate the concurrent expression of several genes, its mutation often leads to major diseases. Moreover, the occurrence of many tumors and cardiovascular and nervous system diseases are closely related to MRG changes. With the development in omics technology, an increasing amount of investigations will be directed toward MRGs because their regulation involves all aspects of an organism’s development. This review focuses on the definition and classification of MRGs as well as their influence on disease regulation.
Collapse
Affiliation(s)
- Wanwan Cai
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Wanbang Zhou
- College of Physical Education, Hunan Normal University, Changsha, Hunan, China
| | - Zhe Han
- University of Maryland School of Medicine, Center for Precision Disease Modeling, Baltimore, MD, USA
| | - Junrong Lei
- College of Physical Education, Hunan Normal University, Changsha, Hunan, China
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Department of Cardiac Surgery, Guangzhou, Guangdong, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Department of Cardiac Surgery, Guangzhou, Guangdong, China
| | - Xiushan Wu
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Wuzhou Yuan
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
19
|
Zhang S, Hamid MR, Wang T, Liao J, Wen L, Zhou Y, Wei P, Zou X, Chen G, Chen J, Zhou G. RSK-3 promotes cartilage regeneration via interacting with rpS6 in cartilage stem/progenitor cells. Theranostics 2020; 10:6915-6927. [PMID: 32550912 PMCID: PMC7295041 DOI: 10.7150/thno.44875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Cartilage stem/progenitor cells (CSPC) are a promising cellular source to promote endogenous cartilage regeneration in osteoarthritis (OA). Our previous work indicates that ribosomal s6 kinase 3 (RSK-3) is a target of 4-aminobiphenyl, a chemical enhancing CSPC-mediated cartilage repair in OA. However, the primary function and mechanism of RSK-3 in CSPC-mediated cartilage pathobiology remain undefined. Methods: We systematically assessed the association of RSK-3 with OA in three mouse strains with varying susceptibility to OA (MRL/MpJ>CBA>STR/Ort), and also RSK-3-/- mice. Bioinformatic analysis was used to identify the possible mechanism of RSK-3 affecting CSPC, which was further verified in OA mice and CSPC with varying RSK-3 expression induced by chemicals or gene modification. Results: We demonstrated that the level of RSK-3 in cartilage was positively correlated with cartilage repair capacities in three mouse strains (MRL/MpJ>CBA>STR/Ort). Enhanced RSK-3 expression by 4-aminobiphenyl markedly attenuated cartilage injury in OA mice and inhibition or deficiency of RSK-3 expression, on the other hand, significantly aggravated cartilage damage. Transcriptional profiling of CSPC from mice suggested the potential role of RSK-3 in modulating cell proliferation. It was further shown that the in vivo and in vitro manipulation of the RSK-3 expression indeed affected the CSPC proliferation. Mechanistically, ribosomal protein S6 (rpS6) was activated by RSK-3 to accelerate CSPC growth. Conclusion: RSK-3 is identified as a key regulator to enhance cartilage repair, at least partly by regulating the functionality of the cartilage-resident stem/progenitor cells.
Collapse
|
20
|
Mallis P, Papapanagiotou A, Katsimpoulas M, Kostakis A, Siasos G, Kassi E, Stavropoulos-Giokas C, Michalopoulos E. Efficient differentiation of vascular smooth muscle cells from Wharton’s Jelly mesenchymal stromal cells using human platelet lysate: A potential cell source for small blood vessel engineering. World J Stem Cells 2020; 12:203-221. [PMID: 32266052 PMCID: PMC7118289 DOI: 10.4252/wjsc.v12.i3.203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/17/2020] [Accepted: 02/01/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The development of fully functional small diameter vascular grafts requires both a properly defined vessel conduit and tissue-specific cellular populations. Mesenchymal stromal cells (MSCs) derived from the Wharton’s Jelly (WJ) tissue can be used as a source for obtaining vascular smooth muscle cells (VSMCs), while the human umbilical arteries (hUAs) can serve as a scaffold for blood vessel engineering.
AIM To develop VSMCs from WJ-MSCs utilizing umbilical cord blood platelet lysate.
METHODS WJ-MSCs were isolated and expanded until passage (P) 4. WJ-MSCs were properly defined according to the criteria of the International Society for Cell and Gene Therapy. Then, these cells were differentiated into VSMCs with the use of platelet lysate from umbilical cord blood in combination with ascorbic acid, followed by evaluation at the gene and protein levels. Specifically, gene expression profile analysis of VSMCs for ACTA2, MYH11, TGLN, MYOCD, SOX9, NANOG homeobox, OCT4 and GAPDH, was performed. In addition, immunofluorescence against ACTA2 and MYH11 in combination with DAPI staining was also performed in VSMCs. HUAs were decellularized and served as scaffolds for possible repopulation by VSMCs. Histological and biochemical analyses were performed in repopulated hUAs.
RESULTS WJ-MSCs exhibited fibroblastic morphology, successfully differentiating into “osteocytes”, “adipocytes” and “chondrocytes”, and were characterized by positive expression (> 90%) of CD90, CD73 and CD105. In addition, WJ-MSCs were successfully differentiated into VSMCs with the proposed differentiation protocol. VSMCs successfully expressed ACTA2, MYH11, MYOCD, TGLN and SOX9. Immunofluorescence results indicated the expression of ACTA2 and MYH11 in VSMCs. In order to determine the functionality of VSMCs, hUAs were isolated and decellularized. Based on histological analysis, decellularized hUAs were free of any cellular or nuclear materials, while their extracellular matrix retained intact. Then, repopulation of decellularized hUAs with VSMCs was performed for 3 wk. Decellularized hUAs were repopulated efficiently by the VSMCs. Biochemical analysis revealed the increase of total hydroyproline and sGAG contents in repopulated hUAs with VSMCs. Specifically, total hydroxyproline and sGAG content after the 1st, 2nd and 3rd wk was 71 ± 10, 74 ± 9 and 86 ± 8 μg hydroxyproline/mg of dry tissue weight and 2 ± 1, 3 ± 1 and 3 ± 1 μg sGAG/mg of dry tissue weight, respectively. Statistically significant differences were observed between all study groups (P < 0.05).
CONCLUSION VSMCs were successfully obtained from WJ-MSCs with the proposed differentiation protocol. Furthermore, hUAs were efficiently repopulated by VSMCs. Differentiated VSMCs from WJ-MSCs could provide an alternative source of cells for vascular tissue engineering.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Aggeliki Papapanagiotou
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
| | - Michalis Katsimpoulas
- Center of Experimental Surgery, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Alkiviadis Kostakis
- Center of Experimental Surgery, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| | - Gerasimos Siasos
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
- First Department of Cardiology, “Hippokration” Hospital, University of Athens Medical School, Athens 15231, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian Univesity of Athens, Athens 15772, Greece
- First Department of Internal Medicine, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | | | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, Athens 11527, Greece
| |
Collapse
|
21
|
Dai L, Qureshi AR, Witasp A, Lindholm B, Stenvinkel P. Early Vascular Ageing and Cellular Senescence in Chronic Kidney Disease. Comput Struct Biotechnol J 2019; 17:721-729. [PMID: 31303976 PMCID: PMC6603301 DOI: 10.1016/j.csbj.2019.06.015] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 06/08/2019] [Accepted: 06/11/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic kidney disease (CKD) is a clinical model of premature ageing characterized by progressive vascular disease, systemic inflammation, muscle wasting and frailty. The predominant early vascular ageing (EVA) process mediated by medial vascular calcification (VC) results in a marked discrepancy between chronological and biological vascular age in CKD. Though the exact underlying mechanisms of VC and EVA are not fully elucidated, accumulating evidence indicates that cellular senescence - and subsequent chronic inflammation through the senescence-associated secretary phenotype (SASP) - plays a fundamental role in its initiation and progression. In this review, we discuss the pathophysiological links between senescence and the EVA process in CKD, with focus on cellular senescence and media VC, and potential anti-ageing therapeutic strategies of senolytic drugs targeting cellular senescence and EVA in CKD.
Collapse
Affiliation(s)
| | | | | | | | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
22
|
Voelkl J, Lang F, Eckardt KU, Amann K, Kuro-O M, Pasch A, Pieske B, Alesutan I. Signaling pathways involved in vascular smooth muscle cell calcification during hyperphosphatemia. Cell Mol Life Sci 2019; 76:2077-2091. [PMID: 30887097 PMCID: PMC6502780 DOI: 10.1007/s00018-019-03054-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 02/06/2023]
Abstract
Medial vascular calcification has emerged as a putative key factor contributing to the excessive cardiovascular mortality of patients with chronic kidney disease (CKD). Hyperphosphatemia is considered a decisive determinant of vascular calcification in CKD. A critical role in initiation and progression of vascular calcification during elevated phosphate conditions is attributed to vascular smooth muscle cells (VSMCs), which are able to change their phenotype into osteo-/chondroblasts-like cells. These transdifferentiated VSMCs actively promote calcification in the medial layer of the arteries by producing a local pro-calcifying environment as well as nidus sites for precipitation of calcium and phosphate and growth of calcium phosphate crystals. Elevated extracellular phosphate induces osteo-/chondrogenic transdifferentiation of VSMCs through complex intracellular signaling pathways, which are still incompletely understood. The present review addresses critical intracellular pathways controlling osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification during hyperphosphatemia. Elucidating these pathways holds a significant promise to open novel therapeutic opportunities counteracting the progression of vascular calcification in CKD.
Collapse
MESH Headings
- Animals
- Calcium Phosphates/chemistry
- Calcium Phosphates/metabolism
- Cell Transdifferentiation
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Gene Expression Regulation
- Humans
- Hyperphosphatemia/complications
- Hyperphosphatemia/genetics
- Hyperphosphatemia/metabolism
- Hyperphosphatemia/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Osteoblasts/metabolism
- Osteoblasts/pathology
- RANK Ligand/genetics
- RANK Ligand/metabolism
- Receptor Activator of Nuclear Factor-kappa B/genetics
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Renal Insufficiency, Chronic/complications
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/pathology
- Signal Transduction
- Vascular Calcification/complications
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
Collapse
Affiliation(s)
- Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria.
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany.
| | - Florian Lang
- Department of Physiology I, Eberhard-Karls University, Wilhelmstr. 56, 72076, Tübingen, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Augustenburgerplatz 1, 13353, Berlin, Germany
| | - Kerstin Amann
- Department of Nephropathology, Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Makoto Kuro-O
- Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Andreas Pasch
- Calciscon AG, Aarbergstrasse 5, 2560, Nidau-Biel, Switzerland
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center Berlin (DHZB), Augustenburger Platz 1, 13353, Berlin, Germany
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Altenberger Strasse 69, 4040, Linz, Austria
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, 13353, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13347, Berlin, Germany
- Berlin Institute of Health (BIH), Anna-Louisa-Karsch Str. 2, 10178, Berlin, Germany
| |
Collapse
|
23
|
Yu Q, Li W, Xie D, Zheng X, Huang T, Xue P, Guo B, Gao Y, Zhang C, Sun P, Li M, Wang G, Cheng X, Zheng Q, Song Z. PI3Kγ promotes vascular smooth muscle cell phenotypic modulation and transplant arteriosclerosis via a SOX9-dependent mechanism. EBioMedicine 2018; 36:39-53. [PMID: 30241919 PMCID: PMC6197754 DOI: 10.1016/j.ebiom.2018.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/31/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
Background Transplant arteriosclerosis (TA) remains the major cause of chronic graft failure in solid organ transplantation. The phenotypic modulation of vascular smooth muscle cells (VSMCs) is a key event for the initiation and progression of neointimal formation and TA. This study aims to explore the role and underlying mechanism of phosphoinositide 3-kinases γ (PI3Kγ) in VSMC phenotypic modulation and TA. Methods The rat model of aortic transplantation was established to detect PI3Kγ expression and its role in neointimal formation and vascular remodeling in vivo. PI3Kγ shRNA transfection was employed to knockdown PI3Kγ gene. Aortic VSMCs was cultured and treated with TNF-α to explore the role and molecular mechanism of PI3Kγ in VSMC phenotypic modulation. Findings Activated PI3Kγ/p-Akt signaling was observed in aortic allografts and in TNF-α-treated VSMCs. Lentivirus-mediated shRNA transfection effectively inhibited PI3Kγ expression in medial VSMCs while restoring the expression of VSMC contractile genes, associated with impaired neointimal formation in aortic allografts. In cultured VSMCs, PI3Kγ blockade with pharmacological inhibitor or genetic knockdown markedly abrogated TNF-α-induced downregulation of VSMC contractile genes and increase in cellular proliferation and migration. Moreover, SOX9 located in nucleus competitively inhibited the interaction of Myocardin and SRF, while PI3Kγ inhibition robustly reduced SOX9 expression and its nuclear translocation and repaired the Myocardin/SRF association. Interpretation These results suggest that PI3Kγ plays a critical role in VSMC phenotypic modulation via a SOX9-dependent mechanism. Therefore, PI3Kγ in VSMCs may represent a promising therapeutic target for the treatment of TA. Fund National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Qihong Yu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dawei Xie
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xichuan Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Huang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Xue
- Departments of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Guo
- Department of Hepatology and Oncology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Yang Gao
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Sun
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoliang Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qichang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
24
|
Augstein A, Mierke J, Poitz DM, Strasser RH. Sox9 is increased in arterial plaque and stenosis, associated with synthetic phenotype of vascular smooth muscle cells and causes alterations in extracellular matrix and calcification. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2526-2537. [PMID: 29777903 DOI: 10.1016/j.bbadis.2018.05.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/23/2018] [Accepted: 05/15/2018] [Indexed: 12/20/2022]
Abstract
Vascular smooth muscle cells (VSMC) exhibit a dual role in progression and maintenance of arteriosclerosis. They are fundamental for plaque stability but also can drive plaque progression. During pathogenic vascular remodeling, VSMC transdifferentiate into a phenotype with enhanced proliferation and migration. Moreover, they exert an increased capacity to generate extracellular matrix proteins. A special lineage of transdifferentiated VSMC expresses Sox9, a multi-functional transcription factor. The aim of the study was to examine the role of Sox9 in phenotypic alterations leading to arteriosclerosis. Using mouse models for arterial stenosis, Sox9 induction in diseased vessels was verified. The phenotypic switch of VSMC from contractile to proliferative nature caused a significant increase of Sox9 expression. Various factors known to be involved in the progression of arteriosclerosis were examined for their ability to modulate Sox9 expression in VSMC. While PDGF-BB resulted in a strong transient upregulation of Sox9, TGF-β1 appeared to be responsible for a moderate, but prolonged increase of Sox9 expression. Beside the regulation, functional studies focused on knockout and overexpression of Sox9. A Sox9-dependent alteration of extracellular matrix could be revealed and was associated with an upregulated calcium deposition. Taken together, Sox9 is identified as important factor of VSMC function by modulation the extracellular matrix composition and calcium deposition, which are important processes in plaque development.
Collapse
Affiliation(s)
- Antje Augstein
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany.
| | - Johannes Mierke
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - David M Poitz
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| | - Ruth H Strasser
- Internal Medicine and Cardiology, Heart Center Dresden, TU Dresden, Germany
| |
Collapse
|
25
|
Lang F, Leibrock C, Pelzl L, Gawaz M, Pieske B, Alesutan I, Voelkl J. Therapeutic Interference With Vascular Calcification-Lessons From Klotho-Hypomorphic Mice and Beyond. Front Endocrinol (Lausanne) 2018; 9:207. [PMID: 29780355 PMCID: PMC5945862 DOI: 10.3389/fendo.2018.00207] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/13/2018] [Indexed: 12/13/2022] Open
Abstract
Medial vascular calcification, a major pathophysiological process associated with cardiovascular disease and mortality, involves osteo-/chondrogenic transdifferentiation of vascular smooth muscle cells (VSMCs). In chronic kidney disease (CKD), osteo-/chondrogenic transdifferentiation of VSMCs and, thus, vascular calcification is mainly driven by hyperphosphatemia, resulting from impaired elimination of phosphate by the diseased kidneys. Hyperphosphatemia with subsequent vascular calcification is a hallmark of klotho-hypomorphic mice, which are characterized by rapid development of multiple age-related disorders and early death. In those animals, hyperphosphatemia results from unrestrained formation of 1,25(OH)2D3 with subsequent retention of calcium and phosphate. Analysis of klotho-hypomorphic mice and mice with vitamin D3 overload uncovered several pathophysiological mechanisms participating in the orchestration of vascular calcification and several therapeutic opportunities to delay or even halt vascular calcification. The present brief review addresses the beneficial effects of bicarbonate, carbonic anhydrase inhibition, magnesium supplementation, mineralocorticoid receptor (MR) blockage, and ammonium salts. The case is made that bicarbonate is mainly effective by decreasing intestinal phosphate absorption, and that carbonic anhydrase inhibition leads to metabolic acidosis, which counteracts calcium-phosphate precipitation and VSMC transdifferentiation. Magnesium supplementation, MR blockage and ammonium salts are mainly effective by interference with osteo-/chondrogenic signaling in VSMCs. It should be pointed out that the, by far, most efficient substances are ammonium salts, which may virtually prevent vascular calcification. Future research will probably uncover further therapeutic options and, most importantly, reveal whether these observations in mice can be translated into treatment of patients suffering from vascular calcification, such as patients with CKD.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- *Correspondence: Florian Lang,
| | - Christina Leibrock
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
- Fresenius Kabi Deutschland GmbH, Bad Homburg, Germany
| | - Lisann Pelzl
- Department of Physiology I, Eberhard Karls-University, Tübingen, Germany
| | - Meinrad Gawaz
- Department of Internal Medicine III, Eberhard Karls-University, Tübingen, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Ioana Alesutan
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Jakob Voelkl
- Department of Internal Medicine and Cardiology, Charité-Universität Medizin Berlin, Berlin, Germany
- Partner Site Berlin, German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
26
|
Dai X, Thiagarajan D, Fang J, Shen J, Annam NP, Yang Z, Jiang H, Ju D, Xie Y, Zhang K, Tseng YY, Yang Z, Rishi AK, Li HJ, Yang M, Li L. SM22α suppresses cytokine-induced inflammation and the transcription of NF-κB inducing kinase (Nik) by modulating SRF transcriptional activity in vascular smooth muscle cells. PLoS One 2017; 12:e0190191. [PMID: 29284006 PMCID: PMC5746259 DOI: 10.1371/journal.pone.0190191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 12/08/2017] [Indexed: 12/13/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) phenotypic modulation is characterized by the downregulation of SMC actin cytoskeleton proteins. Our published study shows that depletion of SM22α (aka SM22, Transgelin, an actin cytoskeleton binding protein) promotes inflammation in SMCs by activating NF-κB signal pathways both in cultured VSMCs and in response to vascular injury. The goal of this study is to investigate the underlying molecular mechanisms whereby SM22 suppresses NF-κB signaling pathways under inflammatory condition. NF-κB inducing kinase (Nik, aka MAP3K14, activated by the LTβR) is a key upstream regulator of NF-κB signal pathways. Here, we show that SM22 overexpression suppresses the expression of NIK and its downstream NF-κB canonical and noncanonical signal pathways in a VSMC line treated with a LTβR agonist. SM22 regulates NIK expression at both transcriptional and the proteasome-mediated post-translational levels in VSMCs depending on the culture condition. By qPCR, chromatin immunoprecipitation and luciferase assays, we found that Nik is a transcription target of serum response factor (SRF). Although SM22 is known to be expressed in the cytoplasm, we found that SM22 is also expressed in the nucleus where SM22 interacts with SRF to inhibit the transcription of Nik and prototypical SRF regulated genes including c-fos and Egr3. Moreover, carotid injury increases NIK expression in Sm22-/- mice, which is partially relieved by adenovirally transduced SM22. These findings reveal for the first time that SM22 is expressed in the nucleus in addition to the cytoplasm of VSMCs to regulate the transcription of Nik and its downstream proinflammatory NF-kB signal pathways as a modulator of SRF during vascular inflammation.
Collapse
Affiliation(s)
- Xiaohua Dai
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Devi Thiagarajan
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Jingye Fang
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Jianbin Shen
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Neeraja Priyanka Annam
- Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, Michigan, United States of America
| | - Zhao Yang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Hong Jiang
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
| | - Donghong Ju
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Youming Xie
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Yan Yuan Tseng
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, Wayne State University, Detroit, Michigan, United States of America
| | - Arun K. Rishi
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
- John D. Dingell VA Medical Center, Detroit, Michigan, United States of America
| | - Hui J. Li
- Department of Medicine, University of Massachusetts, Worcester, Massachusetts, United States of America
| | - Maozhou Yang
- Bone and Joint Center, Henry Ford Hospital, Detroit, Michigan, United States of America
| | - Li Li
- Department of Internal Medicine, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, United States of America
- Cardiovascular Research Institute, Wayne State University, Detroit, Michigan, United States of America
| |
Collapse
|
27
|
Öztürk E, Despot-Slade E, Pichler M, Zenobi-Wong M. RhoA activation and nuclearization marks loss of chondrocyte phenotype in crosstalk with Wnt pathway. Exp Cell Res 2017; 360:113-124. [PMID: 28865751 DOI: 10.1016/j.yexcr.2017.08.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/20/2017] [Accepted: 08/29/2017] [Indexed: 12/24/2022]
Abstract
De-differentiation comprises a major drawback for the use of autologous chondrocytes in cartilage repair. Here, we investigate the role of RhoA and canonical Wnt signaling in chondrocyte phenotype. Chondrocyte de-differentiation is accompanied by an upregulation and nuclear localization of RhoA. Effectors of canonical Wnt signaling including β-catenin and YAP/TAZ are upregulated in de-differentiating chondrocytes in a Rho-dependent manner. Inhibition of Rho activation with C3 transferase inhibits nuclear localization of RhoA, induces expression of chondrogenic markers on 2D and enhances the chondrogenic effect of 3D culturing. Upregulation of chondrogenic markers by Rho inhibition is accompanied by loss of canonical Wnt signaling markers in 3D or on 2D whereas treatment of chondrocytes with Wnt-3a abrogates this effect. However, induction of canonical Wnt signaling inhibits chondrogenic markers on 2D but enhances chondrogenic re-differentiation on 2D with C3 transferase or in 3D. These data provide insights on the context-dependent role of RhoA and Wnt signaling in de-differentiation and on mechanisms to induce chondrogenic markers for therapeutic approaches.
Collapse
Affiliation(s)
- Ece Öztürk
- Cartilage Engineering + Regeneration Laboratory, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Evelin Despot-Slade
- Cartilage Engineering + Regeneration Laboratory, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Michael Pichler
- Cartilage Engineering + Regeneration Laboratory, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland
| | - Marcy Zenobi-Wong
- Cartilage Engineering + Regeneration Laboratory, ETH Zurich, Otto-Stern-Weg 7, 8093 Zurich, Switzerland.
| |
Collapse
|
28
|
Freise C, Bobb V, Querfeld U. Collagen XIV and a related recombinant fragment protect human vascular smooth muscle cells from calcium-/phosphate-induced osteochondrocytic transdifferentiation. Exp Cell Res 2017; 358:242-252. [PMID: 28655510 DOI: 10.1016/j.yexcr.2017.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 02/08/2023]
Abstract
Transdifferentiation of vascular smooth muscle cells (VSMC) promotes the development of vascular calcifications such as arteriosclerosis. The aim was to investigate effects of specific extracellular matrix (ECM) components on transdifferentiation of VSMC to identify novel ECM-based therapeutic tools. Human collagens I & IV (CI, CIV) along with collagen XIV (CXIV) and a CXIV-derived fragment (CXIV-F), both of which induce differentiation, were applied in an in-vitro model of calcium-/phosphate (Ca/P)-induced osteochondrocytic transdifferentiation of human and murine VSMC. Transdifferentiation was determined by RT-PCR and calcium contents of VSMC cultures. Signaling pathways involved were determined by western-blot and luciferase reporter plasmid assays. Under normal culture conditions, CI induced VSMC proliferation and a more epithelioid/synthetic phenotype while CIV and predominantly CXIV provoked opposite effects. CIV and CXIV further blocked Ca/P-induced osteochondrocytic transdifferentiation of VSMC displayed e.g. by reduced gene expressions of Runx2, Sox9, osterix and increased expressions of αSMA and SM22α. This involved impaired activation of ERK1/2, NF-ĸB and Wnt-signaling. Similar preventive effects were achieved by applying CXIV-F. Impaired preventive effects of CXIV by co-treatment with a cluster of differentiation (CD)44 agonist propose CD44 as a CXIV-target structure on VSMC. In conclusion, CXIV and CXIV-F interfere with osteochondrocytic transdifferentiation of VSMC and should be further explored as potential therapeutic tools in vascular calcification.
Collapse
Affiliation(s)
- Christian Freise
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Nephrology, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany; Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Mitte, Hessische Str. 3-4, 10115 Berlin, Germany.
| | - Veronika Bobb
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Nephrology, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany; Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Mitte, Hessische Str. 3-4, 10115 Berlin, Germany
| | - Uwe Querfeld
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Pediatric Nephrology, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany; Center for Cardiovascular Research, Charité - Universitätsmedizin Berlin, Campus Mitte, Hessische Str. 3-4, 10115 Berlin, Germany
| |
Collapse
|
29
|
Klotho suppresses high phosphate-induced osteogenic responses in human aortic valve interstitial cells through inhibition of Sox9. J Mol Med (Berl) 2017; 95:739-751. [PMID: 28332126 DOI: 10.1007/s00109-017-1527-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 02/27/2017] [Accepted: 03/08/2017] [Indexed: 12/19/2022]
Abstract
Elevated level of blood phosphate (Pi) associated with chronic kidney disease (CKD) is a risk factor of aortic valve calcification. Aortic valve interstitial cells (AVICs) display osteogenic responses to high Pi although the underlying mechanism is incompletely understood. Sox9 is a pro-chondrogenic factor and may play a role in ectopic tissue calcification. Circulating and kidney levels of Klotho are reduced in patients with CKD. We hypothesized that Sox9 mediates high Pi-induced osteogenic responses in human AVICs and that Klotho inhibits the responses. Treatment of human AVICs with high Pi increased protein levels of Runt-related transcription factor 2 (Runx2) and alkaline phosphatase (ALP), and a prolonged exposure to high Pi caused calcium deposition. High Pi induced Sox9 upregulation through PKD and Akt activation. Knockdown of Sox9 essentially abolished the effect of high Pi on the osteogenic responses. Lower Klotho levels were observed in calcified aortic valve tissues. Interestingly, high Pi decreased Klotho levels in AVICs from normal valves, and treatment with recombinant Klotho markedly reduced the effect of high Pi on the levels of Sox9, Runx2, and ALP and suppressed calcium deposition. We conclude that high Pi induces human AVIC osteogenic responses through Sox9. Human AVICs express Klotho, and its levels in AVICs are modulated by high Pi and valvular calcification. Importantly, Klotho suppresses the pro-osteogenic effect of high Pi on human AVICs. These novel findings indicate that modulation of Klotho may have therapeutic potential for mitigation of valvular calcification associated with CKD. KEY MESSAGES CAVD associated with chronic kidney disease is a significant clinical problem. High phosphate upregulates Sox9 through AKT and PKD in human AVICs. Calcified human aortic valves have lower levels of Klotho. Klotho suppresses Sox9 upregulation and intranuclear translocation. Klotho inhibits high phosphate-induced osteogenic activity in human AVICs.
Collapse
|
30
|
MRTF-A signaling regulates the acquisition of the contractile phenotype in dedifferentiated chondrocytes. Matrix Biol 2016; 62:3-14. [PMID: 27751947 DOI: 10.1016/j.matbio.2016.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 10/09/2016] [Accepted: 10/10/2016] [Indexed: 11/22/2022]
Abstract
Chondrocyte culture as a monolayer for cell number expansion results in dedifferentiation whereby expanded cells acquire contractile features and increased actin polymerization status. This study determined whether the actin polymerization based signaling pathway, myocardin-related transcription factor-a (MRTF-A) is involved in regulating this contractile phenotype. Serial passaging of chondrocytes in monolayer culture to passage 2 resulted in increased gene and protein expression of the contractile molecules alpha-smooth muscle actin, transgelin and vinculin compared to non-passaged, primary cells. This resulted in a functional change as passaged 2, but not primary, chondrocytes were capable of contracting type I collagen gels in a stress-relaxed contraction assay. These changes were associated with increased actin polymerization and MRTF-A nuclear localization. The involvement of actin was demonstrated by latrunculin B depolymerization of actin which reversed these changes. Alternatively cytochalasin D which activates MRTF-A increased gene and protein expression of α-smooth muscle actin, transgelin and vinculin, whereas CCG1423 which deactivates MRTF-A decreased these molecules. The involvement of MRTF-A signaling was confirmed by gene silencing of MRTF or its co-factor serum response factor. Knockdown experiments revealed downregulation of α-smooth muscle actin and transgelin gene and protein expression, and inhibition of gel contraction. These findings demonstrate that passaged chondrocytes acquire a contractile phenotype and that this change is modulated by the actin-MRTF-A-serum response factor signaling pathway.
Collapse
|
31
|
Balogh E, Tóth A, Tolnai E, Bodó T, Bányai E, Szabó DJ, Petrovski G, Jeney V. Osteogenic differentiation of human lens epithelial cells might contribute to lens calcification. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1724-31. [PMID: 27318027 DOI: 10.1016/j.bbadis.2016.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/24/2016] [Accepted: 06/14/2016] [Indexed: 01/14/2023]
Abstract
Calcification of the human lens has been described in senile cataracts and in young patients with congenital cataract or chronic uveitis. Lens calcification is also a major complication of cataract surgery and plays a role in the opacification of intraocular lenses. A cell-mediated process has been suggested in the background of lens calcification, but so far the exact mechanism remained unexplored. Lens calcification shares remarkable similarities with vascular calcification; in both pathological processes hydroxyapatite accumulates in the soft tissue. Vascular calcification is a regulated, cell-mediated process in which vascular cells undergo osteogenic differentiation. Our objective was to investigate whether human lens epithelial cells (HuLECs) can undergo osteogenic transition in vitro, and whether this process contributes to lens calcification. We used inorganic phosphate (Pi) and Ca to stimulate osteogenic differentiation of HuLECs. Osteogenic stimuli (2.5mmol/L Pi and 1.2mmol/L Ca) induced extracellular matrix mineralization and Ca deposition in HuLECs with the critical involvement of active Pi uptake. Osteogenic stimuli almost doubled mRNA expressions of osteo-/chondrogenic transcription factors Runx2 and Sox9, which was accompanied by a 1.9-fold increase in Runx2 and a 5.5-fold increase in Sox9 protein expressions. Osteogenic stimuli induced mRNA and protein expressions of alkaline phosphatase and osteocalcin in HuLEC. Ca content was higher in human cataractous lenses, compared to non-cataractous controls (n=10). Osteocalcin, an osteoblast-specific protein, was expressed in 2 out of 10 cataractous lenses. We conclude that osteogenic stimuli induce osteogenic differentiation of HuLECs and propose that this mechanism might play a role in lens calcification.
Collapse
Affiliation(s)
- Enikő Balogh
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Andrea Tóth
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Emese Tolnai
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tímea Bodó
- Department of Neurology, Bethesda Children's Hospital, Budapest, Hungary
| | - Emese Bányai
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dóra Júlia Szabó
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Goran Petrovski
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, Szeged, Hungary; Center of Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Viktória Jeney
- Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
32
|
Xu D, Gu JT, Yi B, Chen L, Wang GS, Qian GS, Lu KZ. Requirement of miR-9-dependent regulation of Myocd in PASMCs phenotypic modulation and proliferation induced by hepatopulmonary syndrome rat serum. J Cell Mol Med 2015; 19:2453-61. [PMID: 26147104 PMCID: PMC4594686 DOI: 10.1111/jcmm.12631] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/13/2015] [Indexed: 12/18/2022] Open
Abstract
Hepatopulmonary syndrome (HPS) is characterized by a triad of severe liver disease, intrapulmonary vascular dilation and hypoxaemia. Pulmonary vascular remodelling (PVR) is a key feature of HPS pathology. Our previous studies have established the role of the pulmonary artery smooth muscle cell (PASMC) phenotypic modulation and proliferation in HPS-associated PVR. Myocardin, a robust transcriptional coactivator of serum response factor, plays a critical role in the vascular smooth muscle cell phenotypic switch. However, the mechanism regulating myocardin upstream signalling remains unclear. In this study, treatment of rat PASMCs with serum drawn from common bile duct ligation rats, which model symptoms of HPS, resulted in a significant increase in miR-9 expression correlated with a decrease in expression of myocardin and the phenotypic markers SM-α-actin and smooth muscle-specific myosin heavy chain (SM-MHC). Furthermore, miRNA functional analysis and luciferase reporter assay demonstrated that miR-9 effectively regulated myocardin expression by directly binding to its 3′-untranslated region. Both the knockdown of miR-9 and overexpression of myocardin effectively attenuated the HPS rat serum-induced phenotype switch and proliferation of PASMCs. Taken together, the findings of our present study demonstrate that miR-9 is required in HPS rat serum-induced phenotypic modulation and proliferation of PASMCs for targeting of myocardin and that miR-9 may serve as a potential therapeutic target in HPS.
Collapse
Affiliation(s)
- Duo Xu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jian-teng Gu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bin Yi
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lin Chen
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Guan-song Wang
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Gui-sheng Qian
- Institute of Respiratory Disease, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Kai-zhi Lu
- Department of Anaesthesia, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
33
|
|
34
|
Briot A, Jaroszewicz A, Warren CM, Lu J, Touma M, Rudat C, Hofmann JJ, Airik R, Weinmaster G, Lyons K, Wang Y, Kispert A, Pellegrini M, Iruela-Arispe ML. Repression of Sox9 by Jag1 is continuously required to suppress the default chondrogenic fate of vascular smooth muscle cells. Dev Cell 2015; 31:707-21. [PMID: 25535917 DOI: 10.1016/j.devcel.2014.11.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 09/11/2014] [Accepted: 11/13/2014] [Indexed: 01/15/2023]
Abstract
Acquisition and maintenance of vascular smooth muscle fate are essential for the morphogenesis and function of the circulatory system. Loss of contractile properties or changes in the identity of vascular smooth muscle cells (vSMCs) can result in structural alterations associated with aneurysms and vascular wall calcification. Here we report that maturation of sclerotome-derived vSMCs depends on a transcriptional switch between mouse embryonic days 13 and 14.5. At this time, Notch/Jag1-mediated repression of sclerotome transcription factors Pax1, Scx, and Sox9 is necessary to fully enable vSMC maturation. Specifically, Notch signaling in vSMCs antagonizes sclerotome and cartilage transcription factors and promotes upregulation of contractile genes. In the absence of the Notch ligand Jag1, vSMCs acquire a chondrocytic transcriptional repertoire that can lead to ossification. Importantly, our findings suggest that sustained Notch signaling is essential throughout vSMC life to maintain contractile function, prevent vSMC reprogramming, and promote vascular wall integrity.
Collapse
Affiliation(s)
- Anaïs Briot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Artur Jaroszewicz
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carmen M Warren
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jing Lu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marlin Touma
- Division of Anesthesiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Carsten Rudat
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Jennifer J Hofmann
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rannar Airik
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Gerry Weinmaster
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Karen Lyons
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yibin Wang
- Division of Anesthesiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Andreas Kispert
- Institut fur Molekularbiologie, Medizinische Hochschule Hannover, 30625 Hannover, Germany
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
35
|
Leibrock CB, Alesutan I, Voelkl J, Pakladok T, Michael D, Schleicher E, Kamyabi-Moghaddam Z, Quintanilla-Martinez L, Kuro-o M, Lang F. NH4Cl Treatment Prevents Tissue Calcification in Klotho Deficiency. J Am Soc Nephrol 2015; 26:2423-33. [PMID: 25644113 DOI: 10.1681/asn.2014030230] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 11/30/2014] [Indexed: 11/03/2022] Open
Abstract
Klotho, a cofactor in suppressing 1,25(OH)2D3 formation, is a powerful regulator of mineral metabolism. Klotho-hypomorphic mice (kl/kl) exhibit excessive plasma 1,25(OH)2D3, Ca(2+), and phosphate concentrations, severe tissue calcification, volume depletion with hyperaldosteronism, and early death. Calcification is paralleled by overexpression of osteoinductive transcription factor Runx2/Cbfa1, Alpl, and senescence-associated molecules Tgfb1, Pai-1, p21, and Glb1. Here, we show that NH4Cl treatment in drinking water (0.28 M) prevented soft tissue and vascular calcification and increased the life span of kl/kl mice >12-fold in males and >4-fold in females without significantly affecting extracellular pH or plasma concentrations of 1,25(OH)2D3, Ca(2+), and phosphate. NH4Cl treatment significantly decreased plasma aldosterone and antidiuretic hormone concentrations and reversed the increase of Runx2/Cbfa1, Alpl, Tgfb1, Pai-1, p21, and Glb1 expression in aorta of kl/kl mice. Similarly, in primary human aortic smooth muscle cells (HAoSMCs), NH4Cl treatment reduced phosphate-induced mRNA expression of RUNX2/CBFA1, ALPL, and senescence-associated molecules. In both kl/kl mice and phosphate-treated HAoSMCs, levels of osmosensitive transcription factor NFAT5 and NFAT5-downstream mediator SOX9 were higher than in controls and decreased after NH4Cl treatment. Overexpression of NFAT5 in HAoSMCs mimicked the effect of phosphate and abrogated the effect of NH4Cl on SOX9, RUNX2/CBFA1, and ALPL mRNA expression. TGFB1 treatment of HAoSMCs upregulated NFAT5 expression and prevented the decrease of phosphate-induced NFAT5 expression after NH4Cl treatment. In conclusion, NH4Cl treatment prevents tissue calcification, reduces vascular senescence, and extends survival of klotho-hypomorphic mice. The effects of NH4Cl on vascular osteoinduction involve decrease of TGFB1 and inhibition of NFAT5-dependent osteochondrogenic signaling.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | |
Collapse
|
36
|
Abstract
Myocardin (MYOCD) is a potent transcriptional coactivator that functions primarily in cardiac muscle and smooth muscle through direct contacts with serum response factor (SRF) over cis elements known as CArG boxes found near a number of genes encoding for contractile, ion channel, cytoskeletal, and calcium handling proteins. Since its discovery more than 10 years ago, new insights have been obtained regarding the diverse isoforms of MYOCD expressed in cells as well as the regulation of MYOCD expression and activity through transcriptional, post-transcriptional, and post-translational processes. Curiously, there are a number of functions associated with MYOCD that appear to be independent of contractile gene expression and the CArG-SRF nucleoprotein complex. Further, perturbations in MYOCD gene expression are associated with an increasing number of diseases including heart failure, cancer, acute vessel disease, and diabetes. This review summarizes the various biological and pathological processes associated with MYOCD and offers perspectives to several challenges and future directions for further study of this formidable transcriptional coactivator.
Collapse
Affiliation(s)
- Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
37
|
McGowan SE, McCoy DM. Regulation of fibroblast lipid storage and myofibroblast phenotypes during alveolar septation in mice. Am J Physiol Lung Cell Mol Physiol 2014; 307:L618-31. [PMID: 25150063 DOI: 10.1152/ajplung.00144.2014] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Signaling through platelet-derived growth factor receptor-α (PDGFRα) is required for alveolar septation and participates in alveolar regeneration after pneumonectomy. In both adipose tissue and skeletal muscle, bipotent pdgfrα-expressing progenitors expressing delta-like ligand-1 or sex-determining region Y box 9 (Sox9) may differentiate into either lipid storage cells or myofibroblasts. We analyzed markers of mesenchymal progenitors and differentiation in lung fibroblasts (LF) with different levels (absent, low, or high) of pdgfrα gene expression. A larger proportion of pdgfrα-expressing than nonexpressing LF contained Sox9. Neutral lipids, CD166, and Tcf21 were more abundant in LF with a lower compared with a higher level of pdgfrα gene expression. PDGF-A increased Sox9 in primary LF cultures, suggesting that active signaling through PDGFRα is required to maintain Sox9. As alveolar septation progresses from postnatal day (P) 8 to P12, fewer pdgfrα-expressing LF contain Sox9, whereas more of these LF contain myocardin-like transcription factor-A, showing that Sox9 diminishes as LF become myofibroblasts. At P8, neutral lipid droplets predominate in LF with the lower level of pdgfrα gene expression, whereas transgelin (tagln) was predominantly expressed in LF with higher pdgfrα gene expression. Targeted deletion of pdgfrα in LF, which expressed tagln, reduced Sox9 in α-actin (α-SMA, ACTA2)-containing LF, whereas it increased the abundance of cell surface delta-like protein-1 (as well as peroxisome proliferator-activated receptor-γ and tcf21 mRNA in LF, which also expressed stem cell antigen-1). Thus pdgfrα deletion differentially alters delta-like protein-1 and Sox9, suggesting that targeting different downstream pathways in PDGF-A-responsive LF could identify strategies that promote lung regeneration without initiating fibrosis.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
38
|
Kauffenstein G, Pizard A, Le Corre Y, Vessières E, Grimaud L, Toutain B, Labat C, Mauras Y, Gorgels TG, Bergen AA, Le Saux O, Lacolley P, Lefthériotis G, Henrion D, Martin L. Disseminated arterial calcification and enhanced myogenic response are associated with abcc6 deficiency in a mouse model of pseudoxanthoma elasticum. Arterioscler Thromb Vasc Biol 2014; 34:1045-56. [PMID: 24675664 DOI: 10.1161/atvbaha.113.302943] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Pseudoxanthoma elasticum is an inherited metabolic disorder resulting from ABCC6 gene mutations. It is characterized by progressive calcification and fragmentation of elastic fibers in the skin, retina, and the arterial wall. Despite calcium accumulation in the arteries of patients with pseudoxanthoma elasticum, functional consequences remain unknown. In the present study, we investigated arterial structure and function in Abcc6(-/-) mice, a model of the human disease. APPROACH AND RESULTS Arterial calcium accumulation was evaluated using alizarin red stain and atomic absorption spectrometry. Expression of genes involved in osteochondrogenic differentiation was measured by polymerase chain reaction. Elastic arterial properties were evaluated by carotid echotracking. Vascular reactivity was evaluated using wire and pressure myography and remodeling using histomorphometry. Arterial calcium accumulation was 1.5- to 2-fold higher in Abcc6(-/-) than in wild-type mice. Calcium accumulated locally leading to punctuate pattern. Old Abcc6(-/-) arteries expressed markers of both osteogenic (Runx2, osteopontin) and chondrogenic lineage (Sox9, type II collagen). Abcc6(-/-) arteries displayed slight increase in arterial stiffness and vasoconstrictor tone in vitro tended to be higher in response to phenylephrine and thromboxane A2. Pressure-induced (myogenic) tone was significantly higher in Abcc6(-/-) arteries than in wild type. Arterial blood pressure was not significantly changed in Abcc6(-/-), despite higher variability. CONCLUSIONS Scattered arterial calcium depositions are probably a result of osteochondrogenic transdifferentiation of vascular cells. Lower elasticity and increased myogenic tone without major changes in agonist-dependent contraction evidenced in aged Abcc6(-/-) mice suggest a reduced control of local blood flow, which in turn may alter vascular homeostasis in the long term.
Collapse
Affiliation(s)
- Gilles Kauffenstein
- From the CNRS UMR 6214, INSERM U1083, l'UNAM (G.K., Y.L.C., E.V., L.G., B.T., G.L., D.H., L.M.) and Laboratoire de Pharmacologie-Toxicologie, l'UNAM, Université d'Angers (Y.M.), University Hospital Angers, Angers, France; INSERM, U1116 (A.P., C.L., P.L.), Université de Lorraine, Vandoeuvre-lès-Nancy, France; Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI (O.L.S.); Molecular Ophthalmogenetics, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands (T.G.G., A.A.B.); and Departments of Ophthalmology (A.A.B.) and Clinical Genetics (A.A.B.), Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zheng XL. Myocardin and smooth muscle differentiation. Arch Biochem Biophys 2014; 543:48-56. [DOI: 10.1016/j.abb.2013.12.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/15/2013] [Accepted: 12/18/2013] [Indexed: 01/08/2023]
|
40
|
Andrikou C, Iovene E, Rizzo F, Oliveri P, Arnone MI. Myogenesis in the sea urchin embryo: the molecular fingerprint of the myoblast precursors. EvoDevo 2013; 4:33. [PMID: 24295205 PMCID: PMC4175510 DOI: 10.1186/2041-9139-4-33] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/02/2013] [Indexed: 01/01/2023] Open
Abstract
Background In sea urchin larvae the circumesophageal fibers form a prominent muscle system of mesodermal origin. Although the morphology and later development of this muscle system has been well-described, little is known about the molecular signature of these cells or their precise origin in the early embryo. As an invertebrate deuterostome that is more closely related to the vertebrates than other commonly used model systems in myogenesis, the sea urchin fills an important phylogenetic gap and provides a unique perspective on the evolution of muscle cell development. Results Here, we present a comprehensive description of the development of the sea urchin larval circumesophageal muscle lineage beginning with its mesodermal origin using high-resolution localization of the expression of several myogenic transcriptional regulators and differentiation genes. A few myoblasts are bilaterally distributed at the oral vegetal side of the tip of the archenteron and first appear at the late gastrula stage. The expression of the differentiation genes Myosin Heavy Chain, Tropomyosin I and II, as well as the regulatory genes MyoD2, FoxF, FoxC, FoxL1, Myocardin, Twist, and Tbx6 uniquely identify these cells. Interestingly, evolutionarily conserved myogenic factors such as Mef2, MyoR and Six1/2 are not expressed in sea urchin myoblasts but are found in other mesodermal domains of the tip of the archenteron. The regulatory states of these domains were characterized in detail. Moreover, using a combinatorial analysis of gene expression we followed the development of the FoxF/FoxC positive cells from the onset of expression to the end of gastrulation. Our data allowed us to build a complete map of the Non-Skeletogenic Mesoderm at the very early gastrula stage, in which specific molecular signatures identify the precursors of different cell types. Among them, a small group of cells within the FoxY domain, which also express FoxC and SoxE, have been identified as plausible myoblast precursors. Together, these data support a very early gastrula stage segregation of the myogenic lineage. Conclusions From this analysis, we are able to precisely define the regulatory and differentiation signatures of the circumesophageal muscle in the sea urchin embryo. Our findings have important implications in understanding the evolution of development of the muscle cell lineage at the molecular level. The data presented here suggest a high level of conservation of the myogenic specification mechanisms across wide phylogenetic distances, but also reveal clear cases of gene cooption.
Collapse
Affiliation(s)
| | | | | | | | - Maria Ina Arnone
- Cellular and Developmental Biology, Stazione Zoologica Anton Dohrn, Napoli 80121, Italy.
| |
Collapse
|