1
|
Kim Y, Yeo Y, Kim M, Son YW, Kim J, Kim KL, Kim S, Oh S, Kim Y, Lee H, Park HW, Lee D, Lee SJ, Kang C, Choi H, Park CS, Lee SP, Suh W, Jang JH. A highly mobile adeno-associated virus targeting vascular smooth muscle cells for the treatment of pulmonary arterial hypertension. Nat Biomed Eng 2025:10.1038/s41551-025-01379-8. [PMID: 40301691 DOI: 10.1038/s41551-025-01379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 03/24/2025] [Indexed: 05/01/2025]
Abstract
In pulmonary arterial hypertension (PAH), a phenotypic switch in pulmonary arterial smooth muscle cells (PASMCs) that is primarily caused by aberrant gene regulatory networks can lead to dysregulated vascular remodelling, heart failure or death. No curative therapies for PAH are currently available, presumably because of a lack of viral vectors specifically targeting PASMCs. Here we show that a highly mobile and PASMC-tropic adeno-associated virus variant developed via directed evolution overcomes physical barriers that inhibit its transfer from bronchial airways to vascular layers, ultimately boosting therapeutic efficacy in murine models of PAH. Intratracheal administration of the adeno-associated virus variant carrying a transgene for fibroblast growth factor 12-a key factor regulating the PASMC phenotype-suppressed pulmonary vascular remodelling, prevented the development of PAH in mice and reversed established PAH in rats. The variant's mobility and enhanced tropism for PASMCs may enable curative treatments for PAH.
Collapse
Affiliation(s)
- Yoojin Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
- Nano Science Technology Institute, Yonsei University, Seoul, Korea
| | - Yeongju Yeo
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Minju Kim
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Yong-Wook Son
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Joowon Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Koung Li Kim
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Seohee Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Seokmin Oh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Yunha Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Hyowoo Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Hyun-Woo Park
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Dongsoo Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea
| | - Sung Jin Lee
- R&D Center, GluGene Therapeutics Inc., Seoul, Korea
| | | | | | - Chan Soon Park
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seung-Pyo Lee
- Division of Cardiology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Wonhee Suh
- Department of Global Innovative Drug, the Graduate School of Chung-Ang University, Chung-Ang University, Seoul, Korea.
- College of Pharmacy, Chung-Ang University, Seoul, Korea.
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, Korea.
- R&D Center, GluGene Therapeutics Inc., Seoul, Korea.
| |
Collapse
|
2
|
Wolfson SM, Beigel K, Anderson SE, Deal B, Weiner M, Lee SH, Taylor D, Heo SC, Heuckeroth RO, Hashmi SK. Rapid cyclic stretching induces a synthetic, proinflammatory phenotype in cultured human intestinal smooth muscle, with the potential to alter signaling to adjacent bowel cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.617767. [PMID: 39464046 PMCID: PMC11507745 DOI: 10.1101/2024.10.12.617767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background and Aims Bowel smooth muscle experiences mechanical stress constantly during normal function, and pathologic mechanical stressors in disease states. We tested the hypothesis that pathologic mechanical stress could alter transcription to induce smooth muscle phenotypic class switching. Methods Primary human intestinal smooth muscle cells (HISMCs), seeded on electrospun aligned poly-ε-caprolactone nano-fibrous scaffolds, were subjected to pathologic, high frequency (1 Hz) uniaxial 3% cyclic stretch (loaded) or kept unloaded in culture for 6 hours. Total RNA sequencing, qRT-PCR, and quantitative immunohistochemistry defined loading-induced changes in gene expression. NicheNet predicted how differentially expressed genes might impact HISMCs and other bowel cells. Results Loading induced differential expression of 4537 genes in HISMCs. Loaded HISMCs had a less contractile phenotype, with increased expression of synthetic SMC genes, proinflammatory cytokines, and altered expression of axon guidance molecules, growth factors and morphogens. Many differentially expressed genes encode secreted ligands that could act cell-autonomously on smooth muscle and on other cells in the bowel wall. Discussion HISMCs demonstrate remarkably rapid phenotypic plasticity in response to mechanical stress that may convert contractile HISMCs into proliferative, fibroblast-like cells or proinflammatory cells. These mechanical stress-induced changes in HISMC gene expression may be relevant for human bowel disease.
Collapse
Affiliation(s)
- Sharon M. Wolfson
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Katherine Beigel
- The Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sierra E. Anderson
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Brooke Deal
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Molly Weiner
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | | | - Deanne Taylor
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Chin Heo
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, 220 S 33rd St, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sohaib K. Hashmi
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, 220 S 33rd St, Philadelphia, PA 19104, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hospital of the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Yang S, Zhao Y, Cao S, Liu X, Feng M, Chen Y, Ma C, Zhan T, Zhang Q, Jia H, Zhao Y, Tong M, Yu Y, Liu X, Yang B, Zhang Y. Kanglexin counters vascular smooth muscle cell dedifferentiation and associated arteriosclerosis through inhibiting PDGFR. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155704. [PMID: 38759316 DOI: 10.1016/j.phymed.2024.155704] [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: 02/09/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-β. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-β-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-β-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.
Collapse
Affiliation(s)
- Shuang Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yixiu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shifeng Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinxin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Min Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yi Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chunyue Ma
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tingting Zhan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Qi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Honglin Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ming Tong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuanyuan Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xue Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Baofeng Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| | - Yan Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
4
|
Fang G, Tian Y, Huang S, Zhang X, Liu Y, Li Y, Du J, Gao S. KLF15 maintains contractile phenotype of vascular smooth muscle cells and prevents thoracic aortic dissection by interacting with MRTFB. J Biol Chem 2024; 300:107260. [PMID: 38582447 PMCID: PMC11061230 DOI: 10.1016/j.jbc.2024.107260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/08/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a highly dangerous cardiovascular disorder caused by weakening of the aortic wall, resulting in a sudden tear of the internal face. Progressive loss of the contractile apparatus in vascular smooth muscle cells (VSMCs) is a major event in TAD. Exploring the endogenous regulators essential for the contractile phenotype of VSMCs may aid the development of strategies to prevent TAD. Krüppel-like factor 15 (KLF15) overexpression was reported to inhibit TAD formation; however, the mechanisms by which KLF15 prevents TAD formation and whether KLF15 regulates the contractile phenotype of VSMCs in TAD are not well understood. Therefore, we investigated these unknown aspects of KLF15 function. We found that KLF15 expression was reduced in human TAD samples and β-aminopropionitrile monofumarate-induced TAD mouse model. Klf15KO mice are susceptible to both β-aminopropionitrile monofumarate- and angiotensin II-induced TAD. KLF15 deficiency results in reduced VSMC contractility and exacerbated vascular inflammation and extracellular matrix degradation. Mechanistically, KLF15 interacts with myocardin-related transcription factor B (MRTFB), a potent serum response factor coactivator that drives contractile gene expression. KLF15 silencing represses the MRTFB-induced activation of contractile genes in VSMCs. Thus, KLF15 cooperates with MRTFB to promote the expression of contractile genes in VSMCs, and its dysfunction may exacerbate TAD. These findings indicate that KLF15 may be a novel therapeutic target for the treatment of TAD.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/genetics
- Aortic Aneurysm, Thoracic/pathology
- Dissection, Thoracic Aorta
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Contraction/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Transcription Factors/metabolism
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Guangming Fang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yexuan Tian
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Shan Huang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Xiaoping Zhang
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yan Liu
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Yulin Li
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China
| | - Jie Du
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China.
| | - Shijuan Gao
- Collaborative Innovation Centre for Cardiovascular Disorders, The Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Klessinger D, Mamazhakypov A, Glaeser S, Emig R, Peyronnet R, Meier L, Proelss K, Marenne K, Smolka C, Grundmann S, Pankratz F, Esser PR, Moser M, Zhou Q, Esser JS. Divergent and Compensatory Effects of BMP2 and BMP4 on the VSMC Phenotype and BMP4's Role in Thoracic Aortic Aneurysm Development. Cells 2024; 13:735. [PMID: 38727271 PMCID: PMC11083443 DOI: 10.3390/cells13090735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.
Collapse
MESH Headings
- Animals
- Male
- Mice
- Angiotensin II/pharmacology
- Aortic Aneurysm, Thoracic/metabolism
- Aortic Aneurysm, Thoracic/pathology
- Aortic Aneurysm, Thoracic/genetics
- Apoptosis/drug effects
- Bone Morphogenetic Protein 2/metabolism
- Bone Morphogenetic Protein 4/metabolism
- Disease Models, Animal
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Signal Transduction
Collapse
Affiliation(s)
- Daniel Klessinger
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Argen Mamazhakypov
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg im Breisgau, Germany;
| | - Sophie Glaeser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany; (R.E.); (R.P.)
- CIBSS Centre for Integrative Biological Signalling Studies, Faculty of Biology, University of Freiburg, 79104 Freiburg im Breisgau, Germany
| | - Remi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany; (R.E.); (R.P.)
| | - Lena Meier
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Kora Proelss
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Katia Marenne
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Christian Smolka
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Sebastian Grundmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Franziska Pankratz
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Philipp R. Esser
- Allergy Research Group, Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany;
| | - Martin Moser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| | - Qian Zhou
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Jennifer S. Esser
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg im Breisgau, Germany (C.S.); (S.G.); (F.P.); (M.M.); (Q.Z.)
| |
Collapse
|
6
|
Yang S, Li HW, Tian JY, Wang ZK, Chen Y, Zhan TT, Ma CY, Feng M, Cao SF, Zhao Y, Li X, Ren J, Liu Q, Jin LY, Wang ZQ, Jiang WY, Zhao YX, Zhang Y, Liu X. Myeloid-derived growth factor suppresses VSMC dedifferentiation and attenuates postinjury neointimal formation in rats by activating S1PR2 and its downstream signaling. Acta Pharmacol Sin 2024; 45:98-111. [PMID: 37726422 PMCID: PMC10770085 DOI: 10.1038/s41401-023-01155-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/13/2023] [Indexed: 09/21/2023]
Abstract
Restenosis after angioplasty is caused usually by neointima formation characterized by aberrant vascular smooth muscle cell (VSMC) dedifferentiation. Myeloid-derived growth factor (MYDGF), secreted from bone marrow-derived monocytes and macrophages, has been found to have cardioprotective effects. In this study we investigated the effect of MYDGF to postinjury neointimal formation and the underlying mechanisms. Rat carotid arteries balloon-injured model was established. We found that plasma MYDGF content and the level of MYDGF in injured arteries were significantly decreased after balloon injury. Local application of exogenous MYDGF (50 μg/mL) around the injured vessel during balloon injury markedly ameliorated the development of neointimal formation evidenced by relieving the narrow endovascular diameter, improving hemodynamics, and reducing collagen deposition. In addition, local application of MYDGF inhibited VSMC dedifferentiation, which was proved by reversing the elevated levels of osteopontin (OPN) protein and decreased levels of α-smooth muscle actin (α-SMA) in the left carotid arteries. We showed that PDGF-BB (30 ng/mL) stimulated VSMC proliferation, migration and dedifferentiation in vitro; pretreatment with MYDGF (50-200 ng/mL) concentration-dependently eliminated PDGF-BB-induced cell proliferation, migration and dedifferentiation. Molecular docking revealed that MYDGF had the potential to bind with sphingosine-1-phosphate receptor 2 (S1PR2), which was confirmed by SPR assay and Co-IP analysis. Pretreatment with CCG-1423 (Rho signaling inhibitor), JTE-013 (S1PR2 antagonist) or Ripasudil (ROCK inhibitor) circumvented the inhibitory effects of MYDGF on VSMC phenotypic switching through inhibiting S1PR2 or its downstream RhoA-actin monomers (G-actin) /actin filaments (F-actin)-MRTF-A signaling. In summary, this study proves that MYDGF relieves neointimal formation of carotid arteries in response to balloon injury in rats, and suppresses VSMC dedifferentiation induced by PDGF-BB via S1PR2-RhoA-G/F-actin-MRTF-A signaling pathway. In addition, our results provide evidence for cross talk between bone marrow and vasculature.
Collapse
Affiliation(s)
- Shuang Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Hou-Wei Li
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Jia-Ying Tian
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Zheng-Kai Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yi Chen
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Ting-Ting Zhan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Chun-Yue Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Min Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Shi-Feng Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yu Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Xue Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Jing Ren
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Qian Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Lu-Ying Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Zhi-Qi Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Wen-Yu Jiang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yi-Xiu Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China.
| | - Xue Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, National-Local Joint Engineering Laboratory for Drug Research and Development of Cardio-Cerebrovascular Diseases in Frigid Zone, the National Development and Reform Commission, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150086, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin, 150086, China.
| |
Collapse
|
7
|
Meijer E, Giles R, van Dijk CGM, Maringanti R, Wissing TB, Appels Y, Chrifi I, Crielaard H, Verhaar MC, Smits AI, Cheng C. Effect of Mechanical Stimuli on the Phenotypic Plasticity of Induced Pluripotent Stem-Cell-Derived Vascular Smooth Muscle Cells in a 3D Hydrogel. ACS APPLIED BIO MATERIALS 2023; 6:5716-5729. [PMID: 38032545 PMCID: PMC10731661 DOI: 10.1021/acsabm.3c00840] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023]
Abstract
Introduction: Vascular smooth muscle cells (VSMCs) play a pivotal role in vascular homeostasis, with dysregulation leading to vascular complications. Human-induced pluripotent stem-cell (hiPSC)-derived VSMCs offer prospects for personalized disease modeling and regenerative strategies. Current research lacks comparative studies on the impact of three-dimensional (3D) substrate properties under cyclic strain on phenotypic adaptation in hiPSC-derived VSMCs. Here, we aim to investigate the impact of intrinsic substrate properties, such as the hydrogel's elastic modulus and cross-linking density in a 3D static and dynamic environment, on the phenotypical adaptation of human mural cells derived from hiPSC-derived organoids (ODMCs), compared to aortic VSMCs. Methods and results: ODMCs were cultured in two-dimensional (2D) conditions with synthetic or contractile differentiation medium or in 3D Gelatin Methacryloyl (GelMa) substrates with varying degrees of functionalization and percentages to modulate Young's modulus and cross-linking density. Cells in 3D substrates were exposed to cyclic, unidirectional strain. Phenotype characterization was conducted using specific markers through immunofluorescence and gene expression analysis. Under static 2D culture, ODMCs derived from hiPSCs exhibited a VSMC phenotype, expressing key mural markers, and demonstrated a level of phenotypic plasticity similar to primary human VSMCs. In static 3D culture, a substrate with a higher Young's modulus and cross-linking density promoted a contractile phenotype in ODMCs and VSMCs. Dynamic stimulation in the 3D substrate promoted a switch toward a contractile phenotype in both cell types. Conclusion: Our study demonstrates phenotypic plasticity of human ODMCs in response to 2D biological and 3D mechanical stimuli that equals that of primary human VSMCs. These findings may contribute to the advancement of tailored approaches for vascular disease modeling and regenerative strategies.
Collapse
Affiliation(s)
- Elana
M. Meijer
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Rachel Giles
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Christian G. M. van Dijk
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ranganath Maringanti
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Tamar B. Wissing
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Ymke Appels
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Ihsan Chrifi
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Hanneke Crielaard
- Department
of Biomedical Engineering, Erasmus Medical
Center, Rotterdam 3000 CA, The Netherlands
| | - Marianne C. Verhaar
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
| | - Anthal I.P.M. Smits
- Department
of Biomedical Engineering, Eindhoven University
of Technology; Eindhoven 5612 AE, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology; Eindhoven 5612 AE, The Netherlands
| | - Caroline Cheng
- Department
of Nephrology and Hypertension, Division of Internal Medicine and
Dermatology, University Medical Center Utrecht, Utrecht 3508 GA, The Netherlands
- Regenerative
Medicine Center Utrecht, University Medical
Center Utrecht, Utrecht 3508 GA, The Netherlands
- Experimental
Cardiology, Department of Cardiology, Thorax
Center Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| |
Collapse
|
8
|
Wang L, Moonen JR, Cao A, Isobe S, Li CG, Tojais NF, Taylor S, Marciano DP, Chen PI, Gu M, Li D, Harper RL, El-Bizri N, Kim Y, Stankunas K, Rabinovitch M. Dysregulated Smooth Muscle Cell BMPR2-ARRB2 Axis Causes Pulmonary Hypertension. Circ Res 2023; 132:545-564. [PMID: 36744494 PMCID: PMC10008520 DOI: 10.1161/circresaha.121.320541] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/26/2023] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Mutations in BMPR2 (bone morphogenetic protein receptor 2) are associated with familial and sporadic pulmonary arterial hypertension (PAH). The functional and molecular link between loss of BMPR2 in pulmonary artery smooth muscle cells (PASMC) and PAH pathogenesis warrants further investigation, as most investigations focus on BMPR2 in pulmonary artery endothelial cells. Our goal was to determine whether and how decreased BMPR2 is related to the abnormal phenotype of PASMC in PAH. METHODS SMC-specific Bmpr2-/- mice (BKOSMC) were created and compared to controls in room air, after 3 weeks of hypoxia as a second hit, and following 4 weeks of normoxic recovery. Echocardiography, right ventricular systolic pressure, and right ventricular hypertrophy were assessed as indices of pulmonary hypertension. Proliferation, contractility, gene and protein expression of PASMC from BKOSMC mice, human PASMC with BMPR2 reduced by small interference RNA, and PASMC from PAH patients with a BMPR2 mutation were compared to controls, to investigate the phenotype and underlying mechanism. RESULTS BKOSMC mice showed reduced hypoxia-induced vasoconstriction and persistent pulmonary hypertension following recovery from hypoxia, associated with sustained muscularization of distal pulmonary arteries. PASMC from mutant compared to control mice displayed reduced contractility at baseline and in response to angiotensin II, increased proliferation and apoptosis resistance. Human PASMC with reduced BMPR2 by small interference RNA, and PASMC from PAH patients with a BMPR2 mutation showed a similar phenotype related to upregulation of pERK1/2 (phosphorylated extracellular signal related kinase 1/2)-pP38-pSMAD2/3 mediating elevation in ARRB2 (β-arrestin2), pAKT (phosphorylated protein kinase B) inactivation of GSK3-beta, CTNNB1 (β-catenin) nuclear translocation and reduction in RHOA (Ras homolog family member A) and RAC1 (Ras-related C3 botulinum toxin substrate 1). Decreasing ARRB2 in PASMC with reduced BMPR2 restored normal signaling, reversed impaired contractility and attenuated heightened proliferation and in mice with inducible loss of BMPR2 in SMC, decreasing ARRB2 prevented persistent pulmonary hypertension. CONCLUSIONS Agents that neutralize the elevated ARRB2 resulting from loss of BMPR2 in PASMC could prevent or reverse the aberrant hypocontractile and hyperproliferative phenotype of these cells in PAH.
Collapse
Affiliation(s)
- Lingli Wang
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Jan Renier Moonen
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Aiqin Cao
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Sarasa Isobe
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Caiyun G Li
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Nancy F Tojais
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Shalina Taylor
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - David P Marciano
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Pin-I Chen
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxia Gu
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Dan Li
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Rebecca L Harper
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Nesrine El-Bizri
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - YuMee Kim
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| | - Kryn Stankunas
- Departments of Pathology and of Developmental Biology, and Howard Hughes Medical Institute; Stanford University School of Medicine, Stanford, CA, USA
| | - Marlene Rabinovitch
- BASE Initiative, Betty Irene Moore Children’s Heart Center, Lucile Packard Children’s Hospital
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
BMPER Improves Vascular Remodeling and the Contractile Vascular SMC Phenotype. Int J Mol Sci 2023; 24:ijms24054950. [PMID: 36902380 PMCID: PMC10002482 DOI: 10.3390/ijms24054950] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 02/19/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Dedifferentiated vascular smooth muscle cells (vSMCs) play an essential role in neointima formation, and we now aim to investigate the role of the bone morphogenetic protein (BMP) modulator BMPER (BMP endothelial cell precursor-derived regulator) in neointima formation. To assess BMPER expression in arterial restenosis, we used a mouse carotid ligation model with perivascular cuff placement. Overall BMPER expression after vessel injury was increased; however, expression in the tunica media was decreased compared to untreated control. Consistently, BMPER expression was decreased in proliferative, dedifferentiated vSMC in vitro. C57BL/6_Bmper+/- mice displayed increased neointima formation 21 days after carotid ligation and enhanced expression of Col3A1, MMP2, and MMP9. Silencing of BMPER increased the proliferation and migration capacity of primary vSMCs, as well as reduced contractibility and expression of contractile markers, whereas stimulation with recombinant BMPER protein had the opposite effect. Mechanistically, we showed that BMPER binds insulin-like growth factor-binding protein 4 (IGFBP4), resulting in the modulation of IGF signaling. Furthermore, perivascular application of recombinant BMPER protein prevented neointima formation and ECM deposition in C57BL/6N mice after carotid ligation. Our data demonstrate that BMPER stimulation causes a contractile vSMC phenotype and suggest that BMPER has the potential for a future therapeutic agent in occlusive cardiovascular diseases.
Collapse
|
10
|
Kim Y, Yu N, Jang YE, Lee E, Jung Y, Lee DJ, Taylor WR, Jo H, Kim J, Lee S, Kang SW. Conserved miR-370-3p/BMP-7 axis regulates the phenotypic change of human vascular smooth muscle cells. Sci Rep 2023; 13:2404. [PMID: 36765143 PMCID: PMC9918535 DOI: 10.1038/s41598-022-26711-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/19/2022] [Indexed: 02/12/2023] Open
Abstract
Endothelial dysfunction and inflammatory immune response trigger dedifferentiation of vascular smooth muscle cells (SMCs) from contractile to synthetic phenotype and initiate arterial occlusion. However, the complex vascular remodeling process playing roles in arterial occlusion initiation is largely unknown. We performed bulk sequencing of small and messenger RNAs in a rodent arterial injury model. Bioinformatic data analyses reveal that six miRNAs are overexpressed in injured rat carotids as well as synthetic-type human vascular SMCs. In vitro cell-based assays show that four miRNAs (miR-130b-5p, miR-132-3p, miR-370-3p, and miR-410-3p) distinctly regulate the proliferation of and monocyte adhesion to the vascular SMCs. Individual inhibition of the four selected miRNAs strongly prevents the neointimal hyperplasia in the injured rat carotid arteries. Mechanistically, miR-132-3p and miR-370-3p direct the cell cycle progression, triggering SMC proliferation. Gene ontology analysis of mRNA sequencing data consistently reveal that the miRNA targets include gene clusters that direct proliferation, differentiation, and inflammation. Notably, bone morphogenic protein (BMP)-7 is a prominent target gene of miR-370-3p, and it regulates vascular SMC proliferation in cellular and animal models. Overall, this study first reports that the miR-370-3p/BMP-7 axis determines the vascular SMC phenotype in both rodent and human systems.
Collapse
Affiliation(s)
- Yerin Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Namhee Yu
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
- Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Ye Eun Jang
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eunkyung Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Yeonjoo Jung
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Doo Jae Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - W Robert Taylor
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Jaesang Kim
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sanghyuk Lee
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
| | - Sang Won Kang
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
11
|
Yeo Y, Jeong H, Kim M, Choi Y, Kim KL, Suh W. Crosstalk between BMP signaling and KCNK3 in phenotypic switching of pulmonary vascular smooth muscle cells. BMB Rep 2022; 55:565-570. [PMID: 36016502 PMCID: PMC9712703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/11/2022] [Accepted: 08/12/2022] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and devastating disease whose pathogenesis is associated with a phenotypic switch of pulmonary arterial vascular smooth muscle cells (PASMCs). Bone morphogenetic protein (BMP) signaling and potassium two pore domain channel subfamily K member 3 (KCNK3) play crucial roles in PAH pathogenesis. However, the relationship between BMP signaling and KCNK3 expression in the PASMC phenotypic switching process has not been studied. In this study, we explored the effect of BMPs on KCNK3 expression and the role of KCNK3 in the BMP-mediated PASMC phenotypic switch. Expression levels of BMP receptor 2 (BMPR2) and KCNK3 were downregulated in PASMCs of rats with PAH compared to those in normal controls, implying a possible association between BMP/BMPR2 signaling and KCNK3 expression in the pulmonary vasculature. Treatment with BMP2, BMP4, and BMP7 significantly increased KCNK3 expression in primary human PASMCs (HPASMCs). BMPR2 knockdown and treatment with Smad1/5 signaling inhibitor substantially abrogated the BMP-induced increase in KCNK3 expression, suggesting that KCNK3 expression in HPASMCs is regulated by the canonical BMP-BMPR2-Smad1/5 signaling pathway. Furthermore, KCNK3 knockdown and treatment with a KCNK3 channel blocker completely blocked BMP-mediated anti-proliferation and expression of contractile marker genes in HPAMSCs, suggesting that the expression and functional activity of KCNK3 are required for BMP-mediated acquisition of the quiescent PASMC phenotype. Overall, our findings show a crosstalk between BMP signaling and KCNK3 in regulating the PASMC phenotype, wherein BMPs upregulate KCNK3 expression and KCNK3 then mediates BMP-induced phenotypic switching of PASMCs. Our results indicate that the dysfunction and/or downregulation of BMPR2 and KCNK3 observed in PAH work together to induce aberrant changes in the PASMC phenotype, providing insights into the complex molecular pathogenesis of PAH. [BMB Reports 2022; 55(11): 565-570].
Collapse
Affiliation(s)
- Yeongju Yeo
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Hayoung Jeong
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Minju Kim
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Yanghee Choi
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Koung Li Kim
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Wonhee Suh
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
12
|
Yeo Y, Jeong H, Kim M, Choi Y, Kim KL, Suh W. Crosstalk between BMP signaling and KCNK3 in phenotypic switching of pulmonary vascular smooth muscle cells. BMB Rep 2022; 55:565-570. [PMID: 36016502 PMCID: PMC9712703 DOI: 10.5483/bmbrep.2022.55.11.098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/11/2022] [Accepted: 08/12/2022] [Indexed: 02/18/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and devastating disease whose pathogenesis is associated with a phenotypic switch of pulmonary arterial vascular smooth muscle cells (PASMCs). Bone morphogenetic protein (BMP) signaling and potassium two pore domain channel subfamily K member 3 (KCNK3) play crucial roles in PAH pathogenesis. However, the relationship between BMP signaling and KCNK3 expression in the PASMC phenotypic switching process has not been studied. In this study, we explored the effect of BMPs on KCNK3 expression and the role of KCNK3 in the BMP-mediated PASMC phenotypic switch. Expression levels of BMP receptor 2 (BMPR2) and KCNK3 were downregulated in PASMCs of rats with PAH compared to those in normal controls, implying a possible association between BMP/BMPR2 signaling and KCNK3 expression in the pulmonary vasculature. Treatment with BMP2, BMP4, and BMP7 significantly increased KCNK3 expression in primary human PASMCs (HPASMCs). BMPR2 knockdown and treatment with Smad1/5 signaling inhibitor substantially abrogated the BMP-induced increase in KCNK3 expression, suggesting that KCNK3 expression in HPASMCs is regulated by the canonical BMP-BMPR2-Smad1/5 signaling pathway. Furthermore, KCNK3 knockdown and treatment with a KCNK3 channel blocker completely blocked BMP-mediated anti-proliferation and expression of contractile marker genes in HPAMSCs, suggesting that the expression and functional activity of KCNK3 are required for BMP-mediated acquisition of the quiescent PASMC phenotype. Overall, our findings show a crosstalk between BMP signaling and KCNK3 in regulating the PASMC phenotype, wherein BMPs upregulate KCNK3 expression and KCNK3 then mediates BMP-induced phenotypic switching of PASMCs. Our results indicate that the dysfunction and/or downregulation of BMPR2 and KCNK3 observed in PAH work together to induce aberrant changes in the PASMC phenotype, providing insights into the complex molecular pathogenesis of PAH. [BMB Reports 2022; 55(11): 565-570].
Collapse
Affiliation(s)
- Yeongju Yeo
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Hayoung Jeong
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Minju Kim
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Yanghee Choi
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Koung Li Kim
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| | - Wonhee Suh
- Department of Global Innovative Drug, The Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
13
|
Deshpande A, Shetty PMV, Frey N, Rangrez AY. SRF: a seriously responsible factor in cardiac development and disease. J Biomed Sci 2022; 29:38. [PMID: 35681202 PMCID: PMC9185982 DOI: 10.1186/s12929-022-00820-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
The molecular mechanisms that regulate embryogenesis and cardiac development are calibrated by multiple signal transduction pathways within or between different cell lineages via autocrine or paracrine mechanisms of action. The heart is the first functional organ to form during development, which highlights the importance of this organ in later stages of growth. Knowledge of the regulatory mechanisms underlying cardiac development and adult cardiac homeostasis paves the way for discovering therapeutic possibilities for cardiac disease treatment. Serum response factor (SRF) is a major transcription factor that controls both embryonic and adult cardiac development. SRF expression is needed through the duration of development, from the first mesodermal cell in a developing embryo to the last cell damaged by infarction in the myocardium. Precise regulation of SRF expression is critical for mesoderm formation and cardiac crescent formation in the embryo, and altered SRF levels lead to cardiomyopathies in the adult heart, suggesting the vital role played by SRF in cardiac development and disease. This review provides a detailed overview of SRF and its partners in their various functions and discusses the future scope and possible therapeutic potential of SRF in the cardiovascular system.
Collapse
Affiliation(s)
- Anushka Deshpande
- Department of Internal Medicine III, Cardiology and Angiology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany.,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Prithviraj Manohar Vijaya Shetty
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
14
|
Chen J, Rodriguez M, Miao J, Liao J, Jain PP, Zhao M, Zhao T, Babicheva A, Wang Z, Parmisano S, Powers R, Matti M, Paquin C, Soroureddin Z, Shyy JYJ, Thistlethwaite PA, Makino A, Wang J, Yuan JXJ. Mechanosensitive channel Piezo1 is required for pulmonary artery smooth muscle cell proliferation. Am J Physiol Lung Cell Mol Physiol 2022; 322:L737-L760. [PMID: 35318857 PMCID: PMC9076422 DOI: 10.1152/ajplung.00447.2021] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/10/2022] [Accepted: 03/17/2022] [Indexed: 01/10/2023] Open
Abstract
Concentric pulmonary vascular wall thickening due partially to increased pulmonary artery (PA) smooth muscle cell (PASMC) proliferation contributes to elevating pulmonary vascular resistance (PVR) in patients with pulmonary hypertension (PH). Although pulmonary vasoconstriction may be an early contributor to increasing PVR, the transition of contractile PASMCs to proliferative PASMCs may play an important role in the development and progression of pulmonary vascular remodeling in PH. A rise in cytosolic Ca2+ concentration ([Ca2+]cyt) is a trigger for PASMC contraction and proliferation. Here, we report that upregulation of Piezo1, a mechanosensitive cation channel, is involved in the contractile-to-proliferative phenotypic transition of PASMCs and potential development of pulmonary vascular remodeling. By comparing freshly isolated PA (contractile PASMCs) and primary cultured PASMCs (from the same rat) in a growth medium (proliferative PASMCs), we found that Piezo1, Notch2/3, and CaSR protein levels were significantly higher in proliferative PASMCs than in contractile PASMCs. Upregulated Piezo1 was associated with an increase in expression of PCNA, a marker for cell proliferation, whereas downregulation (with siRNA) or inhibition (with GsMTx4) of Piezo1 attenuated PASMC proliferation. Furthermore, Piezo1 in the remodeled PA from rats with experimental PH was upregulated compared with PA from control rats. These data indicate that PASMC contractile-to-proliferative phenotypic transition is associated with the transition or adaptation of membrane channels and receptors. Upregulated Piezo1 may play a critical role in PASMC phenotypic transition and PASMC proliferation. Upregulation of Piezo1 in proliferative PASMCs may likely be required to provide sufficient Ca2+ to assure nuclear/cell division and PASMC proliferation, contributing to the development and progression of pulmonary vascular remodeling in PH.
Collapse
Affiliation(s)
- Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jinrui Miao
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing Liao
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Pritesh P Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Manjia Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ziyi Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Ryan Powers
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Moreen Matti
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Cole Paquin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Zahra Soroureddin
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Disease and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jason X-J Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
15
|
Lim L, Ki YJ, Kim H, Chu B, Choi IY, Choi DH, Song H. Plantamajoside Attenuates Neointima Formation via Upregulation of Tissue Inhibitor of Metalloproteinases in Balloon-Injured Rats. J Med Food 2022; 25:503-512. [PMID: 35483086 DOI: 10.1089/jmf.2021.k.0162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The abnormal change of vascular smooth muscle cell (VSMC) behavior is an important cellular event leading to neointimal hyperplasia in atherosclerosis and restenosis. Plantamajoside (PMS), a phenylethanoid glycoside compound of the Plantago asiatica, has been reported to have anti-inflammatory, antioxidative, and anticancer activities. In this study, the protective effects of PMS against intimal hyperplasia and the mechanisms underlying the regulation of VSMC behavior were investigated. MTT and BrdU assays were performed to evaluate the cytotoxicity and cell proliferative activity of PMS, respectively. Rat aortic VSMC migrations after treatment with the determined concentration of PMS (50 and 150 μM) were evaluated using wound healing and Boyden chamber assays. The inhibitory effects of PMS on intimal hyperplasia were evaluated in balloon-injured (BI) rat carotid artery. PMS suppressed the proliferation in platelet-derived growth factor-BB-induced VSMC, as confirmed from the decrease in cyclin-dependent kinase (CDK)-2, CDK-4, cyclin D1, and proliferating cell nuclear antigen levels. PMS also inhibited VSMC migration, consistent with the downregulated expression and zymolytic activities of matrix metalloproteinase (MMP)2, MMP9, and MMP13. PMS specifically regulated MMP expression through p38 mitogen-activated protein kinase and focal adhesion kinase pathways. Tissue inhibitor of metalloproteinase (TIMP)1 and TIMP2 levels were upregulated via Smad1. TIMPs inhibited the conversion of pro-MMPs to active MMPs. PMS significantly inhibited neointimal formation in BI rat carotid arteries. In conclusion, PMS inhibits VSMC proliferation and migration by upregulating TIMP1 and TIMP2 expression. Therefore, PMS could be a potential therapeutic agent for vascular atherosclerosis and restenosis treatment.
Collapse
Affiliation(s)
- Leejin Lim
- Cancer Mutation Research Center, Chosun University, Gwangju, Korea
| | - Young-Jae Ki
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Hyeonhwa Kim
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea
| | - Byeongsam Chu
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea
| | - In Young Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Dong-Hyun Choi
- Department of Internal Medicine, Chosun University School of Medicine, Gwangju, Korea
| | - Heesang Song
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju, Korea.,Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju, Korea
| |
Collapse
|
16
|
Kabwe JC, Sawada H, Mitani Y, Oshita H, Tsuboya N, Zhang E, Maruyama J, Miyasaka Y, Ko H, Oya K, Ito H, Yodoya N, Otsuki S, Ohashi H, Okamoto R, Dohi K, Nishimura Y, Mashimo T, Hirayama M, Maruyama K. CRISPR-mediated Bmpr2 point mutation exacerbates late pulmonary vasculopathy and reduces survival in rats with experimental pulmonary hypertension. Respir Res 2022; 23:87. [PMID: 35395852 PMCID: PMC8994407 DOI: 10.1186/s12931-022-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/24/2022] [Indexed: 11/23/2022] Open
Abstract
Background Patients with pulmonary arterial hypertension (PAH) carrying bone morphogenetic protein receptor type 2 (Bmpr2) mutations present earlier with severe hemodynamic compromise and have poorer survival outcomes than those without mutation. The mechanism underlying the worsening clinical phenotype of PAH with Bmpr2 mutations has been largely unaddressed in rat models of pulmonary hypertension (PH) because of the difficulty in reproducing progressive PH in mice and genetic modification in rats. We tested whether a clinically-relevant Bmpr2 mutation affects the progressive features of monocrotaline (MCT) induced-PH in rats. Methods A monoallelic single nucleotide insertion in exon 1 of Bmpr2 (+/44insG) was generated in rats using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9, then PH, pulmonary vascular disease (PVD) and survival after MCT injection with or without a phosphodiesterase type 5 inhibitor, tadalafil, administration were assessed. Results The +/44insG rats had reduced BMPR2 signalling in the lungs compared with wild-type. PH and PVD assessed at 3-weeks after MCT injection were similar in wild-type and +/44insG rats. However, survival at 4-weeks after MCT injection was significantly reduced in +/44insG rats. Among the rats surviving at 4-weeks after MCT administration, +/44insG rats had increased weight ratio of right ventricle to left ventricle plus septum (RV/[LV + S]) and % medial wall thickness (MWT) in pulmonary arteries (PAs). Immunohistochemical analysis showed increased vessels with Ki67-positive cells in the lungs, decreased mature and increased immature smooth muscle cell phenotype markers in the PAs in +/44insG rats compared with wild-type at 3-weeks after MCT injection. Contraction of PA in response to prostaglandin-F2α and endothelin-1 were significantly reduced in the +/44insG rats. The +/44insG rats that had received tadalafil had a worse survival with a significant increase in RV/(LV + S), %MWT in distal PAs and RV myocardial fibrosis compared with wild-type. Conclusions The present study demonstrates that the Bmpr2 mutation promotes dedifferentiation of PA smooth muscle cells, late PVD and RV myocardial fibrosis and adversely impacts both the natural and post-treatment courses of MCT-PH in rats with significant effects only in the late stages and warrants preclinical studies using this new genetic model to optimize treatment outcomes of heritable PAH. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02005-w.
Collapse
Affiliation(s)
- Jane Chanda Kabwe
- The Department of Anesthesiology and Critical Care Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu city, Mie, 514-8507, Japan
| | - Hirofumi Sawada
- The Department of Anesthesiology and Critical Care Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu city, Mie, 514-8507, Japan. .,The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan.
| | - Yoshihide Mitani
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Hironori Oshita
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan.,The Department of Pediatrics, Nagoya City University School of Medicine, Aichi, Japan
| | - Naoki Tsuboya
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Erquan Zhang
- The Department of Anesthesiology and Critical Care Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu city, Mie, 514-8507, Japan.,The Department of Neonatology, Fuzhou Children's Hospital of Fujian Province, Fujian Medical University, Fujian, China
| | - Junko Maruyama
- The Department of Clinical Engineering, Suzuka University of Medical Science, Mie, Japan
| | - Yoshiki Miyasaka
- Institute of Experimental Animal Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hideyoshi Ko
- The Department of Clinical Engineering, Suzuka University of Medical Science, Mie, Japan
| | - Kazunobu Oya
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiromasa Ito
- The Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Noriko Yodoya
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Shoichiro Otsuki
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroyuki Ohashi
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Ryuji Okamoto
- The Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Kaoru Dohi
- The Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Yuhei Nishimura
- The Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Mie, Japan
| | - Tomoji Mashimo
- Institute of Experimental Animal Sciences, Osaka University Graduate School of Medicine, Osaka, Japan.,Laboratory Animal Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masahiro Hirayama
- The Department of Pediatrics, Mie University Graduate School of Medicine, Mie, Japan
| | - Kazuo Maruyama
- The Department of Anesthesiology and Critical Care Medicine, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu city, Mie, 514-8507, Japan
| |
Collapse
|
17
|
Beck-Joseph J, Tabrizian M, Lehoux S. Molecular Interactions Between Vascular Smooth Muscle Cells and Macrophages in Atherosclerosis. Front Cardiovasc Med 2021; 8:737934. [PMID: 34722670 PMCID: PMC8554018 DOI: 10.3389/fcvm.2021.737934] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis is the largest contributor toward life-threatening cardiovascular events. Cellular activity and cholesterol accumulation lead to vascular remodeling and the formation of fatty plaques. Complications arise from blood clots, forming at sites of plaque development, which may detach and result in thrombotic occlusions. Vascular smooth muscle cells and macrophages play dominant roles in atherosclerosis. A firm understanding of how these cells influence and modulate each other is pivotal for a better understanding of the disease and the development of novel therapeutics. Recent studies have investigated molecular interactions between both cell types and their impact on disease progression. Here we aim to review the current knowledge. Intercellular communications through soluble factors, physical contact, and extracellular vesicles are discussed. We also present relevant background on scientific methods used to study the disease, the general pathophysiology and intracellular factors involved in phenotypic modulation of vascular smooth muscle cells. We conclude this review with a discussion of the current state, shortcomings and potential future directions of the field.
Collapse
Affiliation(s)
- Jahnic Beck-Joseph
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Maryam Tabrizian
- Biomat'X Research Laboratories, Department of Biomedical Engineering, McGill University, Montreal, QC, Canada
| | - Stephanie Lehoux
- Department of Medicine, Lady Davis Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
18
|
Childs BG, Zhang C, Shuja F, Sturmlechner I, Trewartha S, Fierro Velasco R, Baker D, Li H, van Deursen JM. Senescent cells suppress innate smooth muscle cell repair functions in atherosclerosis. NATURE AGING 2021; 1:698-714. [PMID: 34746803 PMCID: PMC8570576 DOI: 10.1038/s43587-021-00089-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Senescent cells (SNCs) degenerate the fibrous cap that normally prevents atherogenic plaque rupture, a leading cause of myocardial infarction and stroke. Here we explored the underlying mechanism using pharmacological or transgenic approaches to clear SNCs in the Ldlr -/- mouse model of atherosclerosis. SNC clearance reinforced fully deteriorated fibrous caps in highly advanced lesions, as evidenced by restored vascular smooth muscle cell (VSMC) numbers, elastin content, and overall cap thickness. We found that SNCs inhibit VSMC promigratory phenotype switching in the first interfiber space of the arterial wall directly beneath atherosclerotic plaque, thereby limiting lesion entry of medial VSMCs for fibrous cap assembly or reinforcement. SNCs do so by antagonizing IGF-1 through the secretion of insulin-like growth factor-binding protein 3 (Igfbp3). These data indicate that the intermittent use of senolytic agents or IGFBP-3 inhibition in combination with lipid lowering drugs may provide therapeutic benefit in atherosclerosis.
Collapse
Affiliation(s)
- Bennett G. Childs
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester MN, United States
| | - Fahad Shuja
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Rochester MN, United States
| | - Ines Sturmlechner
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Molecular Genetics Section, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Shawn Trewartha
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Raul Fierro Velasco
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Darren Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester MN, United States
| | - Jan M. van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
- Correspondence:
| |
Collapse
|
19
|
Yogi A, Rukhlova M, Charlebois C, Tian G, Stanimirovic DB, Moreno MJ. Differentiation of Adipose-Derived Stem Cells into Vascular Smooth Muscle Cells for Tissue Engineering Applications. Biomedicines 2021; 9:biomedicines9070797. [PMID: 34356861 PMCID: PMC8301460 DOI: 10.3390/biomedicines9070797] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/15/2021] [Accepted: 06/25/2021] [Indexed: 11/20/2022] Open
Abstract
Synthetic grafts have been developed for vascular bypass surgery, however, the risks of thrombosis and neointimal hyperplasia still limit their use. Tissue engineering with the use of adipose-derived stem cells (ASCs) has shown promise in addressing these limitations. Here we further characterized and optimized the ASC differentiation into smooth muscle cells (VSMCs) induced by TGF-β and BMP-4. TGF-β and BMP-4 induced a time-dependent expression of SMC markers in ASC. Shortening the differentiation period from 7 to 4 days did not impair the functional property of contraction in these cells. Stability of the process was demonstrated by switching cells to regular growth media for up to 14 days. The role of IGFBP7, a downstream effector of TGF-β, was also examined. Finally, topographic and surface patterning of a substrate is recognized as a powerful tool for regulating cell differentiation. Here we provide evidence that a non-woven PET structure does not affect the differentiation of ASC. Taken together, our results indicate that VSMCs differentiated from ASCs are a suitable candidate to populate a PET-based vascular scaffolds. By employing an autologous source of cells we provide a novel alternative to address major issues that reduces long-term patency of currently vascular grafts.
Collapse
Affiliation(s)
- Alvaro Yogi
- Human Health Therapeutics, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A0R6, Canada; (M.R.); (C.C.); (D.B.S.)
- Correspondence: (A.Y.); (M.J.M.); Tel.: +1-613-990-0891 (A.Y.); +1-613-990-0829 (M.J.M.)
| | - Marina Rukhlova
- Human Health Therapeutics, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A0R6, Canada; (M.R.); (C.C.); (D.B.S.)
| | - Claudie Charlebois
- Human Health Therapeutics, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A0R6, Canada; (M.R.); (C.C.); (D.B.S.)
| | - Ganghong Tian
- Medical Devices Research Centre, National Research Council of Canada, 435 Ellice Ave, Winnipeg, MB R3B 1Y6, Canada;
| | - Danica B. Stanimirovic
- Human Health Therapeutics, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A0R6, Canada; (M.R.); (C.C.); (D.B.S.)
| | - Maria J. Moreno
- Human Health Therapeutics, National Research Council of Canada, 1200 Montreal Road, Ottawa, ON K1A0R6, Canada; (M.R.); (C.C.); (D.B.S.)
- Correspondence: (A.Y.); (M.J.M.); Tel.: +1-613-990-0891 (A.Y.); +1-613-990-0829 (M.J.M.)
| |
Collapse
|
20
|
Yang Q, Shi W. Rho/ROCK-MYOCD in regulating airway smooth muscle growth and remodeling. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1-L5. [PMID: 33909498 DOI: 10.1152/ajplung.00034.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Abnormal airway remodeling is a common pathological change seen in chronic respiratory diseases. Altered proliferation and differentiation of airway smooth muscle cells (ASMCs) are the major components of airway remodeling, and the resultant structural abnormalities are difficult to restore. Understanding of airway smooth muscle regulation is urgently needed to identify potential intervention targets. MYOCD (or myocardin) and myocardin-related transcription factors (MRTFs) are key cotranscription factors in muscle growth, which have not been extensively investigated in airway smooth muscle cells. In addition, the RhoA/ROCK signaling pathway is known to play an important role in airway remodeling partly through regulating the proliferation and differentiation of ASMCs, which may be connected with MYOCD/MRTF cotranscription factors [Kumawat et al. (Am J Physiol Lung Cell Mol Physiol 311: L529-L537, 2016); Lagna et al. (J Biol Chem 282: 37244-37255, 2007)]. This review focuses on this newly recognized and potentially important RhoA/ROCK-MYOCD/MRTFs pathway in controlling airway smooth muscle growth and remodeling.
Collapse
Affiliation(s)
- Qin Yang
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California.,Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Wei Shi
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
21
|
Zhao G, Lu H, Chang Z, Zhao Y, Zhu T, Chang L, Guo Y, Garcia-Barrio MT, Chen YE, Zhang J. Single-cell RNA sequencing reveals the cellular heterogeneity of aneurysmal infrarenal abdominal aorta. Cardiovasc Res 2021; 117:1402-1416. [PMID: 32678909 PMCID: PMC8064434 DOI: 10.1093/cvr/cvaa214] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 12/28/2022] Open
Abstract
AIMS The artery contains numerous cell types which contribute to multiple vascular diseases. However, the heterogeneity and cellular responses of these vascular cells during abdominal aortic aneurysm (AAA) progression have not been well characterized. METHODS AND RESULTS Single-cell RNA sequencing was performed on the infrarenal abdominal aortas (IAAs) from C57BL/6J mice at Days 7 and 14 post-sham or peri-adventitial elastase-induced AAA. Unbiased clustering analysis of the transcriptional profiles from >4500 aortic cells identified 17 clusters representing nine-cell lineages, encompassing vascular smooth muscle cells (VSMCs), fibroblasts, endothelial cells, immune cells (macrophages, T cells, B cells, and dendritic cells), and two types of rare cells, including neural cells and erythrocyte cells. Seurat clustering analysis identified four smooth muscle cell (SMC) subpopulations and five monocyte/macrophage subpopulations, with distinct transcriptional profiles. During AAA progression, three major SMC subpopulations were proportionally decreased, whereas the small subpopulation was increased, accompanied with down-regulation of SMC contractile markers and up-regulation of pro-inflammatory genes. Another AAA-associated cellular response is immune cell expansion, particularly monocytes/macrophages. Elastase exposure induced significant expansion and activation of aortic resident macrophages, blood-derived monocytes and inflammatory macrophages. We also identified increased blood-derived reparative macrophages expressing anti-inflammatory cytokines suggesting that resolution of inflammation and vascular repair also persist during AAA progression. CONCLUSION Our data identify AAA disease-relevant transcriptional signatures of vascular cells in the IAA. Furthermore, we characterize the heterogeneity and cellular responses of VSMCs and monocytes/macrophages during AAA progression, which provide insights into their function and the regulation of AAA onset and progression.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/chemically induced
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Cell Lineage
- Cluster Analysis
- Disease Models, Animal
- Gene Expression Profiling
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Monocytes/metabolism
- Monocytes/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Pancreatic Elastase
- Phenotype
- RNA-Seq
- Single-Cell Analysis
- Transcriptome
- Mice
Collapse
Affiliation(s)
- Guizhen Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Haocheng Lu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Ziyi Chang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, PR China
| | - Yang Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Tianqing Zhu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Lin Chang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Yanhong Guo
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Minerva T Garcia-Barrio
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Y Eugene Chen
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, NCRC Bldg26, Room 357S. 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| |
Collapse
|
22
|
Bonetti J, Corti A, Lerouge L, Pompella A, Gaucher C. Phenotypic Modulation of Macrophages and Vascular Smooth Muscle Cells in Atherosclerosis-Nitro-Redox Interconnections. Antioxidants (Basel) 2021; 10:antiox10040516. [PMID: 33810295 PMCID: PMC8066740 DOI: 10.3390/antiox10040516] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Monocytes/macrophages and vascular smooth muscle cells (vSMCs) are the main cell types implicated in atherosclerosis development, and unlike other mature cell types, both retain a remarkable plasticity. In mature vessels, differentiated vSMCs control the vascular tone and the blood pressure. In response to vascular injury and modifications of the local environment (inflammation, oxidative stress), vSMCs switch from a contractile to a secretory phenotype and also display macrophagic markers expression and a macrophagic behaviour. Endothelial dysfunction promotes adhesion to the endothelium of monocytes, which infiltrate the sub-endothelium and differentiate into macrophages. The latter become polarised into M1 (pro-inflammatory), M2 (anti-inflammatory) or Mox macrophages (oxidative stress phenotype). Both monocyte-derived macrophages and macrophage-like vSMCs are able to internalise and accumulate oxLDL, leading to formation of “foam cells” within atherosclerotic plaques. Variations in the levels of nitric oxide (NO) can affect several of the molecular pathways implicated in the described phenomena. Elucidation of the underlying mechanisms could help to identify novel specific therapeutic targets, but to date much remains to be explored. The present article is an overview of the different factors and signalling pathways implicated in plaque formation and of the effects of NO on the molecular steps of the phenotypic switch of macrophages and vSMCs.
Collapse
Affiliation(s)
- Justine Bonetti
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| | - Alessandro Corti
- Department of Translational Research NTMS, University of Pisa Medical School, 56126 Pisa, Italy;
| | - Lucie Lerouge
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| | - Alfonso Pompella
- Department of Translational Research NTMS, University of Pisa Medical School, 56126 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-2218-537
| | - Caroline Gaucher
- CITHEFOR, Université de Lorraine, F-54000 Nancy, France; (J.B.); (L.L.); (C.G.)
| |
Collapse
|
23
|
Yeo Y, Yi ES, Kim JM, Jo EK, Seo S, Kim RI, Kim KL, Sung JH, Park SG, Suh W. FGF12 (Fibroblast Growth Factor 12) Inhibits Vascular Smooth Muscle Cell Remodeling in Pulmonary Arterial Hypertension. Hypertension 2020; 76:1778-1786. [PMID: 33100045 DOI: 10.1161/hypertensionaha.120.15068] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Loss of BMP (bone morphogenic protein) signaling induces a phenotype switch of pulmonary arterial smooth muscle cells (PASMCs), which is the pathological basis of pulmonary vascular remodeling in pulmonary arterial hypertension (PAH). Here, we identified FGF12 (fibroblast growth factor 12) as a novel regulator of the BMP-induced phenotype change in PASMCs and elucidated its role in pulmonary vascular remodeling during PAH development. Using murine models of PAH and lung specimens of patients with PAH, we observed that FGF12 expression was significantly reduced in PASMCs. In human PASMCs, FGF12 expression was increased by canonical BMP signaling. FGF12 knockdown blocked the antiproliferative and prodifferentiation effect of BMP on human PASMCs, suggesting that FGF12 is required for the BMP-mediated acquisition of the quiescent and differentiated PASMC phenotype. Mechanistically, FGF12 regulated the BMP-induced phenotype change by inducing MEF2a (myocyte enhancer factor 2a) phosphorylation via p38MAPK signaling, thereby modulating the expression of MEF2a target genes involved in cell proliferation and differentiation. Furthermore, we observed that TG (transgenic) mice with smooth muscle cell-specific FGF12 overexpression were protected from chronic hypoxia-induced PAH development, pulmonary vascular remodeling, and right ventricular hypertrophy. Consistent with the in vitro data using human PASMCs, FGF12 TG mice showed increased MEF2a phosphorylation and a substantial change in MEF2a target gene expression, compared with the WT (wild type) controls. Overall, our findings demonstrate a novel BMP/FGF12/MEF2a pathway regulating the PASMC phenotype switch and suggest FGF12 as a potential target for the development of therapeutics for ameliorating pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Yeongju Yeo
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Eunhee S Yi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN (E.S.Y.)
| | - Jeong-Min Kim
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Eun-Kyung Jo
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Songyi Seo
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Ryul-I Kim
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Koung Li Kim
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| | - Jong-Hyuk Sung
- College of Pharmacy, Yonsei University, Incheon, Korea (J.-H.S.)
| | - Sang Gyu Park
- College of Pharmacy, Ajou University, Suwon, Korea (S.G.P.)
| | - Wonhee Suh
- From the Department of Global Innovative Drug, Graduate School of Chung-Ang University, College of Pharmacy, Chung-Ang University, Seoul, Korea (Y.Y., J.-M.K., E.-K.J., S.S., R.-I.K., K.L.K., W.S.)
| |
Collapse
|
24
|
da Silva Madaleno C, Jatzlau J, Knaus P. BMP signalling in a mechanical context - Implications for bone biology. Bone 2020; 137:115416. [PMID: 32422297 DOI: 10.1016/j.bone.2020.115416] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/12/2023]
Abstract
Bone Morphogenetic Proteins (BMPs) are extracellular multifunctional signalling cytokines and members of the TGFβ super family. These pleiotropic growth factors crucially promote bone formation, remodeling and healing after injury. Additionally, bone homeostasis is systematically regulated by mechanical inputs from the environment, which are incorporated into the bone cells' biochemical response. These inputs range from compression and tension induced by the movement of neighboring muscle, to fluid shear stress induced by interstitial fluid flow in the canaliculi and in the vascular system. Although BMPs are widely applied in a clinic context to promote fracture healing, it is still elusive how mechanical inputs modulate this signalling pathway, hindering an efficient and side-effect free application of these ligands in bone healing. This review aims to summarize the current understanding in how mechanical cues (tension, compression, shear force and hydrostatic pressure) and substrate stiffness modulate BMP signalling. We highlight the time-dependent effects in modulating immediate early up to long-term effects of mechano-BMP crosstalk during bone formation and remodeling, considering the interplay with other already established mechanosensitive pathways, such as MRTF/SRF and Hippo signalling.
Collapse
Affiliation(s)
- Carolina da Silva Madaleno
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany
| | - Jerome Jatzlau
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; Berlin Brandenburg School of Regenerative Therapies (BSRT), Charité Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
25
|
Park N, Kang H. BMP-Induced MicroRNA-101 Expression Regulates Vascular Smooth Muscle Cell Migration. Int J Mol Sci 2020; 21:ijms21134764. [PMID: 32635504 PMCID: PMC7369869 DOI: 10.3390/ijms21134764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022] Open
Abstract
Proliferation and migration of vascular smooth muscle cells (VSMCs) are implicated in blood vessel development, maintenance of vascular homeostasis, and pathogenesis of vascular disorders. MicroRNAs (miRNAs) mediate the regulation of VSMC functions in response to microenvironmental signals. Because a previous study reported that miR-101, a tumor-suppressive miRNA, is a critical regulator of cell proliferation in vascular disease, we hypothesized that miR-101 controls important cellular processes in VSMCs. The present study aimed to elucidate the effects of miR-101 on VSMC function and its molecular mechanisms. We revealed that miR-101 regulates VSMC proliferation and migration. We showed that miR-101 expression is induced by bone morphogenetic protein (BMP) signaling, and we identified dedicator of cytokinesis 4 (DOCK4) as a novel target of miR-101. Our results suggest that the BMP–miR-101–DOCK4 axis mediates the regulation of VSMC function. Our findings help further the understanding of vascular physiology and pathology.
Collapse
Affiliation(s)
- Nanju Park
- Department of Life Sciences, Incheon National University, Incheon 22012, Korea;
| | - Hara Kang
- Department of Life Sciences, Incheon National University, Incheon 22012, Korea;
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea
- Correspondence: ; Tel.: +82-32-835-8238; Fax: +82-32-835-0763
| |
Collapse
|
26
|
Huycke TR, Miller BM, Gill HK, Nerurkar NL, Sprinzak D, Mahadevan L, Tabin CJ. Genetic and Mechanical Regulation of Intestinal Smooth Muscle Development. Cell 2020; 179:90-105.e21. [PMID: 31539501 DOI: 10.1016/j.cell.2019.08.041] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/31/2019] [Accepted: 08/22/2019] [Indexed: 11/30/2022]
Abstract
The gastrointestinal tract is enveloped by concentric and orthogonally aligned layers of smooth muscle; however, an understanding of the mechanisms by which these muscles become patterned and aligned in the embryo has been lacking. We find that Hedgehog acts through Bmp to delineate the position of the circumferentially oriented inner muscle layer, whereas localized Bmp inhibition is critical for allowing formation of the later-forming, longitudinally oriented outer layer. Because the layers form at different developmental stages, the muscle cells are exposed to unique mechanical stimuli that direct their alignments. Differential growth within the early gut tube generates residual strains that orient the first layer circumferentially, and when formed, the spontaneous contractions of this layer align the second layer longitudinally. Our data link morphogen-based patterning to mechanically controlled smooth muscle cell alignment and provide a mechanistic context for potentially understanding smooth muscle organization in a wide variety of tubular organs.
Collapse
Affiliation(s)
- Tyler R Huycke
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Bess M Miller
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Hasreet K Gill
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nandan L Nerurkar
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - David Sprinzak
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - L Mahadevan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA; Kavli Institute for Bionano Science and Technology, Harvard University, Cambridge, MA 02138, USA
| | - Clifford J Tabin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Abstract
Vascular smooth muscle cells (VSMCs) are a unique cell type that has unusual plasticity controlled by environmental stimuli. As an abnormal increase of VSMC proliferation is associated with various vascular diseases, tight regulation of VSMC phenotypes is essential for maintaining vascular homeostasis. Hypoxia is one environmental stress that stimulates VSMC proliferation. Emerging evidence has indicated that microRNAs (miRNAs) are critical regulators in the hypoxic responses of VSMCs. Therefore, we previously investigated miRNAs modulated by hypoxia in VSMCs and found that miR-1260b is one of the most upregulated miRNAs under hypoxia. However, the mechanism that underlies the regulation of VSMCs via miR-1260b in response to hypoxia has not been explored. Here we demonstrated that hypoxia-induced miR-1260b promotes VSMC proliferation. We also identified growth differentiation factor 11 (GDF11), a member of the TGF-β superfamily, as a novel target of miR-1260b. miR-1260b directly targets the 3’UTR of GDF11. Downregulation of GDF11 inhibited Smad signaling and consequently enhanced the proliferation of VSMCs. Our findings suggest that miR-1260b-mediated GDF11-Smad-dependent signaling is an essential regulatory mechanism in the proliferation of VSMCs, and this axis is modulated by hypoxia to promote abnormal VSMC proliferation. Therefore, our study unveils a novel function of miR-1260b in the pathological proliferation of VSMCs under hypoxia.
Collapse
Affiliation(s)
- Minhyeong Seong
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
| | - Hara Kang
- Division of Life Sciences, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Korea
- Institute for New Drug Development, Incheon National University, Incheon 22012, Korea
| |
Collapse
|
28
|
Hypoxia Promotes Vascular Smooth Muscle Cell (VSMC) Differentiation of Adipose-Derived Stem Cell (ADSC) by Regulating Mettl3 and Paracrine Factors. Stem Cells Int 2020; 2020:2830565. [PMID: 32148516 PMCID: PMC7053496 DOI: 10.1155/2020/2830565] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 12/19/2019] [Accepted: 12/31/2019] [Indexed: 12/18/2022] Open
Abstract
Adipose-derived stem cell (ADSC) is an alternative and less invasive source of mesenchymal stem cells which can be used to develop biological treatment strategies for tissue regeneration, and their therapeutic applications hinge on an understanding of their physiological characteristics. N6-Methyladenosine (m6A) is the most common chemical modification of mRNAs and has recently been revealed to play important roles in cell lineage differentiation and development. However, the role of m6A modification in the vascular smooth muscle cell (VSMC) differentiation of ADSCs remains unclear. Herein, we investigated the expression of N6-adenosine methyltransferases (Mettl3) and demethylases (Fto and Alkbh5) and found that Mettl3 was upregulated in ADSCs undergoing vascular smooth muscle differentiation induction. Moreover, silence of Mettle3 reduced the expression level of VSMC-specific markers, including α-SMA, SM22α, calponin, and SM-MHC. Meanwhile, Mettl3 knockdown also decreased the expression of paracrine factors, including VEGF, HGF, TGF-β, GM-CSF, bFGF, and SDF-1. In addition, our results suggested that hypoxia stress promotes the ADSC differentiate into VMSCs and regulates the secretion of VEGF, HGF, TGF-β, GM-CSF, bFGF, and SDF-1 by mediating Mettl3 gene expression. These observations might contribute to novel progress in understanding the role of epitranscriptomic regulation in the VSMC differentiation of ADSCs and provide a promising perspective for new therapeutic strategies for tissue regeneration.
Collapse
|
29
|
Yang GS, Zheng B, Qin Y, Zhou J, Yang Z, Zhang XH, Zhao HY, Yang HJ, Wen JK. Salvia miltiorrhiza-derived miRNAs suppress vascular remodeling through regulating OTUD7B/KLF4/NMHC IIA axis. Theranostics 2020; 10:7787-7811. [PMID: 32685020 PMCID: PMC7359079 DOI: 10.7150/thno.46911] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 05/31/2020] [Indexed: 12/15/2022] Open
Abstract
Objective: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are essential for vascular remodeling. Natural compounds with diterpene chinone or phenolic acid structure from Salvia miltiorrhiza, an eminent medicinal herb widely used to treat cardiovascular diseases in China, can effectively attenuate vascular remodeling induced by vascular injury. However, it remains unknown whether Salvia miltiorrhiza-derived miRNAs can protect VSMCs from injury by environmental stimuli. Here, we explored the role and underlying mechanisms of Salvia miltiorrhiza-derived Sal-miR-1 and 3 in the regulation of VSMC migration and monocyte adhesion to VSMCs induced by thrombin. Methods: A mouse model for intimal hyperplasia was established by the ligation of carotid artery and the injured carotid arteries were in situ-transfected with Sal-miR-1 and 3 using F-127 pluronic gel. The vascular protective effects of Sal-miR-1 and 3 were assessed via analysis of intimal hyperplasia with pathological morphology. VSMC migration and adhesion were analyzed by the wound healing, transwell membrane assays, and time-lapse imaging experiment. Using loss- and gain-of-function approaches, Sal-miR-1 and 3 regulation of OTUD7B/KLF4/NMHC IIA axis was investigated by using luciferase assay, co-immunoprecipitation, chromatin immunoprecipitation, western blotting, etc. Results:Salvia miltiorrhiza-derived Sal-miR-1 and 3 can enter the mouse body after intragastric administration, and significantly suppress intimal hyperplasia induced by carotid artery ligation. In cultured VSMCs, these two miRNAs inhibit thrombin-induced the migration of VSMCs and monocyte adhesion to VSMCs. Mechanistically, Sal-miR-1 and 3 abrogate OTUD7B upregulation by thrombin via binding to the different sites of the OTUD7B 3'UTR. Most importantly, OTUD7B downregulation by Sal-miR-1 and 3 attenuates KLF4 protein levels via decreasing its deubiquitylation, whereas decreased KLF4 relieves its repression of transcription of NMHC IIA gene and thus increases NMHC IIA expression levels. Further, increased NMHC IIA represses VSMC migration and monocyte adhesion to VSMCs via maintaining the contractile phenotype of VSMCs. Conclusions: Our studies not only found the novel bioactive components from Salvia miltiorrhiza but also clarified the molecular mechanism underlying Sal-miR-1 and 3 inhibition of VSMC migration and monocyte adhesion to VSMCs. These results add important knowledge to the pharmacological actions and bioactive components of Salvia miltiorrhiza. Sal-miR-1 and 3-regulated OTUD7B/KLF4/NMHC IIA axis may represent a therapeutic target for vascular remodeling.
Collapse
Affiliation(s)
- Gao-shan Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
- Department of Biochemistry and Molecular Biology, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Yan Qin
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
- Central Laboratory, Affiliated Hospital of Hebei University, Baoding, China
| | - Jing Zhou
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
- Department of Endocrine, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhan Yang
- Department of Science and Technology, The second hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin-hua Zhang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Hong-ye Zhao
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Hao-jie Yang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
| | - Jin-kun Wen
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neural and Vascular Biology, China Administration of Education, Hebei Medical University, Shijiazhuang, China
- ✉ Corresponding author: Jin-kun Wen, Department of Biochemistry and Molecular Biology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, China. E-mail:
| |
Collapse
|
30
|
Zhang X, Huang T, Zhai H, Peng W, Zhou Y, Li Q, Yang H. Inhibition of lysine-specific demethylase 1A suppresses neointimal hyperplasia by targeting bone morphogenetic protein 2 and mediating vascular smooth muscle cell phenotype. Cell Prolif 2019; 53:e12711. [PMID: 31737960 PMCID: PMC6985674 DOI: 10.1111/cpr.12711] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/17/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Vascular disorders are associated with phenotypical switching of vascular smooth muscle cells (VSMCs). We investigated the effect of bone morphogenetic protein (BMP)-2 in controlling VSMC phenotype and vascular disorder progression. Lysine (K)-specific demethylase 1A (KDM1A) has been identified to target BMP-2 and is employed as a therapeutic means of regulating BMP-2 expression in VSMCs. MATERIALS AND METHODS VSMCs were stimulated with angiotensin II, and the expression of KDM1A and BMP-2 was detected. VSMC proliferation, apoptosis, and phenotype were evaluated. An in vivo aortic injury model was established, and VSMC behaviour was evaluated by the expression of key markers. The activation of BMP-2-associated signalling pathways was examined. RESULTS We confirmed the inhibitory effect of KDM1A on BMP-2 activity and demonstrated that KDM1A inhibition prevented VSMC transformation from a contractile to synthetic phenotype. In angiotensin II-treated VSMCs, KDM1A inhibition triggered a decrease in cell proliferation and inflammatory response. In vivo, KDM1A inhibition alleviated post-surgery neointimal formation and collagen deposition, preventing VSMCs from switching into a synthetic phenotype and suppressing disease onset. These processes were mediated by BMP-2 through canonical small mothers against decapentaplegic signalling, which was associated with the activation of BMP receptors 1A and 1B. CONCLUSIONS The regulatory correlation between KDM1A and BMP-2 offers insights into vascular remodelling and VSMC phenotypic modulation. The reported findings contribute to the development of innovative strategies against vascular disorders.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Huang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Zhai
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenpeng Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhou
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifeng Yang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
31
|
Wang M, Zhang L, Zhu W, Zhang J, Kim SH, Wang Y, Ni L, Telljohann R, Monticone RE, McGraw K, Liu L, de Cabo R, Lakatta EG. Calorie Restriction Curbs Proinflammation That Accompanies Arterial Aging, Preserving a Youthful Phenotype. J Am Heart Assoc 2019; 7:e009112. [PMID: 30371211 PMCID: PMC6222931 DOI: 10.1161/jaha.118.009112] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background Aging exponentially increases the incidence of morbidity and mortality of quintessential cardiovascular disease mainly due to arterial proinflammatory shifts at the molecular, cellular, and tissue levels within the arterial wall. Calorie restriction (CR) in rats improves arterial function and extends both health span and life span. How CR affects the proinflammatory landscape of molecular, cellular, and tissue phenotypic shifts within the arterial wall in rats, however, remains to be elucidated. Methods and Results Aortae were harvested from young (6‐month‐old) and old (24‐month‐old) Fischer 344 rats, fed ad libitum and a second group maintained on a 40% CR beginning at 1 month of age. Histopathologic and morphometric analysis of the arterial wall demonstrated that CR markedly reduced age‐associated intimal medial thickening, collagen deposition, and elastin fractionation/degradation within the arterial walls. Immunostaining/blotting showed that CR effectively prevented an age‐associated increase in the density of platelet‐derived growth factor, matrix metalloproteinase type II activity, and transforming growth factor beta 1 and its downstream signaling molecules, phospho‐mothers against decapentaplegic homolog‐2/3 (p‐SMAD‐2/3) in the arterial wall. In early passage cultured vascular smooth muscle cells isolated from AL and CR rat aortae, CR alleviated the age‐associated vascular smooth muscle cell phenotypic shifts, profibrogenic signaling, and migration/proliferation in response to platelet‐derived growth factor. Conclusions CR reduces matrix and cellular proinflammation associated with aging that occurs within the aortic wall and that are attributable to platelet‐derived growth factor signaling. Thus, CR reduces the platelet‐derived growth factor–associated signaling cascade, contributing to the postponement of biological aging and preservation of a more youthful aortic wall phenotype.
Collapse
Affiliation(s)
- Mingyi Wang
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Li Zhang
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD.,3 Department of Cardiology Nanfang Hospital Southern Medical University Guangzhou China
| | - Wanqu Zhu
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Jing Zhang
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Soo Hyuk Kim
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Yushi Wang
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD.,4 Department of Cardiology The First Hospital of Jilin University Changchun China
| | - Leng Ni
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD.,5 Department of Vascular Surgery Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Richard Telljohann
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Robert E Monticone
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Kimberly McGraw
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Lijuan Liu
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Rafael de Cabo
- 2 Experimental Gerontology Section, Translational Gerontology Branch National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| | - Edward G Lakatta
- 1 Laboratory of Cardiovascular Science National Institute on Aging National Institutes of Health Biomedical Research Center (BRC) Baltimore MD
| |
Collapse
|
32
|
Wang S, Cao W, Gao S, Nie X, Zheng X, Xing Y, Chen Y, Bao H, Zhu D. TUG1 Regulates Pulmonary Arterial Smooth Muscle Cell Proliferation in Pulmonary Arterial Hypertension. Can J Cardiol 2019; 35:1534-1545. [DOI: 10.1016/j.cjca.2019.07.630] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/07/2019] [Accepted: 07/16/2019] [Indexed: 01/07/2023] Open
|
33
|
Prusinski Fernung LE, Yang Q, Sakamuro D, Kumari A, Mas A, Al-Hendy A. Endocrine disruptor exposure during development increases incidence of uterine fibroids by altering DNA repair in myometrial stem cells. Biol Reprod 2019; 99:735-748. [PMID: 29688260 DOI: 10.1093/biolre/ioy097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/20/2018] [Indexed: 12/20/2022] Open
Abstract
Despite the major negative impact uterine fibroids (UFs) have on female reproductive health, little is known about early events that initiate development of these tumors. Somatic fibroid-causing mutations in mediator complex subunit 12 (MED12), the most frequent genetic alterations in UFs (up to 85% of tumors), are implicated in transforming normal myometrial stem cells (MSCs) into tumor-forming cells, though the underlying mechanism(s) leading to these mutations remains unknown. It is well accepted that defective DNA repair increases the risk of acquiring tumor-driving mutations, though defects in DNA repair have not been explored in UF tumorigenesis. In the Eker rat UF model, a germline mutation in the Tsc2 tumor suppressor gene predisposes to UFs, which arise due to "second hits" in the normal allele of this gene. Risk for developing these tumors is significantly increased by early-life exposure to endocrine-disrupting chemicals (EDCs), suggesting increased UF penetrance is modulated by early drivers for these tumors. We analyzed DNA repair capacity using analyses of related gene and protein expression and DNA repair function in MSCs from adult rats exposed during uterine development to the model EDC diethylstilbestrol. Adult MSCs isolated from developmentally exposed rats demonstrated decreased DNA end-joining ability, higher levels of DNA damage, and impaired ability to repair DNA double-strand breaks relative to MSCs from age-matched, vehicle-exposed rats. These data suggest that early-life developmental EDC exposure alters these MSCs' ability to repair and reverse DNA damage, providing a driver for acquisition of mutations that may promote the development of these tumors in adult life.
Collapse
Affiliation(s)
| | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daitoku Sakamuro
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, USA
| | - Alpana Kumari
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, Georgia, USA
| | - Aymara Mas
- Reproductive Medicine Research Group, La Fe Health Research Institute, Valencia, Spain.,Igenomix, Paterna, Valencia, Spain.,Department of Obstetrics and Gynecology, Valencia University and INCLIVA, Valencia, Spain
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
34
|
Li L, Li Y, Tang C. The role of microRNAs in the involvement of vascular smooth muscle cells in the development of atherosclerosis. Cell Biol Int 2019; 43:1102-1112. [PMID: 31066128 DOI: 10.1002/cbin.11164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 05/05/2019] [Indexed: 01/06/2023]
Abstract
MicroRNAs (miRNAs) are a class of nonprotein-encoding RNAs of ~22 nucleotides in length that bind to or complement each other with a target gene messenger RNA (mRNA) to promote mRNA degradation or inhibit translation of the target mRNA. The protein required [such as Toll-like receptor (TLR) proteins] is controlled at an optimal level. By affecting protein translation, miRNAs have become powerful regulators of biological processes, including development, differentiation, cell proliferation, and apoptosis. MiRNAs are involved in the regulation of proliferation, migration, and apoptosis of vascular smooth muscle cells (VSMCs), thereby affecting the formation of atherosclerosis (AS). In recent years, the role and mechanism of miRNAs involved in AS development in VSMCs have been studied extensively. In the current study, the results and progress in miRNA research are reviewed.
Collapse
Affiliation(s)
- Linqing Li
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yongjun Li
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China
| | - Chengchun Tang
- School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
35
|
Zabini D, Granton E, Hu Y, Miranda MZ, Weichelt U, Breuils Bonnet S, Bonnet S, Morrell NW, Connelly KA, Provencher S, Ghanim B, Klepetko W, Olschewski A, Kapus A, Kuebler WM. Loss of SMAD3 Promotes Vascular Remodeling in Pulmonary Arterial Hypertension via MRTF Disinhibition. Am J Respir Crit Care Med 2019; 197:244-260. [PMID: 29095649 DOI: 10.1164/rccm.201702-0386oc] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Vascular remodeling in pulmonary arterial hypertension (PAH) results from smooth muscle cell hypertrophy and proliferation of vascular cells. Loss of BMPR-II (bone morphogenetic protein receptor 2) signaling and increased signaling via TGF-β (transforming growth factor β) and its downstream mediators SMAD (small body size [a C. elegans protein] mothers against decapentaplegic [a Drosophila protein family])-2/3 has been proposed to drive lung vascular remodeling; yet, proteomic analyses indicate a loss of SMAD3 in PAH. OBJECTIVES We proposed that SMAD3 may be dysregulated in PAH and that loss of SMAD3 may present a pathophysiological master switch by disinhibiting its interaction partner, MRTF (myocardin-related transcription factor), which drives muscle protein expression. METHODS SMAD3 levels were measured in lungs from PAH patients, rats treated either with Sugen/hypoxia or monocrotaline (MCT), and in mice carrying a BMPR2 mutation. In vitro, effects of SMAD3 or BMPR2 silencing or SMAD3 overexpression on cell proliferation or smooth muscle hypertrophy were assessed. In vivo, the therapeutic and prophylactic potential of CCG1423, an inhibitor of MRTF, was investigated in Sugen/hypoxia rats. MEASUREMENTS AND MAIN RESULTS SMAD3 was downregulated in lungs of patients with PAH and in pulmonary arteries of three independent PAH animal models. TGF-β treatment replicated the loss of SMAD3 in human pulmonary artery smooth muscle cells (huPASMCs) and human pulmonary artery endothelial cells. SMAD3 silencing increased proliferation and migration in huPASMCs and human pulmonary artery endothelial cells. Coimmunoprecipitation revealed reduced interaction of MRTF with SMAD3 in TGF-β-treated huPASMCs and pulmonary arteries of PAH animal models. In huPASMCs, loss of SMAD3 or BMPR-II increased smooth muscle actin expression, which was attenuated by MRTF inhibition. Conversely, SMAD3 overexpression prevented TGF-β-induced activation of an MRTF reporter and reduced actin stress fibers in BMPR2-silenced huPASMCs. MRTF inhibition attenuated PAH and lung vascular remodeling in Sugen/hypoxia rats. CONCLUSIONS Loss of SMAD3 presents a novel pathomechanism in PAH that promotes vascular cell proliferation and-via MRTF disinhibition-hypertrophy of huPASMCs, thereby reconciling the parallel induction of a synthetic and contractile huPASMC phenotype.
Collapse
Affiliation(s)
- Diana Zabini
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Elise Granton
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yijie Hu
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Maria Zena Miranda
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Ulrike Weichelt
- 3 Department of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Breuils Bonnet
- 4 Pulmonary Hypertension Group of the Institute of Cardiology and Pulmonology, Laval University, Quebec City, Québec, Canada
| | - Sébastien Bonnet
- 4 Pulmonary Hypertension Group of the Institute of Cardiology and Pulmonology, Laval University, Quebec City, Québec, Canada
| | - Nicholas W Morrell
- 5 Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Kim A Connelly
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Steeve Provencher
- 4 Pulmonary Hypertension Group of the Institute of Cardiology and Pulmonology, Laval University, Quebec City, Québec, Canada
| | - Bahil Ghanim
- 6 Department of Thoracic Surgery, Medical University, Vienna, Austria; and
| | - Walter Klepetko
- 6 Department of Thoracic Surgery, Medical University, Vienna, Austria; and
| | - Andrea Olschewski
- 2 Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Andras Kapus
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,7 Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- 1 Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,7 Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
36
|
Ngai D, Lino M, Bendeck MP. Cell-Matrix Interactions and Matricrine Signaling in the Pathogenesis of Vascular Calcification. Front Cardiovasc Med 2018; 5:174. [PMID: 30581820 PMCID: PMC6292870 DOI: 10.3389/fcvm.2018.00174] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022] Open
Abstract
Vascular calcification is a complex pathological process occurring in patients with atherosclerosis, type 2 diabetes, and chronic kidney disease. The extracellular matrix, via matricrine-receptor signaling plays important roles in the pathogenesis of calcification. Calcification is mediated by osteochondrocytic-like cells that arise from transdifferentiating vascular smooth muscle cells. Recent advances in our understanding of the plasticity of vascular smooth muscle cell and other cells of mesenchymal origin have furthered our understanding of how these cells transdifferentiate into osteochondrocytic-like cells in response to environmental cues. In the present review, we examine the role of the extracellular matrix in the regulation of cell behavior and differentiation in the context of vascular calcification. In pathological calcification, the extracellular matrix not only provides a scaffold for mineral deposition, but also acts as an active signaling entity. In recent years, extracellular matrix components have been shown to influence cellular signaling through matrix receptors such as the discoidin domain receptor family, integrins, and elastin receptors, all of which can modulate osteochondrocytic differentiation and calcification. Changes in extracellular matrix stiffness and composition are detected by these receptors which in turn modulate downstream signaling pathways and cytoskeletal dynamics, which are critical to osteogenic differentiation. This review will focus on recent literature that highlights the role of cell-matrix interactions and how they influence cellular behavior, and osteochondrocytic transdifferentiation in the pathogenesis of cardiovascular calcification.
Collapse
Affiliation(s)
- David Ngai
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Marsel Lino
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON, Canada.,Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Pardali E, Makowski LM, Leffers M, Borgscheiper A, Waltenberger J. BMP-2 induces human mononuclear cell chemotaxis and adhesion and modulates monocyte-to-macrophage differentiation. J Cell Mol Med 2018; 22:5429-5438. [PMID: 30102472 PMCID: PMC6201342 DOI: 10.1111/jcmm.13814] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/07/2018] [Accepted: 06/29/2018] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a cardiovascular risk factor which leads to atherosclerosis, an inflammatory disease characterized by the infiltration of mononuclear cells in the vessel. Bone morphogenetic protein (BMP)‐2 is a cytokine which has been recently shown to be elevated in atherosclerosis and T2DM and to contribute to vascular inflammation. However, the role of BMP‐2 in the regulation of mononuclear cell function remains to be established. Herein, we demonstrate that BMP‐2 induced human monocyte chemotaxis via phosphoinositide 3 kinase and mitogen‐activated protein kinases. Inhibition of endogenous BMP‐2 signalling, by Noggin or a BMP receptor inhibitor, interfered with monocyte migration. Although BMP‐2 expression was increased in monocytes from T2DM patients, it could still stimulate their migration. Furthermore, BMP‐2 interfered with their differentiation into M2 macrophages. Finally, BMP‐2 both induced the adhesion of monocytes to fibronectin and endothelial cells (ECs), and promoted the adhesive properties of ECs, by increasing expression of adhesion and pro‐inflammatory molecules. Our data demonstrate that BMP‐2 could exert its pro‐inflammatory effects by inducing monocyte migration and adhesiveness to ECs and by interfering with the monocyte differentiation into M2 macrophages. Our findings provide novel insights into the mechanisms by which BMP‐2 may contribute to the development of atherosclerosis.
Collapse
Affiliation(s)
- Evangelia Pardali
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Lena-Maria Makowski
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Merle Leffers
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Andreas Borgscheiper
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, University Hospital of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| |
Collapse
|
38
|
Huang X, Yue Z, Wu J, Chen J, Wang S, Wu J, Ren L, Zhang A, Deng P, Wang K, Wu C, Ding X, Ye P, Xia J. MicroRNA-21 Knockout Exacerbates Angiotensin II–Induced Thoracic Aortic Aneurysm and Dissection in Mice With Abnormal Transforming Growth Factor-β–SMAD3 Signaling. Arterioscler Thromb Vasc Biol 2018. [DOI: 10.1161/atvbaha.117.310694] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Objective—
Thoracic aortic aneurysm and dissection (TAAD) are severe vascular conditions. Dysfunctional transforming growth factor-β (TGF-β) signaling in vascular smooth muscle cells and elevated angiotensin II (AngII) levels are implicated in the development of TAAD. In this study, we investigated whether these 2 factors lead to TAAD in a mouse model and explored the possibility of using microRNA-21 (
miR-21
) for the treatment of TAAD.
Approach and Results—
TAAD was developed in
Smad3
(mothers against decapentaplegic homolog 3) heterozygous (S3
+/−
) mice infused with AngII. We found that p-ERK (phosphorylated extracellular regulated protein kinases)– and p-JNK (phosphorylated c-Jun N-terminal kinase)–associated
miR-21
was higher in TAAD lesions. We hypothesize that downregulation of
miR-21
mitigate TAAD formation. However,
Smad3
+/−
:miR-21
−/−
(S3
+/−
21
−/−
) mice exhibited conspicuous TAAD formation after AngII infusion. The vascular wall was dilated, and aortic rupture occurred within 23 days during AngII infusion. We then examined canonical and noncanonical TGF-β signaling and found that
miR-21
knockout in S3
+/−
mice increased SMAD7 and suppressed canonical TGF-β signaling. Vascular smooth muscle cells lacking TGF-β signals tended to switch from a contractile to a synthetic phenotype. The silencing of
Smad7
with lentivirus prevented AngII-induced TAAD formation in S3
+/−
21
−/−
mice.
Conclusions—
Our study demonstrated that
miR-21
knockout exacerbated AngII-induced TAAD formation in mice, which was associated with TGF-β signaling dysfunction. Therapeutic strategies targeting TAAD should consider unexpected side effects associated with alterations in TGF-β signaling.
Collapse
Affiliation(s)
- Xiaofan Huang
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Zhang Yue
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Jia Wu
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
- Key Laboratory for Molecular Diagnosis of Hubei Province, Central Hospital of Wuhan (J.W.)
| | - Jiuling Chen
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Sihua Wang
- Department of Thoracic Surgery, Union Hospital (S.W.)
| | - Jie Wu
- Central Laboratory, Central Hospital of Wuhan (J.W.)
| | - Linyun Ren
- Department of Anesthesia, Central Hospital of Wuhan (L.R.), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anchen Zhang
- Department of Cardiovascular Medicine, Central Hospital of Wuhan (A.Z., P.Y.)
| | - Peng Deng
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Ke Wang
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Chuangyan Wu
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Xiangchao Ding
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
| | - Ping Ye
- Department of Cardiovascular Medicine, Central Hospital of Wuhan (A.Z., P.Y.)
| | - Jiahong Xia
- From the Department of Cardiovascular Surgery, Union Hospital (X.H., Z.Y., J.C., J.W., P.D., K.W., C.W., X.D., J.X.)
- Department of Cardiovascular Surgery, Central Hospital of Wuhan (J.X.)
| |
Collapse
|
39
|
Wang D, Uhrin P, Mocan A, Waltenberger B, Breuss JM, Tewari D, Mihaly-Bison J, Huminiecki Ł, Starzyński RR, Tzvetkov NT, Horbańczuk J, Atanasov AG. Vascular smooth muscle cell proliferation as a therapeutic target. Part 1: molecular targets and pathways. Biotechnol Adv 2018; 36:1586-1607. [PMID: 29684502 DOI: 10.1016/j.biotechadv.2018.04.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/15/2018] [Accepted: 04/18/2018] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases are a major cause of human death worldwide. Excessive proliferation of vascular smooth muscle cells contributes to the etiology of such diseases, including atherosclerosis, restenosis, and pulmonary hypertension. The control of vascular cell proliferation is complex and encompasses interactions of many regulatory molecules and signaling pathways. Herein, we recapitulated the importance of signaling cascades relevant for the regulation of vascular cell proliferation. Detailed understanding of the mechanism underlying this process is essential for the identification of new lead compounds (e.g., natural products) for vascular therapies.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria; Institute of Clinical Chemistry, University Hospital Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Pavel Uhrin
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria.
| | - Andrei Mocan
- Department of Pharmaceutical Botany, "Iuliu Hațieganu" University of Medicine and Pharmacy, Strada Gheorghe Marinescu 23, 400337 Cluj-Napoca, Romania; Institute for Life Sciences, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Mănăştur 3-5, 400372 Cluj-Napoca, Romania
| | - Birgit Waltenberger
- Institute of Pharmacy/Pharmacognosy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Johannes M Breuss
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Kumaun University, Bhimtal, 263136 Nainital, Uttarakhand, India
| | - Judit Mihaly-Bison
- Center for Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Schwarzspanierstrasse 17, 1090 Vienna, Austria
| | - Łukasz Huminiecki
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Rafał R Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Nikolay T Tzvetkov
- Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany; NTZ Lab Ltd., Krasno Selo 198, 1618 Sofia, Bulgaria
| | - Jarosław Horbańczuk
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, ul. Postepu 36A, Jastrzębiec, 05-552 Magdalenka, Poland; Department of Pharmacognosy, University of Vienna, Althanstrasse 14, A-1090 Vienna, Austria.
| |
Collapse
|
40
|
Gu W, Hong X, Potter C, Qu A, Xu Q. Mesenchymal stem cells and vascular regeneration. Microcirculation 2018; 24. [PMID: 27681821 DOI: 10.1111/micc.12324] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/20/2016] [Indexed: 12/22/2022]
Abstract
In recent years, MSCs have emerged as a promising therapeutic cell type in regenerative medicine. They hold great promise for treating cardiovascular diseases, such as myocardial infarction and limb ischemia. MSCs may be utilized in both cell-based therapy and vascular graft engineering to restore vascular function, thereby providing therapeutic benefits to patients. The efficacy of MSCs lies in their multipotent differentiation ability toward vascular smooth muscle cells, endothelial cells and other cell types, as well as their capacity to secrete various trophic factors, which are potent in promoting angiogenesis, inhibiting apoptosis and modulating immunoreaction. Increasing our understanding of the mechanisms of MSC involvement in vascular regeneration will be beneficial in boosting present therapeutic approaches and developing novel ones to treat cardiovascular diseases. In this review, we aim to summarize current progress in characterizing the in vivo identity of MSCs, to discuss mechanisms involved in cell-based therapy utilizing MSCs, and to explore current and future strategies for vascular regeneration.
Collapse
Affiliation(s)
- Wenduo Gu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Xuechong Hong
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Claire Potter
- Cardiovascular Division, King's College London BHF Centre, London, UK
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre, London, UK
| |
Collapse
|
41
|
Hypoxia Enhances Differentiation of Adipose Tissue-Derived Stem Cells toward the Smooth Muscle Phenotype. Int J Mol Sci 2018; 19:ijms19020517. [PMID: 29419805 PMCID: PMC5855739 DOI: 10.3390/ijms19020517] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 01/31/2018] [Accepted: 02/01/2018] [Indexed: 11/17/2022] Open
Abstract
Smooth muscle differentiated adipose tissue-derived stem cells are a valuable resource for regeneration of gastrointestinal tissues, such as the gut and sphincters. Hypoxia has been shown to promote adipose tissue-derived stem cells proliferation and maintenance of pluripotency, but the influence of hypoxia on their smooth myogenic differentiation remains unexplored. This study investigated the phenotype and contractility of adipose-derived stem cells differentiated toward the smooth myogenic lineage under hypoxic conditions. Oxygen concentrations of 2%, 5%, 10%, and 20% were used during differentiation of adipose tissue-derived stem cells. Real time reverse transcription polymerase chain reaction and immunofluorescence staining were used to detect the expression of smooth muscle cells-specific markers, including early marker smooth muscle alpha actin, middle markers calponin, caldesmon, and late marker smooth muscle myosin heavy chain. The specific contractile properties of cells were verified with both a single cell contraction assay and a gel contraction assay. Five percent oxygen concentration significantly increased the expression levels of α-smooth muscle actin, calponin, and myosin heavy chain in adipose-derived stem cell cultures after 2 weeks of induction (p < 0.01). Cells differentiated in 5% oxygen conditions showed greater contraction effect (p < 0.01). Hypoxia influences differentiation of smooth muscle cells from adipose stem cells and 5% oxygen was the optimal condition to generate smooth muscle cells that contract from adipose stem cells.
Collapse
|
42
|
Zhang W, Zhu T, Wu W, Ge X, Xiong X, Zhang Z, Hu C. LOX-1 mediated phenotypic switching of pulmonary arterial smooth muscle cells contributes to hypoxic pulmonary hypertension. Eur J Pharmacol 2017; 818:84-95. [PMID: 29069578 DOI: 10.1016/j.ejphar.2017.10.037] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/19/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
In pulmonary hypertension (PH), pulmonary arterial smooth muscle cells (PASMCs) are dedifferentiated, undergoing a contractile-to-synthetic phenotypic switching. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) plays diverse roles in the cardiovascular system, but its contribution to PH remains to be fully defined. The present study was undertaken to explore the role of LOX-1 in PASMCs dedifferentiation in hypoxia-induced pulmonary vascular remodeling and PH. In a rat model of hypoxic PH, pulmonary vascular remodeling was accompanied by increased expression of LOX-1 in pulmonary arteries. In primary rat PASMCs, hypoxia-induced PASMCs dedifferentiation occurred concomitantly with LOX-1 upregulation. Inhibition of LOX-1 by either siRNA knockdown or neutralizing antibody significantly ameliorated PASMCs dedifferentiation. Mechanistically, LOX-1 promotes PASMCs dedifferentiation under hypoxic conditions via ERK1/2-Elk-1/MRTF-A/SRF signaling pathway. In conclusion, our data uncovers an important role of LOX-1 in the maintenance of PASMCs phenotype. Therapeutic targeting of LOX-1/ERK1/2-Elk-1/MRTF-A/SRF signaling axis would be exploited to treat hypoxic PH.
Collapse
Affiliation(s)
- Weifang Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Department of Pharmacy, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Tiantian Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Weihua Wu
- School of pharmaceutical Sciences, Hunan University of Medicine, Huaihua, Hunan 418000, China
| | - Xiaoyue Ge
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Xiaoming Xiong
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan 410078, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan 410078, China.
| | - Changping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
43
|
Dale M, Fitzgerald MP, Liu Z, Meisinger T, Karpisek A, Purcell LN, Carson JS, Harding P, Lang H, Koutakis P, Batra R, Mietus CJ, Casale G, Pipinos I, Baxter BT, Xiong W. Premature aortic smooth muscle cell differentiation contributes to matrix dysregulation in Marfan Syndrome. PLoS One 2017; 12:e0186603. [PMID: 29040313 PMCID: PMC5645122 DOI: 10.1371/journal.pone.0186603] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022] Open
Abstract
Thoracic aortic aneurysm and dissection are life-threatening complications of Marfan syndrome (MFS). Studies of human and mouse aortic samples from late stage MFS demonstrate increased TGF-β activation/signaling and diffuse matrix changes. However, the role of the aortic smooth muscle cell (SMC) phenotype in early aneurysm formation in MFS has yet to be fully elucidated. As our objective, we investigated whether an altered aortic SMC phenotype plays a role in aneurysm formation in MFS. We describe previously unrecognized concordant findings in the aortas of a murine model of MFS, mgR, during a critical and dynamic phase of early development. Using Western blot, gelatin zymography, and histological analysis, we demonstrated that at postnatal day (PD) 7, before aortic TGF-β levels are increased, there is elastic fiber fragmentation/disorganization and increased levels of MMP-2 and MMP-9. Compared to wild type (WT) littermates, aortic SMCs in mgR mice express higher levels of contractile proteins suggesting a switch to a more mature contractile phenotype. In addition, tropoelastin levels are decreased in mgR mice, a finding consistent with a premature switch to a contractile phenotype. Proliferation assays indicate a decrease in the proliferation rate of mgR cultured SMCs compared to WT SMCs. KLF4, a regulator of smooth muscle cell phenotype, was decreased in aortic tissue of mgR mice. Finally, overexpression of KLF4 partially reversed this phenotypic change in the Marfan SMCs. This study indicates that an early phenotypic switch appears to be associated with initiation of important metabolic changes in SMCs that contribute to subsequent pathology in MFS.
Collapse
Affiliation(s)
- Matthew Dale
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Matthew P. Fitzgerald
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Zhibo Liu
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Trevor Meisinger
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Andrew Karpisek
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Laura N. Purcell
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jeffrey S. Carson
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Paul Harding
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Haili Lang
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Panagiotis Koutakis
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Rishi Batra
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Constance J. Mietus
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - George Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Iraklis Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - B. Timothy Baxter
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Wanfen Xiong
- Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- * E-mail:
| |
Collapse
|
44
|
Yang D, Ma M, Zhou W, Yang B, Xiao C. Inhibition of miR-32 activity promoted EMT induced by PM2.5 exposure through the modulation of the Smad1-mediated signaling pathways in lung cancer cells. CHEMOSPHERE 2017; 184:289-298. [PMID: 28601662 DOI: 10.1016/j.chemosphere.2017.05.152] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/21/2017] [Accepted: 05/27/2017] [Indexed: 05/20/2023]
Abstract
Epithelial mesenchymal transition (EMT) is a crucial morphological event during tumor progression. The present study reported that EMT could be triggered by airborne fine particulate matter (PM) with a mean diameter of less than 2.5 μm (PM2.5) in human lung cancer cells. We also aimed to elucidate the possible mechanisms of these processes. The results showed that treatment with PM2.5 promoted the activity of the SMAD family member 1 (Smad1)-mediated signaling pathway and downregulated the expression of the inhibitory Smad proteins Smad6 and Smad7 in lung cancer cells. Moreover, the knockdown of Smad1 suppressed the EMT process induced by PM2.5 exposure. Our data further revealed that miR-32 has a negative effect on PM2.5-induced EMT. The results showed that the expression level of miR-32 was significantly upregulated in the PM2.5-induced EMT process. The knockdown of miR-32 enhances the activity of the Smad1-mediated signaling pathway, which promotes the EMT process induced by PM2.5. Thus, these findings indicate that PM2.5 can induce the EMT process through the Smad1-mediated signaling pathway, and miR-32 may act as an EMT inhibitor in lung cancer cells.
Collapse
Affiliation(s)
- Dan Yang
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China; Department of Pharmacology, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China
| | - Mingyue Ma
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China; Department of Toxicology, School of Public Health, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China
| | - Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China
| | - Biao Yang
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China
| | - Chunling Xiao
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, North Huanghe Street, Huanggu District, Shenyang City, 110034, PR China.
| |
Collapse
|
45
|
Gökçinar-Yagci B, Çelebi-Saltik B. Comparison of different culture conditions for smooth muscle cell differentiation of human umbilical cord vein CD146+ perivascular cells. Cell Tissue Bank 2017; 18:501-511. [DOI: 10.1007/s10561-017-9656-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/12/2017] [Indexed: 12/12/2022]
|
46
|
MiR-26a contributes to the PDGF-BB-induced phenotypic switch of vascular smooth muscle cells by suppressing Smad1. Oncotarget 2017; 8:75844-75853. [PMID: 29100273 PMCID: PMC5652667 DOI: 10.18632/oncotarget.17998] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 05/06/2017] [Indexed: 12/21/2022] Open
Abstract
The phenotypic switch of vascular smooth muscle cells (VSMCs) is a key event in the pathogenesis of various vascular diseases, such as atherosclerosis and post-angioplasty restenosis. Small non-coding microRNAs (miRNAs) have emerged as critical modulators of VSMC function. In the present study, miR-26a was significantly increased in cultured VSMCs stimulated by platelet-derived growth factor-BB (PDGF-BB) and in arteries with neointimal lesion formation. Moreover, we demonstrated that miR-26a regulates the expression of VSMC differentiation marker genes such as α-smooth muscle actin (α-SMA), calponin and smooth muscle myosin heavy chain (SM-MHC) in PDGF-BB-treated VSMCs. We further confirmed that the regulatory effect of miR-26a during the phenotypic transition occurs through its target gene Smad1, which is a critical mediator of the pro-contractile signal transmitted by bone morphogenetic protein (BMP) and transforming growth factor-beta (TGF-β). This discovery proposed a new channel for communication between PDGF and the BMP/TGF-β family. We concluded that miR-26a is an important regulator in the PDGF-BB-mediated VSMC phenotypic transition by targeting Smad1. Interventions aimed at miR-26a may be promising in treating numerous proliferative vascular disorders.
Collapse
|
47
|
Tuning of major signaling networks (TGF-β, Wnt, Notch and Hedgehog) by miRNAs in human stem cells commitment to different lineages: Possible clinical application. Biomed Pharmacother 2017; 91:849-860. [PMID: 28501774 DOI: 10.1016/j.biopha.2017.05.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/29/2017] [Accepted: 05/04/2017] [Indexed: 02/07/2023] Open
Abstract
Two distinguishing characteristics of stem cells, their continuous division in the undifferentiated state and growth into any cell types, are orchestrated by a number of cell signaling pathways. These pathways act as a niche factor in controlling variety of stem cells. The core stem cell signaling pathways include Wingless-type (Wnt), Hedgehog (HH), and Notch. Additionally, they critically regulate the self-renewal and survival of cancer stem cells. Conversely, stem cells' main properties, lineage commitment and stemness, are tightly controlled by epigenetic mechanisms such as DNA methylation, histone modifications and non-coding RNA-mediated regulatory events. MicroRNAs (miRNAs) are cellular switches that modulate stem cells outcomes in response to diverse extracellular signals. Numerous scientific evidences implicating miRNAs in major signal transduction pathways highlight new crosstalks of cellular processes. Aberrant signaling pathways and miRNAs levels result in developmental defects and diverse human pathologies. This review discusses the crosstalk between the components of main signaling networks and the miRNA machinery, which plays a role in the context of stem cells development and provides a set of examples to illustrate the extensive relevance of potential novel therapeutic targets.
Collapse
|
48
|
Aji K, Zhang Y, Aimaiti A, Wang Y, Rexiati M, Azhati B, Tusong H, Cui L, Wang C. MicroRNA-145 regulates the differentiation of human adipose-derived stem cells to smooth muscle cells via targeting Krüppel-like factor 4. Mol Med Rep 2017; 15:3787-3795. [PMID: 28440409 DOI: 10.3892/mmr.2017.6478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 11/04/2016] [Indexed: 11/05/2022] Open
Abstract
Understanding the molecular mechanisms underlying human adipose-derived stem cell (hASC) differentiation to smooth muscle may contribute to the development of effective therapies for relevant muscle defects, such as bladder wall and urethral defects. A previous study described the differentiation of hASCs to smooth muscle cells (SMCs) by transforming growth factor-β1 (TGF-β1) and bone morphogenetic protein‑4 (BMP4) treatment. The present study investigated whether microRNA-145 (miR‑145) may be involved in the process of hASC differentiation. The expression of miR‑145 was significantly increased during differentiation of ASCs to SMCs. SMC‑specific genes and proteins, including a‑smooth muscle actin (α‑SMA), smooth muscle protein‑22α(SM22α), calponin and myosin heavy chain (SM‑MHC) were upregulated by transfection of a miR‑145 mimic. By contrast, these factors were downregulated following introduction of antisense oligonucleotides. In addition, Krüppel‑like factor 4 (KLF4) levels, which decreased during the differentiation of hASCs, were downregulated when the cells were transfected miR‑145 mimics. Futhermore, inhibition of KLF4 by treatment with short‑interfering‑RNA against KLF4, resulted in increased expression of SMC‑specific genes and proteins. In conclusion, the results of the present study demonstrated that by regulating KLF4, miR‑145 may be involved in regulating smooth muscle differentiation of ASCs induced by TGF‑β1 and BMP4.
Collapse
Affiliation(s)
- Kaisaier Aji
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Yun Zhang
- Department of Orthopedics, Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Abudusaimi Aimaiti
- Department of Joint Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Yujie Wang
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Mulati Rexiati
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Baihetiya Azhati
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Hamulati Tusong
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Lei Cui
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Chen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
49
|
Cai P, Kovacs L, Dong S, Wu G, Su Y. BMP4 inhibits PDGF-induced proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L638-L648. [PMID: 28235949 PMCID: PMC5451598 DOI: 10.1152/ajplung.00260.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 02/09/2017] [Accepted: 02/23/2017] [Indexed: 11/22/2022] Open
Abstract
In the present study, we investigated the effect of bone morphogenetic protein 4 (BMP4) on PDGF-induced cell proliferation and collagen synthesis in pulmonary artery smooth muscle cells (PASMCs). Normal human PASMCs were incubated with and without PDGF-BB in the absence and presence of BMP4 for 0.5 to 24 h. The protein levels of collagen-I, p-Smad2/3, p-Smad1/5, and intracellular active TGF-β1, calpain activity, and cell proliferation were then measured. The results showed that BMP4 induced an increase in p-Smad1/5 but had no effect on the protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in control PASMCs. Nevertheless, BMP4 attenuated increases in cell proliferation and protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in PASMCs exposed to PDGF-BB. Moreover, BMP4 increased PKA activity and inhibition of PKA prevented the inhibitory effects of BMP4 on PDGF-BB-induced calpain activation in normal PASMCs. The PKA activator forskolin recapitulated the suppressive effect of BMP4 on PDGF-induced calpain activation. Furthermore, BMP4 prevented a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in normal PASMCs. Finally, BMP4 did not attenuate PDGF-induced increases in cell proliferation, collagen-I protein levels, and calpain activation and did not induce PKA activation and did not prevent a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in PASMCs from idiopathic pulmonary arterial hypertension (PAH) patients. These data demonstrate that BMP4 inhibits PDGF-induced cell proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in normal PASMCs. The inhibitory effects of BMP4 on PDGF-induced cell proliferation, collagen synthesis, and calpain-2 activation are impaired in PASMCs from PAH patients, which may contribute to pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Pengcheng Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Sam Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia.,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; and
| |
Collapse
|
50
|
Understanding the requirements of self-expandable stents for heart valve replacement: Radial force, hoop force and equilibrium. J Mech Behav Biomed Mater 2017; 68:252-264. [PMID: 28219851 DOI: 10.1016/j.jmbbm.2017.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/03/2017] [Accepted: 02/05/2017] [Indexed: 11/22/2022]
Abstract
A proper interpretation of the forces developed during stent crimping and deployment is of paramount importance for a better understanding of the requirements for successful heart valve replacement. The present study combines experimental and computational methods to assess the performance of a nitinol stent for tissue-engineered heart valve implantation. To validate the stent model, the mechanical response to parallel plate compression and radial crimping was evaluated experimentally. Finite element simulations showed good agreement with the experimental findings. The computational models were further used to determine the hoop force on the stent and radial force on a rigid tool during crimping and self-expansion. In addition, stent deployment against ovine and human pulmonary arteries was simulated to determine the hoop force on the stent-artery system and the equilibrium diameter for different degrees of oversizing.
Collapse
|