1
|
Wong D, Qiu H. New insights into the pharmacological inhibition of SRF activity: Key inhibitory targets and mechanisms. Vascul Pharmacol 2024; 157:107443. [PMID: 39586415 DOI: 10.1016/j.vph.2024.107443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Serum Response Factor (SRF) is a critical regulatory transcription factor widely expressed across cell types and is essential for animal survival. Excessive SRF activity has been linked to various pathological conditions and diseases, including cardiovascular diseases, cancers and neurodegenerative disorders, making the inhibition of SRF hyperactivity a promising therapeutic strategy. This review summarizes recent advancements in the discovery and development of SRF inhibitors, their regulatory mechanisms, and their respective molecular foundations. These insights deepen our understanding of current therapeutic potentials, paving the way for novel approaches to treat diseases associated with SRF hyperactivity.
Collapse
Affiliation(s)
- Daniel Wong
- Translational Cardiovascular Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| | - Hongyu Qiu
- Translational Cardiovascular Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA; Clinical Translational Sciences (CTS) and Bio5 Institution, University of Arizona, Tucson, AZ 8572, USA.
| |
Collapse
|
2
|
Yan R, Song A, Zhang C. The Pathological Mechanisms and Therapeutic Molecular Targets in Arteriovenous Fistula Dysfunction. Int J Mol Sci 2024; 25:9519. [PMID: 39273465 PMCID: PMC11395150 DOI: 10.3390/ijms25179519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
The number of patients with end-stage renal disease (ESRD) requiring hemodialysis is increasing worldwide. Although arteriovenous fistula (AVF) is the best and most important vascular access (VA) for hemodialysis, its primary maturation failure rate is as high as 60%, which seriously endangers the prognosis of hemodialysis patients. After AVF establishment, the venous outflow tract undergoes hemodynamic changes, which are translated into intracellular signaling pathway cascades, resulting in an outward and inward remodeling of the vessel wall. Outward remodeling refers to the thickening of the vessel wall and the dilation of the lumen to accommodate the high blood flow in the AVF, while inward remodeling is mainly characterized by intimal hyperplasia. More and more studies have shown that the two types of remodeling are closely related in the occurrence and development of, and jointly determining the final fate of, AVF. Therefore, it is essential to investigate the underlying mechanisms involved in outward and inward remodeling for identifying the key targets in alleviating AVF dysfunction. In this review, we summarize the current clinical diagnosis, monitoring, and treatment techniques for AVF dysfunction and discuss the possible pathological mechanisms related to improper outward and inward remodeling in AVF dysfunction, as well as summarize the similarities and differences between the two remodeling types in molecular mechanisms. Finally, the representative therapeutic targets of potential clinical values are summarized.
Collapse
Affiliation(s)
- Ruiwei Yan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
3
|
Visconti A, Qiu H. Recent advances in serum response factor posttranslational modifications and their therapeutic potential in cardiovascular and neurological diseases. Vascul Pharmacol 2024; 156:107421. [PMID: 39209126 PMCID: PMC11626983 DOI: 10.1016/j.vph.2024.107421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Serum Response Factor (SRF) is a key regulatory transcription factor present in various cell types throughout the body, playing essential roles in cellular functions under physiological conditions. Mutations and abnormal expression of SRF have been linked to the development of various diseases and disorders. Recent evidence highlights that post-translational modifications (PTMs) are critical for regulating SRF function in different cell types and contribute to disease pathogenesis. Targeting SRF-related PTMs is emerging as a promising therapeutic approach for treating SRF-associated diseases. In this review, we summarize recent advances in understanding SRF PTMs and their underlying regulatory mechanisms. We also explore the implications of SRF-PTM in related cardiovascular and neurological diseases and their potential for therapeutic intervention. This information underscores the significance of SRF PTMs in both physiological and pathological contexts, enhancing our understanding of disease mechanisms and paving the way for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Alexander Visconti
- Cardiovascular Translational Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA.
| | - Hongyu Qiu
- Cardiovascular Translational Research Center, Department of Internal Medicine, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ 85004, USA; Clinical Translational Sciences (CTS) and Bio5 Institution, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
4
|
Kajuluri LP, Lyu QR, Doja J, Kumar A, Wilson MP, Sgrizzi SR, Rezaeimanesh E, Miano JM, Morgan KG. Calponin 1 inhibits agonist-induced ERK activation and decreases calcium sensitization in vascular smooth muscle. J Cell Mol Med 2024; 28:e18025. [PMID: 38147352 PMCID: PMC10805486 DOI: 10.1111/jcmm.18025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 10/07/2023] [Indexed: 12/27/2023] Open
Abstract
Smooth muscle cell (SMC) contraction and vascular tone are modulated by phosphorylation and multiple modifications of the thick filament, and thin filament regulation of SMC contraction has been reported to involve extracellular regulated kinase (ERK). Previous studies in ferrets suggest that the actin-binding protein, calponin 1 (CNN1), acts as a scaffold linking protein kinase C (PKC), Raf, MEK and ERK, promoting PKC-dependent ERK activation. To gain further insight into this function of CNN1 in ERK activation and the regulation of SMC contractility in mice, we generated a novel Calponin 1 knockout mouse (Cnn1 KO) by a single base substitution in an intronic CArG box that preferentially abolishes expression of CNN1 in vascular SMCs. Using this new Cnn1 KO mouse, we show that ablation of CNN1 has two effects, depending on the cytosolic free calcium level: (1) in the presence of elevated intracellular calcium caused by agonist stimulation, Cnn1 KO mice display a reduced amplitude of stress and stiffness but an increase in agonist-induced ERK activation; and (2) during intracellular calcium depletion, in the presence of an agonist, Cnn1 KO mice exhibit increased duration of SM tone maintenance. Together, these results suggest that CNN1 plays an important and complex modulatory role in SMC contractile tone amplitude and maintenance.
Collapse
Affiliation(s)
- Lova Prasadareddy Kajuluri
- Vascular Biology Laboratory, Department of Health SciencesBoston UniversityBostonMassachusettsUSA
- Present address:
Cardiovascular Research CenterMassachusetts General HospitalCharlestownMassachusettsUSA
| | - Qing Rex Lyu
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
- Medical Research CenterChongqing General HospitalChongqingChina
| | - Jaser Doja
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | - Ajay Kumar
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | | | - Samantha R. Sgrizzi
- Vascular Biology Laboratory, Department of Health SciencesBoston UniversityBostonMassachusettsUSA
| | - Elika Rezaeimanesh
- Vascular Biology Laboratory, Department of Health SciencesBoston UniversityBostonMassachusettsUSA
| | - Joseph M. Miano
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGeorgiaUSA
| | - Kathleen G. Morgan
- Vascular Biology Laboratory, Department of Health SciencesBoston UniversityBostonMassachusettsUSA
| |
Collapse
|
5
|
Neural Pericytes: A genomic archival state programmed by CHromatin topology. Eur J Cell Biol 2022; 101:151211. [DOI: 10.1016/j.ejcb.2022.151211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/04/2022] [Accepted: 02/25/2022] [Indexed: 11/22/2022] Open
|
6
|
Yang S, Jiang X, Xiao X, Niu C, Xu Y, Huang Z, Kang YJ, Feng L. Controlling the Poly(ε-caprolactone) Degradation to Maintain the Stemness and Function of Adipose-Derived Mesenchymal Stem Cells in Vascular Regeneration Application. Macromol Biosci 2020; 21:e2000226. [PMID: 33094556 DOI: 10.1002/mabi.202000226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/11/2020] [Accepted: 09/29/2020] [Indexed: 02/05/2023]
Abstract
Biodegradable poly(ε-caprolactone) (PCL) scaffolds with adipose-derived mesenchymal stem cells (ADSCs) have been used in vascular regeneration studies. An evaluation method of the effect of PCL degradation products (DP) on the viability, stemness, and differentiation capacities of ADSCs is established. ADSCs are cultured in medium containing different concentrations of PCL DP before evaluating the effect of PCL DP on the cell apoptosis and proliferation, cell surface antigens, adipogenic and osteogenic differentiation capacities, and capacities to differentiate into endothelial cells and smooth muscle cells. The results demonstrate that PCL DP exceed 0.05 mg mL-1 may change the stemness and differentiation capacities of ADSCs. Therefore, to control the proper concentration of PCL DP is essential for ADSCs in vascular regeneration application.
Collapse
Affiliation(s)
- Shaojie Yang
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Xia Jiang
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Xiong Xiao
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Chuan Niu
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Yue Xu
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Ziwei Huang
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Y James Kang
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| | - Li Feng
- S. Yang, Dr. X. Jiang, X. Xiao, C. Niu, Y. Xu, Z. Huang, Prof. Y. J. Kang, Prof. L. Feng, Regenerative Medicine Research Center, Sichuan University West China Hospital, No. 4 Keyuan Road, Wuhou District, Chengdu, 610041, China
| |
Collapse
|
7
|
Chen Y, Su X, Qin Q, Yu Y, Jia M, Zhang H, Li H, Pei L. New insights into phenotypic switching of VSMCs induced by hyperhomocysteinemia: Role of endothelin-1 signaling. Biomed Pharmacother 2020; 123:109758. [DOI: 10.1016/j.biopha.2019.109758] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/20/2019] [Accepted: 11/29/2019] [Indexed: 12/30/2022] Open
|
8
|
Nagao M, Lyu Q, Zhao Q, Wirka RC, Bagga J, Nguyen T, Cheng P, Kim JB, Pjanic M, Miano JM, Quertermous T. Coronary Disease-Associated Gene TCF21 Inhibits Smooth Muscle Cell Differentiation by Blocking the Myocardin-Serum Response Factor Pathway. Circ Res 2020; 126:517-529. [PMID: 31815603 PMCID: PMC7274203 DOI: 10.1161/circresaha.119.315968] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022]
Abstract
RATIONALE The gene encoding TCF21 (transcription factor 21) has been linked to coronary artery disease risk by human genome-wide association studies in multiple racial ethnic groups. In murine models, Tcf21 is required for phenotypic modulation of smooth muscle cells (SMCs) in atherosclerotic tissues and promotes a fibroblast phenotype in these cells. In humans, TCF21 expression inhibits risk for coronary artery disease. The molecular mechanism by which TCF21 regulates SMC phenotype is not known. OBJECTIVE To better understand how TCF21 affects the SMC phenotype, we sought to investigate the possible mechanisms by which it regulates the lineage determining MYOCD (myocardin)-SRF (serum response factor) pathway. METHODS AND RESULTS Modulation of TCF21 expression in human coronary artery SMC revealed that TCF21 suppresses a broad range of SMC markers, as well as key SMC transcription factors MYOCD and SRF, at the RNA and protein level. We conducted chromatin immunoprecipitation-sequencing to map SRF-binding sites in human coronary artery SMC, showing that binding is colocalized in the genome with TCF21, including at a novel enhancer in the SRF gene, and at the MYOCD gene promoter. In vitro genome editing indicated that the SRF enhancer CArG box regulates transcription of the SRF gene, and mutation of this conserved motif in the orthologous mouse SRF enhancer revealed decreased SRF expression in aorta and heart tissues. Direct TCF21 binding and transcriptional inhibition at colocalized sites were established by reporter gene transfection assays. Chromatin immunoprecipitation and protein coimmunoprecipitation studies provided evidence that TCF21 blocks MYOCD and SRF association by direct TCF21-MYOCD interaction. CONCLUSIONS These data indicate that TCF21 antagonizes the MYOCD-SRF pathway through multiple mechanisms, further establishing a role for this coronary artery disease-associated gene in fundamental SMC processes and indicating the importance of smooth muscle response to vascular stress and phenotypic modulation of this cell type in coronary artery disease risk.
Collapse
Affiliation(s)
- Manabu Nagao
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Qing Lyu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Rochester, NY 14624
| | - Quanyi Zhao
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Robert C Wirka
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Joetsaroop Bagga
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Trieu Nguyen
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Paul Cheng
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Milos Pjanic
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine & Dentistry, 601 Elmwood Ave, Rochester, NY 14624
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305
| |
Collapse
|
9
|
Song TF, Huang LW, Yuan Y, Wang HQ, He HP, Ma WJ, Huo LH, Zhou H, Wang N, Zhang TC. LncRNA MALAT1 regulates smooth muscle cell phenotype switch via activation of autophagy. Oncotarget 2018; 9:4411-4426. [PMID: 29435112 PMCID: PMC5796983 DOI: 10.18632/oncotarget.23230] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 11/14/2017] [Indexed: 12/26/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs), switching from a differentiated to a proliferative phenotype, contribute to various vascular diseases. However, the role of long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 MALAT1 in the phenotype switching of VSMCs remains unclear. Here, we report that the knockdown of MALAT1 promotes the transformation of smooth muscle cells from a proliferative phenotype to a differentiated phenotype. MALAT1 knockdown inhibited cellular proliferation and migration, leading to significant cell cycle arrest in the G2 phase. MALAT1 was downregulated in bone morphogenetic protein-7 (BMP-7)-induced cellular differentiation, while MALAT1 was upregulated in platelet-derived growth factor-BB (PDGF-BB)-induced cellular proliferation. PDGF induced the transformation of smooth muscle cells into a proliferative phenotype accompanied by an increase in autophagy. The downregulation of MALAT1 attenuated PDGF-BB-induced proliferation and migration by inhibiting autophagy. MALAT1 could act as a competing endogenous RNA (ceRNA) to regulate autophagy-related 7 (ATG7) gene expression by sponging miR142-3p. The present study reveals a novel mechanism by which MALAT1 promotes the transformation of smooth muscle cells from contraction to synthetic phenotypes.
Collapse
Affiliation(s)
- Tie-Feng Song
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Li-Wen Huang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Ying Yuan
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Hui-qin Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Hong-Peng He
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Wen-Jian Ma
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Li-Hong Huo
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Hao Zhou
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Nan Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, P.R. China
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Wuhan 430000, P.R. China
| |
Collapse
|
10
|
Chettimada S, Joshi SR, Dhagia V, Aiezza A, Lincoln TM, Gupte R, Miano JM, Gupte SA. Vascular smooth muscle cell contractile protein expression is increased through protein kinase G-dependent and -independent pathways by glucose-6-phosphate dehydrogenase inhibition and deficiency. Am J Physiol Heart Circ Physiol 2016; 311:H904-H912. [PMID: 27521420 PMCID: PMC5114469 DOI: 10.1152/ajpheart.00335.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/06/2016] [Indexed: 11/22/2022]
Abstract
Homeostatic control of vascular smooth muscle cell (VSMC) differentiation is critical for contractile activity and regulation of blood flow. Recently, we reported that precontracted blood vessels are relaxed and the phenotype of VSMC is regulated from a synthetic to contractile state by glucose-6-phosphate dehydrogenase (G6PD) inhibition. In the current study, we investigated whether the increase in the expression of VSMC contractile proteins by inhibition and knockdown of G6PD is mediated through a protein kinase G (PKG)-dependent pathway and whether it regulates blood pressure. We found that the expression of VSMC-restricted contractile proteins, myocardin (MYOCD), and miR-1 and miR-143 are increased by G6PD inhibition or knockdown. Importantly, RNA-sequence analysis of aortic tissue from G6PD-deficient mice revealed uniform increases in VSMC-restricted genes, particularly those regulated by the MYOCD-serum response factor (SRF) switch. Conversely, expression of Krüppel-like factor 4 (KLF4) is decreased by G6PD inhibition. Interestingly, the G6PD inhibition-induced expression of miR-1 and contractile proteins was blocked by Rp-β-phenyl-1,N2-etheno-8-bromo-guanosine-3',5'-cyclic monophosphorothioate, a PKG inhibitor. On the other hand, MYOCD and miR-143 levels are increased by G6PD inhibition through a PKG-independent manner. Furthermore, blood pressure was lower in the G6PD-deficient compared with wild-type mice. Therefore, our results suggest that the expression of VSMC contractile proteins induced by G6PD inhibition occurs via PKG1α-dependent and -independent pathways.
Collapse
MESH Headings
- Animals
- Aorta/drug effects
- Aorta/metabolism
- Blotting, Western
- Cattle
- Chromatography, Liquid
- Contractile Proteins/drug effects
- Contractile Proteins/genetics
- Contractile Proteins/metabolism
- Cyclic GMP-Dependent Protein Kinase Type I/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinase Type I/metabolism
- Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors
- Cyclic GMP-Dependent Protein Kinases/metabolism
- Gene Knockdown Techniques
- Glucosephosphate Dehydrogenase/antagonists & inhibitors
- Glucosephosphate Dehydrogenase/genetics
- Immunoprecipitation
- Kruppel-Like Factor 4
- Kruppel-Like Transcription Factors/drug effects
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Mice
- MicroRNAs/drug effects
- MicroRNAs/genetics
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/drug effects
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Polymerase Chain Reaction
- Rats
- Serum Response Factor/drug effects
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Tandem Mass Spectrometry
- Trans-Activators/drug effects
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Sukrutha Chettimada
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Sachindra Raj Joshi
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Vidhi Dhagia
- Pharmacology, New York Medical College, Valhalla, New York
| | - Alessandro Aiezza
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | | | - Rakhee Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| | - Joseph M Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Sachin A Gupte
- Biochemistry & Molecular Biology, University of South Alabama, Mobile, Alabama; Pharmacology, New York Medical College, Valhalla, New York
| |
Collapse
|
11
|
Moharil J, Lei P, Tian J, Gaile DP, Andreadis ST. Lentivirus Live Cell Array for Quantitative Assessment of Gene and Pathway Activation during Myogenic Differentiation of Mesenchymal Stem Cells. PLoS One 2015; 10:e0141365. [PMID: 26505747 PMCID: PMC4624764 DOI: 10.1371/journal.pone.0141365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/06/2015] [Indexed: 11/19/2022] Open
Abstract
Stem cell differentiation involves multiple cascades of transcriptional regulation that govern the cell fate. To study the real-time dynamics of this complex process, quantitative and high throughput live cell assays are required. Herein, we developed a lentiviral library of promoters and transcription factor binding sites to quantitatively capture the gene expression dynamics over a period of several days during myogenic differentiation of human mesenchymal stem cells (MSCs) harvested from two different anatomic locations, bone marrow and hair follicle. Our results enabled us to monitor the sequential activation of signaling pathways and myogenic gene promoters at various stages of differentiation. In conjunction with chemical inhibitors, the lentiviral array (LVA) results also revealed the relative contribution of key signaling pathways that regulate the myogenic differentiation. Our study demonstrates the potential of LVA to monitor the dynamics of gene and pathway activation during MSC differentiation as well as serve as a platform for discovery of novel molecules, genes and pathways that promote or inhibit complex biological processes.
Collapse
Affiliation(s)
- Janhavi Moharil
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
- Department of Biostatistics, University at Buffalo, State University of New York, Kimball, Buffalo, NY 14214–3000, United States of America
| | - Pedro Lei
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
| | - Jun Tian
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
| | - Daniel P. Gaile
- Department of Biostatistics, University at Buffalo, State University of New York, Kimball, Buffalo, NY 14214–3000, United States of America
| | - Stelios T. Andreadis
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, 908 Furnas Hall, Amherst, NY 14260–4200, United States of America
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Amherst, NY 14260–4200, United States of America
- Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, United States of America
- * E-mail:
| |
Collapse
|
12
|
Rinaldi B, Finicelli M, Donniacuo M, Di Bernardo G, Gritti G, Gaudio SD, Forte A, Peluso G, Cipollaro M, Rossi F, Galderisi U. G-CSF contributes at the healing of tunica media of arteriotomy-injured rat carotids by promoting differentiation of vascular smooth muscle cells. J Cell Physiol 2015; 231:215-23. [DOI: 10.1002/jcp.25074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 06/08/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Rinaldi
- Department of Experimental Medicine, Section of Pharmacology “L. Donatelli”, Centre of Excellence for Cardiovascular Diseases; Second University of Naples; Naples Italy
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Mauro Finicelli
- Institute of Biomedicine and Bioresources (IBBR); CNR; Naples Italy
| | - Maria Donniacuo
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Giovanni Di Bernardo
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Giulia Gritti
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
| | - Stefania Del Gaudio
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Amalia Forte
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | | | - Marilena Cipollaro
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology L. Donatelli, Regional Centre for Pharmacovigilance and Pharmacoepidemiology; Second University of Naples; Naples Italy
- Institute of Biomedicine and Bioresources (IBBR); CNR; Naples Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Biotechnology, and Molecular Biology Section; Second University of Naples; Naples Italy
| |
Collapse
|
13
|
Variability in vascular smooth muscle cell stretch-induced responses in 2D culture. Vasc Cell 2015; 7:7. [PMID: 26301087 PMCID: PMC4546126 DOI: 10.1186/s13221-015-0032-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 08/12/2015] [Indexed: 01/27/2023] Open
Abstract
The pulsatile nature of blood flow exposes vascular smooth muscle cells (VSMCs) in the vessel wall to mechanical stress, in the form of circumferential and longitudinal stretch. Cyclic stretch evokes VSMC proliferation, apoptosis, phenotypic switching, migration, alignment, and vascular remodeling. Given that these responses have been observed in many cardiovascular diseases, a defined understanding of their underlying mechanisms may provide critical insight into the pathophysiology of cardiovascular derangements. Cyclic stretch-triggered VSMC responses and their effector mechanisms have been studied in vitro using tension systems that apply either uniaxial or equibiaxial stretch to cells grown on an elastomer-bottomed culture plate and ex vivo by stretching whole vein segments with small weights. This review will focus mainly on VSMC responses to the in vitro application of mechanical stress, outlining the inconsistencies in acquired data, and comparing them to in vivo or ex vivo findings. Major discrepancies in data have been seen in mechanical stress-induced proliferation, apoptosis, and phenotypic switching responses, depending on the stretch conditions. These discrepancies stem from variations in stretch conditions such as degree, axis, duration, and frequency of stretch, wave function, membrane coating, cell type, cell passage number, culture media content, and choice of in vitro model. Further knowledge into the variables that cause these incongruities will allow for improvement of the in vitro application of cyclic stretch.
Collapse
|
14
|
Modulation of cysteine-rich protein 2 expression in vascular injury and atherosclerosis. Mol Biol Rep 2015; 41:7033-41. [PMID: 25034893 DOI: 10.1007/s11033-014-3591-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Vascular smooth muscle cells (VSMCs) of the arterial wall normally display a differentiated and contractile phenotype. In response to arterial injury, VSMCs switch to a synthetic phenotype, contributing to vascular remodeling. Cysteine-rich protein 2 (CRP2) is a cytoskeletal protein expressed in VSMCs and blunts VSMC migration in part by sequestering the scaffolding protein p130Cas at focal adhesions. CRP2 deficiency in mice increases neointima formation following arterial injury. The goal of this study was to use Csrp2 promoter-lacZ transgenic mice to analyze CRP2 expression during VSMC phenotypic modulation. In a neointima formation model after carotid artery cessation of blood flow, lacZ reporter activity and smooth muscle (SM) α-actin expression in the media were rapidly downregulated 4 days after carotid ligation. Fourteen days after ligation, there was a high level expression of both Csrp2 promoter activity and SM α-actin protein expression in neointimal cells. In atherosclerosis prone mice fed an atherogenic diet, Csrp2 promoter activity was detected within complex atherosclerotic lesions. Interestingly, Csrp2 promoter activity was also present in the fibrous caps of complicated atherosclerotic lesions, indicating that CRP2 might contribute to plaque stability. These findings support the concept that CRP2 contributes to the phenotypic modulation of VSMCs during vascular disease. Modulating transcription to increase CRP2 expression during vascular injury might attenuate vascular remodeling. In addition, increased CRP2 expression at the fibrous caps of advanced lesions might also serve to protect atherosclerotic plaques from rupture.
Collapse
|
15
|
Salemi S, Tremp M, Plock JA, Andersson KE, Gobet R, Sulser T, Eberli D. Differentiated adipose-derived stem cells for bladder bioengineering. Scand J Urol 2015; 49:407-14. [DOI: 10.3109/21681805.2015.1004642] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
16
|
Han Y, Slivano OJ, Christie CK, Cheng AW, Miano JM. CRISPR-Cas9 genome editing of a single regulatory element nearly abolishes target gene expression in mice--brief report. Arterioscler Thromb Vasc Biol 2015; 35:312-5. [PMID: 25538209 PMCID: PMC4304932 DOI: 10.1161/atvbaha.114.305017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE To ascertain the importance of a single regulatory element in the control of Cnn1 expression using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) genome editing. APPROACH AND RESULTS The CRISPR/Cas9 system was used to produce 3 of 18 founder mice carrying point mutations in an intronic CArG box of the smooth muscle cell-restricted Cnn1 gene. Each founder was bred for germline transmission of the mutant CArG box and littermate interbreeding to generate homozygous mutant (Cnn1(ΔCArG/ΔCArG)) mice. Quantitative reverse transcription polymerase chain reaction, Western blotting, and confocal immunofluorescence microscopy showed dramatic reductions in Cnn1 mRNA and CNN1 protein expression in Cnn1(ΔCArG/ΔCArG) mice with no change in other smooth muscle cell-restricted genes and little evidence of off-target edits elsewhere in the genome. In vivo chromatin immunoprecipitation assay revealed a sharp decrease in binding of serum response factor to the mutant CArG box. Loss of CNN1 expression was coincident with an increase in Ki-67 positive cells in the normal vessel wall. CONCLUSIONS CRISPR/Cas9 genome editing of a single CArG box nearly abolishes Cnn1 expression in vivo and evokes increases in smooth muscle cell DNA synthesis. This facile genome editing system paves the way for a new generation of studies designed to test the importance of individual regulatory elements in living animals, including regulatory variants in conserved sequence blocks linked to human disease.
Collapse
Affiliation(s)
- Yu Han
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Orazio J Slivano
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Christine K Christie
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Albert W Cheng
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.)
| | - Joseph M Miano
- From the Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester Medical Center, Rochester, NY (Y.H., O.J.S., C.K.C., J.M.M.); and Jackson Laboratories, Bar Harbor, ME (A.W.C.).
| |
Collapse
|
17
|
Biswas Shivhare S, Bulmer JN, Innes BA, Hapangama DK, Lash GE. Altered vascular smooth muscle cell differentiation in the endometrial vasculature in menorrhagia. Hum Reprod 2014; 29:1884-94. [PMID: 25006206 DOI: 10.1093/humrep/deu164] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
STUDY QUESTION How does the smooth muscle content and differentiation stage of vascular smooth muscle cells (VSMCs) in endometrial blood vessels change according to the different phases of the menstrual cycle and is this altered in women with menorrhagia? SUMMARY ANSWER The smooth muscle content (as a proportion of the vascular cross-sectional area) of endometrial blood vessels remained unchanged during the normal menstrual cycle and in menorrhagia; however, expression of the VSMC differentiation markers, smoothelin and calponin, was dysregulated in endometrial blood vessels in samples from women with menorrhagia compared with controls. WHAT IS KNOWN ALREADY Menorrhagia affects 30% of women of reproductive age and is the leading indication for hysterectomy. Previous studies have suggested important structural and functional roles for endometrial blood vessels, including impaired vascular contractility. Differentiation of VSMC from a synthetic to contractile state is associated with altered cellular phenotype that contributes to normal blood flow and pressure. This vascular maturation process has been little studied in endometrium both across the normal menstrual cycle and in menorrhagia. STUDY DESIGN, SIZE, DURATION Endometrial biopsies were taken from hysterectomy specimens or by pipelle biopsy prior to hysterectomy in controls without endometrial pathology and in women with menorrhagia (n = 7 for each of proliferative, early-secretory, mid-secretory and late-secretory phases for both groups). Biopsies were formalin fixed and embedded in paraffin wax. PARTICIPANTS/MATERIALS, SETTING, METHODS Paraffin-embedded sections were immunostained for α smooth muscle actin (αSMA), myosin heavy chain (MyHC), H-caldesmon, desmin, smoothelin and calponin (h1 or basic). VSMC content was measured in 25 αSMA(+) vascular cross sections per sample and expressed as a ratio of the muscular area:gross vascular cross-sectional area. VSMC differentiation was analysed by the presence/absence of differentiation markers compared with αSMA expression. Smoothelin and calponin expression was also analysed in relation to total number of blood vessels by double immunostaining for endothelial cell markers. MAIN RESULTS AND THE ROLE OF CHANCE Study of VSMC differentiation markers revealed decreased expression of calponin both in αSMA(+) vessels (P = 0.008) and in relation to total number of vessels (P = 0.001) in late secretory phase endometrium in menorrhagia compared with controls. Smoothelin expression in αSMA(+) vessels was increased (P = 0.03) in menorrhagia, although this was not significant in relation to the total number of vessels. In normal endometrium, the proportion of blood vessels expressing αSMA increased from 63% in proliferative endometrium to 81% in the late secretory phase (P = 0.002). The overall arterial muscle content did not differ between control and menorrhagia at any phase of the menstrual cycle, occupying 78-81% of gross vascular cross-sectional area during the different menstrual cycle phases. LIMITATIONS, REASONS FOR CAUTION This study included both straight and spiral arterioles and analysed only stratum functionalis. The VSMC differentiation with respect to αSMA expression is an observational study and the data are presented as presence or absence of the differentiation markers in each field of view, corresponding with the vascular cross sections included in the study of vascular muscle content. WIDER IMPLICATIONS OF THE FINDINGS Smoothelin and calponin have been widely implicated as important regulators of vascular tone, vascular contractility and rate of blood flow. Our results have uncovered a disparate pattern of calponin expression, potentially indicating a dysfunctional contraction mechanism in the endometrial blood vessels in menorrhagia, thus implicating calponin as a potential therapeutic target. STUDY FUNDING/COMPETING INTERESTS This study was funded by Wellbeing of Women (RG1342) and Newcastle University. There are no competing interests to declare. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Sourima Biswas Shivhare
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Judith N Bulmer
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Barbara A Innes
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dharani K Hapangama
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool Women's Hospital, Crown Street, Liverpool L8 7SS, UK
| | - Gendie E Lash
- Reproductive and Vascular Biology Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
18
|
Kirchmer MN, Franco A, Albasanz-Puig A, Murray J, Yagi M, Gao L, Dong ZM, Wijelath ES. Modulation of vascular smooth muscle cell phenotype by STAT-1 and STAT-3. Atherosclerosis 2014; 234:169-75. [PMID: 24657387 DOI: 10.1016/j.atherosclerosis.2014.02.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 02/24/2014] [Accepted: 02/27/2014] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Smooth muscle cell (SMC) de-differentiation is a key step that leads to pathological narrowing of blood vessels. De-differentiation involves a reduction in the expression of the SMC contractile genes that are the hallmark of quiescent SMCs. While there is considerable evidence linking inflammation to vascular diseases, very little is known about the mechanisms by which inflammatory signals lead to SMC de-differentiation. Given that the Signal Transducers and Activators of Transcription (STAT) transcriptional factors are the key signaling molecules activated by many inflammatory cytokines and growth factors, the aim of the present study was to determine if STAT transcriptional factors play a role SMC de-differentiation. METHODS AND RESULTS Using shRNA targeted to STAT-1 and STAT-3, we show by real time RT-PCR and Western immunoblots that STAT-1 significantly reduces SMC contractile gene expression. In contrast, STAT-3 promotes expression of SMC contractile genes. Over-expression studies of STAT-1 and STAT-3 confirmed our observation that STAT-1 down-regulates whereas STAT-3 promotes SMC contractile gene expression. Bioinformatics analysis shows that promoters of all SMC contractile genes contain STAT binding sites. Finally, using ChIP analysis, we show that both STAT-1 and STAT-3 associate with the calponin gene. CONCLUSION These data indicate that the balance of STAT-1 and STAT-3 influences the differentiation status of SMCs. Increased levels of STAT-1 promote SMC de-differentiation, whereas high levels of STAT-3 drive SMC into a more mature phenotype. Thus, inhibition of STAT-1 may represent a novel target for therapeutic intervention in the control of vascular diseases such as atherosclerosis and restenosis.
Collapse
Affiliation(s)
- Mayumi Namekata Kirchmer
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Anais Franco
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Adaia Albasanz-Puig
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Jacqueline Murray
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Mayumi Yagi
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Lu Gao
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Zhao Ming Dong
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA
| | - Errol S Wijelath
- Department of Surgery, Division of Vascular Surgery, VA Puget Sound Health Care System and The University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
19
|
Tajsic T, Morrell NW. Smooth muscle cell hypertrophy, proliferation, migration and apoptosis in pulmonary hypertension. Compr Physiol 2013; 1:295-317. [PMID: 23737174 DOI: 10.1002/cphy.c100026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Pulmonary hypertension is a multifactorial disease characterized by sustained elevation of pulmonary vascular resistance (PVR) and pulmonary arterial pressure (PAP). Central to the pathobiology of this disease is the process of vascular remodelling. This process involves structural and functional changes to the normal architecture of the walls of pulmonary arteries (PAs) that lead to increased muscularization of the muscular PAs, muscularization of the peripheral, previously nonmuscular, arteries of the respiratory acinus, formation of neointima, and formation of plexiform lesions. Underlying or contributing to the development of these lesions is hypertrophy, proliferation, migration, and resistance to apoptosis of medial cells and this article is concerned with the cellular and molecular mechanisms of these processes. In the first part of the article we focus on the concept of smooth muscle cell phenotype and the difficulties surrounding the identification and characterization of the cell/cells involved in the remodelling of the vessel media and we review the general mechanisms of cell hypertrophy, proliferation, migration and apoptosis. Then, in the larger part of the article, we review the factors identified thus far to be involved in PH intiation and/or progression and review and discuss their effects on pulmonary artery smooth muscle cells (PASMCs) the predominant cells in the tunica media of PAs.
Collapse
Affiliation(s)
- Tamara Tajsic
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | |
Collapse
|
20
|
Fintha A, Gasparics Á, Fang L, Erdei Z, Hamar P, Mózes MM, Kökény G, Rosivall L, Sebe A. Characterization and role of SCAI during renal fibrosis and epithelial-to-mesenchymal transition. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:388-400. [PMID: 23178076 DOI: 10.1016/j.ajpath.2012.10.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Revised: 09/27/2012] [Accepted: 10/16/2012] [Indexed: 01/17/2023]
Abstract
During progressive tubulointerstitial fibrosis, renal tubular epithelial cells transform into α-smooth muscle actin (SMA)-expressing myofibroblasts via epithelial-to-mesenchymal transition (EMT). SMA expression is regulated by transforming growth factor (TGF)-β1 and cell contact disruption, through signaling events targeting the serum response factor-myocardin-related transcription factor (MRTF) complex. MRTFs are important regulators of fibrosis, tumor cell invasion, and metastasis. Consistent with the role of MRTFs in tumor progression, suppressor of cancer cell invasion (SCAI) was recently identified as a negative regulator of MRTF. Herein, we studied the role of SCAI in a fibrotic EMT model established on LLC-PK1 cells. SCAI overexpression prevented SMA promoter activation induced by TGF-β1. When co-expressed, it inhibited the stimulatory effects of MRTF-A, MRTF-B or the constitutive active forms of RhoA, Rac1, or Cdc42 on the SMA promoter. SCAI interfered with TGF-β1-induced SMA, connective tissue growth factor, and calponin protein expression; it rescued TGF-β1-induced E-cadherin down-regulation. IHC studies on human kidneys showed that SCAI expression is reduced during fibrosis. Kidneys of diabetic rats and mice with unilateral ureteral obstruction depicted significant loss of SCAI expression. In parallel with the decrease of SCAI protein expression, diabetic rat and mouse kidneys with unilateral ureteral obstruction showed SMA expression, as evidenced by using Western blot analysis. Finally, TGF-β1 treatment of LLC-PK1 cells attenuated SCAI protein expression. These data suggest that SCAI is a novel transcriptional cofactor that regulates EMT and renal fibrosis.
Collapse
Affiliation(s)
- Attila Fintha
- 2(nd) Department of Pathology, Semmelweis University, 1089 Budapest, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Expression and promoter analysis of a highly restricted integrin alpha gene in vascular smooth muscle. Gene 2012; 513:82-9. [PMID: 23142384 DOI: 10.1016/j.gene.2012.10.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 09/18/2012] [Accepted: 10/29/2012] [Indexed: 11/19/2022]
Abstract
Full genome annotation requires gene expression analysis and elucidation of promoter activity. Here, we analyzed the expression and promoter of a highly restricted integrin gene, Itga8. RNase protection and quantitative RT-PCR showed Itga8 to be expressed most abundantly in vascular smooth muscle cells (SMC). Transcription start site mapping of Itga8 revealed the immediate 5' promoter region to be poorly conserved with orthologous sequences in the human genome. Further comparative sequence analysis showed a number of conserved non-coding sequence modules around the Itga8 gene. The immediate promoter region and an upstream conserved sequence module were each found to contain a CArG box, which is a binding site for serum response factor (SRF). Luciferase reporter assays revealed activity of several Itga8 promoter constructs with no apparent restricted activity to SMC types. Further, neither SRF nor its coactivator, Myocardin (MYOCD), was able to induce several distinct Itga8 promoter constructs. Transgenic mouse studies failed to reveal Itga8 promoter activity, indicating distal regulatory elements likely control this gene's in vivo expression profile. Interestingly, although the promoter was unresponsive to SRF/MYOCD, the endogenous Itga8 gene showed increases in expression upon ectopic MYOCD expression even though knockdown of SRF both in vitro and in vivo failed to demonstrate a corresponding change in Itga8. Collectively, these data demonstrate that Itga8 expression is CArG-SRF independent, but MYOCD dependent through an as yet unknown sequence module that is distal from the promoter region.
Collapse
|
22
|
Shi J, Dong Y, Cui MZ, Xu X. Lysophosphatidic acid induces increased BACE1 expression and Aβ formation. Biochim Biophys Acta Mol Basis Dis 2012; 1832:29-38. [PMID: 23036978 DOI: 10.1016/j.bbadis.2012.09.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/19/2012] [Accepted: 09/25/2012] [Indexed: 01/08/2023]
Abstract
The abnormal production and accumulation of β-amyloid peptide (Aβ), which is produced from amyloid precursor protein (APP) by the sequential actions of β-secretase and γ-secretase, are thought to be the initial causative events in the development of Alzheimer's disease (AD). Accumulating evidence suggests that vascular factors play an important role in the pathogenesis of AD. Specifically, studies have suggested that one vascular factor in particular, oxidized low density lipoprotein (oxLDL), may play an important role in regulating Aβ formation in AD. However, the mechanism by which oxLDL modulates Aβ formation remains elusive. In this study, we report several new findings that provide biochemical evidence suggesting that the cardiovascular risk factor oxLDL may contribute to Alzheimer's disease by increasing Aβ production. First, we found that lysophosphatidic acid (LPA), the most bioactive component of oxLDL induces increased production of Aβ. Second, our data strongly indicate that LPA induces increased Aβ production via upregulating β-secretase expression. Third, our data strongly support the notion that different isoforms of protein kinase C (PKC) may play different roles in regulating APP processing. Specifically, most PKC members, such as PKCα, PKCβ, and PKCε, are implicated in regulating α-secretase-mediated APP processing; however, PKCδ, a member of the novel PKC subfamily, is involved in LPA-induced upregulation of β-secretase expression and Aβ production. These findings may contribute to a better understanding of the mechanisms by which the cardiovascular risk factor oxLDL is involved in Alzheimer's disease.
Collapse
Affiliation(s)
- Jing Shi
- Department of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | |
Collapse
|
23
|
Pfisterer L, Feldner A, Hecker M, Korff T. Hypertension impairs myocardin function: a novel mechanism facilitating arterial remodelling. Cardiovasc Res 2012; 96:120-9. [PMID: 22843699 DOI: 10.1093/cvr/cvs247] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AIMS Hypertension evokes detrimental changes in the arterial vessel wall that facilitate stiffening and thus lead to a further rise in mean blood pressure, eventually causing heart failure. The underlying pathophysiological remodelling process is elicited by an increase in wall stress (WS) and is strictly dependent on the activation of vascular smooth muscle cells (SMC). However, it remains unclear as to why these cells fail to maintain their contractile and quiescent phenotype in a hypertensive environment. METHODS AND RESULTS In this context, we reveal that the knockdown of myocardin--a pivotal transcriptional determinant of the contractile SMC phenotype--is sufficient to induce SMC proliferation. In line with this observation, immunofluorescence analysis of the media of remodelling arteries from hypertensive mice demonstrated a significant decrease in the abundance of myocardin and an increase in SMC proliferation. Subsequent analyses of isolated perfused mouse arteries and human cultured SMCs exposed to cyclic stretch (i.e. mimicking one component of WS) suggested that this biomechanical force facilitates serine phosphorylation of myocardin. Furthermore, this biomechanical stimulus promotes rapid translocation of myocardin from the nucleus to the cytoplasm, inhibits its mRNA expression, and causes proteasomal degradation of the cytoplasmic protein. CONCLUSIONS Collectively, these findings suggest that hypertension negates the activity of myocardin in SMCs on multiple levels, hence eliminating a crucial determinant of SMC quiescence. This mechanism may control the initial switch from the contractile towards the synthetic SMC phenotype during hypertension and may offer an interesting novel approach to prevent cardiovascular disease.
Collapse
Affiliation(s)
- Larissa Pfisterer
- Institute of Physiology and Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
24
|
Potential role of LMP2 as an anti-oncogenic factor in human uterine leiomyosarcoma: morphological significance of calponin h1. FEBS Lett 2012; 586:1824-31. [PMID: 22659265 DOI: 10.1016/j.febslet.2012.05.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/12/2012] [Accepted: 05/14/2012] [Indexed: 01/04/2023]
Abstract
Uterine leiomyosarcoma (LMS) is a highly metastatic smooth muscle neoplasm for which calponin h1 is suspected to have a biological role as a tumor-suppressor. We earlier reported that LMP2-null mice spontaneously develop uterine LMS through malignant transformation of the myometrium, thus implicating this protein as an anti-tumorigenic candidate as well. In the present study, we show that LMP2 may negatively regulate LMS independently of its role in the proteasome. Moreover, several lines of evidence indicate that although calponin h1 does not directly influence tumorigenesis, it clearly affects LMP2-induced cellular morphological changes. Modulation of LMP2 may lead to new therapeutic approaches in human uterine LMS.
Collapse
|
25
|
Abstract
Smooth muscle cells (SMCs) possess remarkable phenotypic plasticity that allows rapid adaptation to fluctuating environmental cues, including during development and progression of vascular diseases such as atherosclerosis. Although much is known regarding factors and mechanisms that control SMC phenotypic plasticity in cultured cells, our knowledge of the mechanisms controlling SMC phenotypic switching in vivo is far from complete. Indeed, the lack of definitive SMC lineage-tracing studies in the context of atherosclerosis, and difficulties in identifying phenotypically modulated SMCs within lesions that have down-regulated typical SMC marker genes, and/or activated expression of markers of alternative cell types including macrophages, raise major questions regarding the contributions of SMCs at all stages of atherogenesis. The goal of this review is to rigorously evaluate the current state of our knowledge regarding possible phenotypes exhibited by SMCs within atherosclerotic lesions and the factors and mechanisms that may control these phenotypic transitions.
Collapse
Affiliation(s)
- Delphine Gomez
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, 415 Lane Road, PO Box 801394, Room 1322 Medical Research Building 5, Charlottesville, VA 22908, USA
| | | |
Collapse
|
26
|
Nanda V, Miano JM. Leiomodin 1, a new serum response factor-dependent target gene expressed preferentially in differentiated smooth muscle cells. J Biol Chem 2011; 287:2459-67. [PMID: 22157009 DOI: 10.1074/jbc.m111.302224] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Smooth muscle cell (SMC) differentiation is defined largely by a number of cell-restricted genes governed directly by the serum response factor (SRF)/myocardin (MYOCD) transcriptional switch. Here, we describe a new SRF/MYOCD-dependent, SMC-restricted gene known as Leiomodin 1 (Lmod1). Conventional and quantitative RT-PCRs indicate that Lmod1 mRNA expression is enriched in SMC-containing tissues of the mouse, whereas its two paralogs, Lmod2 and Lmod3, exhibit abundant expression in skeletal and cardiac muscle with very low levels in SMC-containing tissues. Western blotting and immunostaining of various adult and embryonic mouse tissues further confirm SMC-specific expression of the LMOD1 protein. Comparative genomic analysis of the human LMOD1 and LMOD2 genes with their respective mouse and rat orthologs shows high conservation between the three exons and several noncoding sequences, including the immediate 5' promoter region. Two conserved CArG boxes are present in both the LMOD1 and LMOD2 promoter regions, although LMOD1 displays much higher promoter activity and is more responsive to SRF/MYOCD stimulation. Gel shift assays demonstrate clear binding between SRF and the two CArG boxes in human LMOD1. Although the CArG boxes in LMOD1 and LMOD2 are similar, only LMOD1 displays SRF or MYOCD-dependent activation. Transgenic mouse studies reveal wild type LMOD1 promoter activity in cardiac and vascular SMC. Such activity is abolished upon mutation of both CArG boxes. Collectively, these data demonstrate that Lmod1 is a new SMC-restricted SRF/MYOCD target gene.
Collapse
Affiliation(s)
- Vivek Nanda
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | |
Collapse
|
27
|
Alexander MR, Owens GK. Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. Annu Rev Physiol 2011; 74:13-40. [PMID: 22017177 DOI: 10.1146/annurev-physiol-012110-142315] [Citation(s) in RCA: 556] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The vascular smooth muscle cell (SMC) in adult animals is a highly specialized cell whose principal function is contraction. However, this cell displays remarkable plasticity and can undergo profound changes in phenotype during repair of vascular injury, during remodeling in response to altered blood flow, or in various disease states. There has been extensive progress in recent years in our understanding of the complex mechanisms that control SMC differentiation and phenotypic plasticity, including the demonstration that epigenetic mechanisms play a critical role. In addition, recent evidence indicates that SMC phenotypic switching in adult animals involves the reactivation of embryonic stem cell pluripotency genes and that mesenchymal stem cells may be derived from SMC and/or pericytes. This review summarizes the current state of our knowledge in this field and identifies some of the key unresolved challenges and questions that we feel require further study.
Collapse
Affiliation(s)
- Matthew R Alexander
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia 22908, USA.
| | | |
Collapse
|
28
|
Long X, Slivano OJ, Cowan SL, Georger MA, Lee TH, Miano JM. Smooth muscle calponin: an unconventional CArG-dependent gene that antagonizes neointimal formation. Arterioscler Thromb Vasc Biol 2011; 31:2172-80. [PMID: 21817093 PMCID: PMC3179981 DOI: 10.1161/atvbaha.111.232785] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Smooth muscle calponin (CNN1) contains multiple conserved intronic CArG elements that bind serum response factor and display enhancer activity in vitro. The objectives here were to evaluate these CArG elements for activity in transgenic mice and determine the effect of human CNN1 on injury-induced vascular remodeling. METHODS AND RESULTS Mice carrying a lacZ reporter under control of intronic CArG elements in the human CNN1 gene failed to show smooth muscle cell (SMC)-restricted activity. However, deletion of the orthologous sequences in mice abolished endogenous Cnn1 promoter activity, suggesting their necessity for in vivo Cnn1 expression. Mice carrying a 38-kb bacterial artificial chromosome (BAC) harboring the human CNN1 gene displayed SMC- restricted expression of the corresponding CNN1 protein, as measured by immunohistochemistry and Western blotting. Extensive BAC recombineering studies revealed the absolute necessity of a single intronic CArG element for correct SMC-restricted expression of human CNN1. Overexpressing human CNN1 suppressed neointimal formation following arterial injury. Mice with an identical BAC carrying mutations in CArG elements that inhibit human CNN1 expression showed outward remodeling and neointimal formation. CONCLUSIONS A single intronic CArG element is necessary but insufficient for proper CNN1 expression in vivo. CNN1 overexpression antagonizes arterial injury-induced neointimal formation.
Collapse
MESH Headings
- Animals
- Binding Sites
- Blotting, Western
- Calcium-Binding Proteins/deficiency
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/pathology
- Cell Line
- Cell Proliferation
- Chromosomes, Artificial, Bacterial
- Disease Models, Animal
- Gene Expression Regulation
- Genes, Reporter
- Humans
- Immunohistochemistry
- Introns
- Lac Operon
- Luciferases/genetics
- Luciferases/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microfilament Proteins/deficiency
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Promoter Regions, Genetic
- Rats
- Serum Response Element
- Serum Response Factor/metabolism
- Transfection
- Tunica Intima/metabolism
- Tunica Intima/pathology
- Calponins
Collapse
Affiliation(s)
- Xiaochun Long
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Orazio J. Slivano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Sarah L. Cowan
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Mary A. Georger
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Ting-Hein Lee
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Joseph M. Miano
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
29
|
Asaduzzaman M, Kinoshita S, Siddique BS, Asakawa S, Watabe S. Multiple cis-elements in the 5'-flanking region of embryonic/larval fast-type of the myosin heavy chain gene of torafugu, MYH(M743-2), function in the transcriptional regulation of its expression. Gene 2011; 489:41-54. [PMID: 21893174 DOI: 10.1016/j.gene.2011.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 08/10/2011] [Accepted: 08/17/2011] [Indexed: 01/10/2023]
Abstract
The myosin heavy chain gene, MYH(M743-2), is highly expressed in fast muscle fibers of torafugu embryos and larvae, suggesting its functional roles for embryonic and larval muscle development. However, the transcriptional regulatory mechanism involved in its expression remained unknown. Here, we analyzed the 2075bp 5'-flanking region of torafugu MYH(M743-2) to examine the spatial and temporal regulation by using transgenic and transient expression techniques in zebrafish embryos. Combining both transient and transgenic analyses, we demonstrated that the 2075bp 5'-flanking sequences was sufficient for its expression in skeletal, craniofacial and pectoral fin muscles. The immunohistochemical observation revealed that the zebrafish larvae from the stable transgenic line consistently expressed enhanced green fluorescent protein (EGFP) in fast muscle fibers. Promoter deletion analyses demonstrated that the minimum 468bp promoter region could direct MYH(M743-2) expression in zebrafish larvae. We discovered that the serum response factor (SRF)-like binding sites are required for promoting MYH(M743-2) expression and myoblast determining factor (MyoD) and myocyte enhancer factor-2 (MEF2) binding sites participate in the transcriptional control of MYH(M743-2) expression in fast skeletal muscles. We further discovered that MyoD binding sites, but not MEF2, participate in the transcriptional regulation of MYH(M743-2) expression in pectoral fin and craniofacial muscles. These results clearly demonstrated that multiple cis-elements in the 5'-flanking region of MYH(M743-2) function in the transcriptional control of its expression.
Collapse
Affiliation(s)
- Md Asaduzzaman
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo 113-8657, Japan
| | | | | | | | | |
Collapse
|
30
|
Benson CC, Zhou Q, Long X, Miano JM. Identifying functional single nucleotide polymorphisms in the human CArGome. Physiol Genomics 2011; 43:1038-48. [PMID: 21771879 DOI: 10.1152/physiolgenomics.00098.2011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Regulatory SNPs (rSNPs) reside primarily within the nonprotein coding genome and are thought to disturb normal patterns of gene expression by altering DNA binding of transcription factors. Nevertheless, despite the explosive rise in SNP association studies, there is little information as to the function of rSNPs in human disease. Serum response factor (SRF) is a widely expressed DNA-binding transcription factor that has variable affinity to at least 1,216 permutations of a 10 bp transcription factor binding site (TFBS) known as the CArG box. We developed a robust in silico bioinformatics screening method to evaluate sequences around RefSeq genes for conserved CArG boxes. Utilizing a predetermined phastCons threshold score, we identified 8,252 strand-specific CArGs within an 8 kb window around the transcription start site of 5,213 genes, including all previously defined SRF target genes. We then interrogated this CArG dataset for the presence of previously annotated common polymorphisms. We found a total of 118 unique CArG boxes harboring a SNP within the 10 bp CArG sequence and 1,130 CArG boxes with SNPs located just outside the CArG element. Gel shift and luciferase reporter assays validated SRF binding and functional activity of several new CArG boxes. Importantly, SNPs within or just outside the CArG box often resulted in altered SRF binding and activity. Collectively, these findings demonstrate a powerful approach to computationally define rSNPs in the human CArGome and provide a foundation for similar analyses of other TFBS. Such information may find utility in genetic association studies of human disease where little insight is known regarding the functionality of rSNPs.
Collapse
Affiliation(s)
- Craig C Benson
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | |
Collapse
|
31
|
Culture media for the differentiation of mesenchymal stromal cells. Acta Biomater 2011; 7:463-77. [PMID: 20688199 DOI: 10.1016/j.actbio.2010.07.037] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Revised: 07/20/2010] [Accepted: 07/27/2010] [Indexed: 02/08/2023]
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues such as bone marrow aspirates, fat or umbilical cord blood. These cells have the ability to proliferate in vitro and differentiate into a series of mesoderm-type lineages, including osteoblasts, chondrocytes, adipocytes, myocytes and vascular cells. Due to this ability, MSCs provide an appealing source of progenitor cells which may be used in the field of tissue regeneration for both research and clinical purposes. The key factors for successful MSC proliferation and differentiation in vitro are the culture conditions. Hence, we here summarize the culture media and their compositions currently available for the differentiation of MSCs towards osteogenic, chondrogenic, adipogenic, endothelial and vascular smooth muscle phenotypes. However, optimal combination of growth factors, cytokines and serum supplements and their concentration within the media is essential for the in vitro culture and differentiation of MSCs and thereby for their application in advanced tissue engineering.
Collapse
|
32
|
Werth D, Grassi G, Konjer N, Dapas B, Farra R, Giansante C, Kandolf R, Guarnieri G, Nordheim A, Heidenreich O. Proliferation of human primary vascular smooth muscle cells depends on serum response factor. Eur J Cell Biol 2010; 89:216-24. [PMID: 20096952 DOI: 10.1016/j.ejcb.2009.12.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Smooth muscle cells (SMCs) can switch between a differentiated/contractile and an alternative proliferative phenotype. The transcription factor serum response factor (SRF) has been implicated in the regulation of gene expression profiles determining both phenotypes. Whereas strong evidence exists for a role of SRF in SMC differentiation, the contribution of SRF to SMC proliferation is less well defined. For primary human vascular SMCs in particular, existing data are non-conclusive. To study SRF functions in primary human vascular SMCs, we used an siRNA approach. siRNA-mediated SRF suppression affected the expression of established SRF target genes such as smooth muscle alpha-actin (ACTA2) or SM22alpha (TAGLN) and decreased both F-actin formation and cell migration. Furthermore, SRF knockdown caused a cell-cycle arrest in G1 associated with reduced hyperphosphorylated pRB, cyclin A and SKP2 levels, and increased p27(kip1) (CDKN1B) protein levels. SRF-depleted cells expressed senescence-associated beta-galactosidase indicating an irreversible G1 arrest. siRNA-mediated suppression of SKP2 triggered senescence to a similar extent as SRF depletion, indicating that SRF knockdown-induced senescence may be dependent on a decrease in SKP2. Thus, SRF is an essential regulator of primary human vascular SMC proliferation and senescence. Interfering with SRF function may therefore be a promising strategy for the treatment of hyperproliferative SMC disorders such as atherosclerosis and in-stent restenosis.
Collapse
Affiliation(s)
- Daniela Werth
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, Eberhard Karls University of Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen CH, Wu ML, Lee YC, Layne MD, Yet SF. Intronic CArG box regulates cysteine-rich protein 2 expression in the adult but not in developing vasculature. Arterioscler Thromb Vasc Biol 2010; 30:835-42. [PMID: 20075421 DOI: 10.1161/atvbaha.109.197251] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE An absence of cysteine-rich protein 2 (CRP2) enhances vascular smooth muscle cell (VSMC) migration and increases neointima formation after arterial injury; therefore, CRP2 plays an important role in the response to vascular injury. The goal of the present study was to elucidate the molecular mechanisms that preserve CRP2 expression in the adult vasculature and thus might serve to inhibit the response to injury. METHODS AND RESULTS We generated a series of transgenic mice harboring potential Csrp2 regulatory regions with a lacZ reporter. We determined that the 12-kb first intron was necessary for transgene activity in adult but not in developing vasculature. Within the intron we identified a 6.3-kb region that contains 2 CArG boxes. Serum response factor preferentially bound to CArG2 box in gel mobility shift and chromatin immunoprecipitation assays; additionally, serum response factor coactivator myocardin factors activated CRP2 expression via the CArG2 box. Mutational analysis revealed that CArG2 box was important in directing lacZ expression in VSMC of adult vessels. CONCLUSIONS Although CRP2 expression during development is independent of CArG box regulatory sites, CRP2 expression in adult VSMC requires CArG2 element within the first intron. Our results suggest that distinct mechanisms regulate CRP2 expression in VSMC that are controlled by separate embryonic and adult regulatory modules.
Collapse
Affiliation(s)
- Chung-Huang Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| | | | | | | | | |
Collapse
|
34
|
Kimura Y, Morita T, Hayashi K, Miki T, Sobue K. Myocardin functions as an effective inducer of growth arrest and differentiation in human uterine leiomyosarcoma cells. Cancer Res 2010; 70:501-11. [PMID: 20068148 DOI: 10.1158/0008-5472.can-09-1469] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Myocardin is an important transcriptional regulator in smooth and cardiac muscle development. We noticed that the expression of myocardin was markedly downregulated in human uterine leiomyosarcoma cells. Restoration of myocardin expression induced the reexpression of smooth muscle marker proteins and the formation of well-developed actin fibers. A concomitant increase in the expression of a cyclin-dependent kinase inhibitor, p21, led to significantly reduced cell proliferation, via p21's inhibition of the G(1)-S transition. A p21 promoter-reporter assay showed that myocardin markedly increased p21's promoter activity. Furthermore, a serum response factor (SRF)-binding cis-element CArG box in the p21 promoter region was required for this myocardin effect. Chromatin immunoprecipitation and DNA-protein binding assays showed that myocardin indirectly bound to the CArG box in the p21 promoter through the interaction with SRF. Furthermore, immunohistochemistry revealed that the levels of myocardin and p21 were both lower in leiomyosarcoma samples than in normal smooth muscle tissue. Taken together, our results indicate that the downregulation of myocardin expression facilitates cell cycle progression via the reduction of p21 expression in human leimyosarcomas and suggest that myocardin could be a useful therapeutic target for this disease.
Collapse
Affiliation(s)
- Yasunori Kimura
- Department of Neuroscience, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | | | | | | | | |
Collapse
|
35
|
Ailawadi G, Moehle CW, Pei H, Walton SP, Yang Z, Kron IL, Lau CL, Owens GK. Smooth muscle phenotypic modulation is an early event in aortic aneurysms. J Thorac Cardiovasc Surg 2009; 138:1392-9. [PMID: 19931668 DOI: 10.1016/j.jtcvs.2009.07.075] [Citation(s) in RCA: 250] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 06/30/2009] [Accepted: 07/29/2009] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Vascular smooth muscle cells can undergo profound changes in phenotype, defined by coordinated repression of smooth muscle cell marker genes and production of matrix metalloproteinases in response to injury. However, little is known of the role of smooth muscle cells in aortic aneurysms. We hypothesized that smooth muscle cells undergo phenotypic modulation early in the development of aortic aneurysms. METHODS Abdominal aortas from C57B6 mice (n = 79) were perfused with elastase or saline (control) and harvested at 1, 3, 7, or 14 days. Aortas were analyzed by means of quantitative polymerase chain reaction and immunohistochemistry for smooth muscle cell marker genes, including SM22A, smooth muscle alpha-actin, and matrix metalloproteinases 2 and 9. In complimentary experiments human aneurysms (n = 10) and control aorta (n = 10) were harvested at the time of surgical intervention and analyzed. RESULTS By 14 days, aortic diameter was larger after elastase perfusion compared with control diameter (100% +/- 9.6% vs 59.5% +/- 18.9%, P = .0002). At 7 days, elastase-perfused mice had a 78% and 85% reduction in SM22 alpha and smooth muscle alpha-actin expression, respectively, compared with that seen in control animals well before aneurysms were present, and these values remained repressed at 14 days. Immunohistochemistry confirmed less SM22 alpha and smooth muscle alpha-actin in experimental aneurysms at 14 days in concert with increased matrix metalloproteinase 2 and 9 expression at 7 and 14 days. Similarly, human aneurysms had less SM22 alpha and smooth muscle alpha-actin and increased matrix metalloproteinase 2 and 9 staining, compared with control values, as determined by means of quantitative polymerase chain reaction. CONCLUSIONS Aneurysms demonstrate smooth muscle cell phenotypic modulation characterized by downregulation of smooth muscle cell marker genes and upregulation of matrix metalloproteinases. These events in experimental models occur before aneurysm formation. Targeting smooth muscle cells to a reparative phenotype might provide a novel therapy in the treatment of aortic aneurysms.
Collapse
Affiliation(s)
- Gorav Ailawadi
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Gong Z, Calkins G, Cheng EC, Krause D, Niklason LE. Influence of culture medium on smooth muscle cell differentiation from human bone marrow-derived mesenchymal stem cells. Tissue Eng Part A 2009; 15:319-30. [PMID: 19115826 DOI: 10.1089/ten.tea.2008.0161] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) represent an appealing source of smooth muscle cells (SMCs) for engineering small-diameter vascular grafts due to the limited availability and replicative capacity of somatic SMCs. However, lack of standardization of hMSC culture conditions has limited some progress in hMSC research. Because, at the moment, a chemically defined, serum-free medium without growth factors is not capable of amplifying hMSCs in vitro, the usage of serum (either human serum or fetal bovine serum [FBS]) continues in hMSC research. The emergence of commercial hMSCs and hMSC media opened a series of questions regarding the compatibility of commercial and homemade hMSCs and hMSC media. In this study, two types of commonly used FBS-containing hMSC media-MSCGM (containing 10% FBS) and MesenPro (containing 2% FBS), along with our homemade medium (low-glucose Dulbecco's modified Eagle's medium plus 10% selected lot FBS)-were compared in their ability to support SMC differentiation from hMSCs. The effects of FBS level, medium supplements (ascorbic acid, copper, etc.), and growth factors (transforming growth factor beta1) were also examined for their impact on SMC differentiation. It was discovered that MesenPro and transforming growth factor beta1 are the strongest SMC inducers from hMSCs. In contrast, hMSCs grown in homemade (10% Dulbecco's modified Eagle's medium) and commercial MSCGM media remained undifferentiated. FBS concentration did not affect SMC differentiation when 10% FBS was compared with 2%. Finally, the mechanism underlying SMC differentiation from hMSCs grown in FBS-containing medium was explored by following the expression changes of serum response factor during the establishment of hMSC culture.
Collapse
Affiliation(s)
- Zhaodi Gong
- Department of Anesthesiology & Biomedical Engineering, Vascular Biology & Transplantation (VBT) Program, Yale University School of Medicine, New Haven, Connecticut 06520-8089, USA
| | | | | | | | | |
Collapse
|
37
|
Long X, Tharp DL, Georger MA, Slivano OJ, Lee MY, Wamhoff BR, Bowles DK, Miano JM. The smooth muscle cell-restricted KCNMB1 ion channel subunit is a direct transcriptional target of serum response factor and myocardin. J Biol Chem 2009; 284:33671-82. [PMID: 19801679 DOI: 10.1074/jbc.m109.050419] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Large conductance calcium-activated potassium (MaxiK) channels play a pivotal role in maintaining normal arterial tone by regulating the excitation-contraction coupling process. MaxiK channels comprise alpha and beta subunits encoded by Kcnma and the cell-restricted Kcnmb genes, respectively. Although the functionality of MaxiK channel subunits has been well studied, the molecular regulation of their transcription and modulation in smooth muscle cells (SMCs) is incomplete. Using several model systems, we demonstrate down-regulation of Kcnmb1 mRNA upon SMC phenotypic modulation in vitro and in vivo. As part of a broad effort to define all functional CArG elements in the genome (i.e. the CArGome), we discovered two conserved CArG boxes located in the proximal promoter and first intron of the human KCNMB1 gene. Gel shift and chromatin immunoprecipitation assays confirmed serum response factor (SRF) binding to both CArG elements. A luciferase assay showed myocardin (MYOCD)-mediated transactivation of the KCNMB1 promoter in a CArG element-dependent manner. In vivo analysis of the human KCNMB1 promoter disclosed activity in embryonic heart and aortic SMCs; mutation of both conserved CArG elements completely abolished in vivo promoter activity. Forced expression of MYOCD increased Kcnmb1 expression in a variety of rodent and human non-SMC lines with no effect on expression of the Kcnma1 subunit. Conversely, knockdown of Srf resulted in decreases of endogenous Kcnmb1. Functional studies demonstrated MYOCD-induced, iberiotoxin-sensitive potassium currents in porcine coronary SMCs. These results reveal the first ion channel subunit as a direct target of SRF-MYOCD transactivation, providing further insight into the role of MYOCD as a master regulator of the SMC contractile phenotype.
Collapse
Affiliation(s)
- Xiaochun Long
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Blaker AL, Taylor JM, Mack CP. PKA-dependent phosphorylation of serum response factor inhibits smooth muscle-specific gene expression. Arterioscler Thromb Vasc Biol 2009; 29:2153-60. [PMID: 19778940 DOI: 10.1161/atvbaha.109.197285] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Our goal was to identify phosphorylation sites that regulate serum response factor (SRF) activity to gain a better understanding of the signaling mechanisms that regulate SRF's involvement in smooth muscle cell (SMC)-specific and early response gene expression. METHODS AND RESULTS By screening phosphorylation-deficient and mimetic mutations in SRF(-/-) embryonic stem cells, we identified T159 as a phosphorylation site that significantly inhibits SMC-specific gene expression in an embryonic stem cell model of SMC differentiation. This residue conforms to a highly conserved consensus cAMP-dependent protein kinase (PKA) site, and in vitro and in vivo labeling studies demonstrated that it was phosphorylated by PKA. Results from gel shift and chromatin immunoprecipitation assays demonstrated that T159 phosphorylation inhibited SRF binding to SMC-specific CArG elements. Interestingly, the myocardin factors could at least partially rescue the effects of the T159D mutation under some conditions, but this response was promoter specific. Finally, PKA signaling had much less of an effect on c-fos promoter activity and SRF binding to the c-fos CArG. CONCLUSIONS Our results indicate that phosphorylation of SRF by PKA inhibits SMC-specific transcription suggesting a novel signaling mechanism for the control of SMC phenotype.
Collapse
Affiliation(s)
- Alicia L Blaker
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | |
Collapse
|
39
|
Sun Q, Taurin S, Sethakorn N, Long X, Imamura M, Wang DZ, Zimmer WE, Dulin NO, Miano JM. Myocardin-dependent activation of the CArG box-rich smooth muscle gamma-actin gene: preferential utilization of a single CArG element through functional association with the NKX3.1 homeodomain protein. J Biol Chem 2009; 284:32582-90. [PMID: 19797053 DOI: 10.1074/jbc.m109.033910] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Serum response factor (SRF) is a ubiquitously expressed transcription factor that binds a 10-bp element known as the CArG box, located in the proximal regulatory region of hundreds of target genes. SRF activates target genes in a cell- and context-dependent manner by assembling unique combinations of cofactors over CArG elements. One particularly strong SRF cofactor, myocardin (MYOCD), acts as a component of a molecular switch for smooth muscle cell (SMC) differentiation by activating cytoskeletal and contractile genes harboring SRF-binding CArG elements. Here we report that the human ACTG2 promoter, containing four conserved CArG elements, displays SMC-specific basal activity and is highly induced in the presence of MYOCD. Stable transfection of a non-SMC cell type with Myocd elicits elevations in endogenous Actg2 mRNA. Gel shift and luciferase assays reveal a strong bias for MYOCD-dependent transactivation through CArG2 of the human ACTG2 promoter. Substitution of CArG2 with other CArGs, including a consensus CArG element, fails to reconstitute full MYOCD-dependent ACTG2 promoter stimulation. Mutation of an adjacent binding site for NKX3.1 reduces MYOCD-dependent transactivation of the ACTG2 promoter. Co-immunoprecipitation, glutathione S-transferase pulldown, and luciferase assays show a physical and functional association between MYOCD and NKX3.1; no such functional relationship is evident with the related NKX2.5 transcription factor despite its interaction with MYOCD. These results demonstrate the ability of MYOCD to discriminate among several juxtaposed CArG elements, presumably through its novel partnership with NKX3.1, to optimally transactivate the human ACTG2 promoter.
Collapse
Affiliation(s)
- Qiang Sun
- Aab Cardiovascular Research Institute of the University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
The role of lysophosphatidic acid receptors in phenotypic modulation of vascular smooth muscle cells. Mol Biol Rep 2009; 37:2675-86. [DOI: 10.1007/s11033-009-9798-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Accepted: 09/02/2009] [Indexed: 12/29/2022]
|
41
|
Munoz-Pinto DJ, Bulick AS, Hahn MS. Uncoupled investigation of scaffold modulus and mesh size on smooth muscle cell behavior. J Biomed Mater Res A 2009; 90:303-16. [PMID: 19402139 DOI: 10.1002/jbm.a.32492] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although scaffold material properties are known to critically impact cell behavior, it has proven difficult to correlate specific cell responses to isolated scaffold parameters, inhibiting rational design of scaffold material properties. The aim of this study was to validate a systematic approach for evaluating the influence of initial scaffold modulus and mesh size on cell extracellular matrix (ECM) deposition and phenotype. Poly(ethylene glycol) diacrylate (PEGDA) hydrogels were selected for this study because of their tunable material properties. Following screening of six distinct PEGDA hydrogels, three formulations were identified which permitted uncoupled investigation of scaffold mesh size and modulus within the target incremental modulus range of approximately 100-300 kPa. Smooth muscle cells (SMCs) were encapsulated within these three formulations, and cell ECM deposition and phenotype were evaluated following 21 days of culture. Although elastin content appeared to be correlated with scaffold mesh size and modulus to a similar degree, levels of collagen and serum response factor (SRF), a key regulator of SMC phenotype, were more strongly correlated with mesh size. To gain insight into the cell signaling underlying these observed correlations, variations in cell metabolic state and in RhoA signaling were semi-quantitatively evaluated. Both RhoA activity, which is largely modulated by scaffold mechanics in 2D, and cell metabolic activity were highly correlated with hydrogel mesh size. These results indicate that the effects of scaffold mechanics on RhoA activity in 3D may be distinct from those in 2D and underscore the need for uncoupled investigation of scaffold parameters on cell behavior. Furthermore, the present data suggest that RhoA signaling and cell metabolic regulation may be closely linked.
Collapse
Affiliation(s)
- Dany J Munoz-Pinto
- Department of Chemical Engineering, Texas A&M University, College Station, Texas, USA
| | | | | |
Collapse
|
42
|
Bulick AS, Muñoz-Pinto DJ, Qu X, Mani M, Cristancho D, Urban M, Hahn MS. Impact of endothelial cells and mechanical conditioning on smooth muscle cell extracellular matrix production and differentiation. Tissue Eng Part A 2009; 15:815-25. [PMID: 19108675 DOI: 10.1089/ten.tea.2008.0179] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The aim of the current study was to explore the separate and coupled effects of endothelial cell (EC) presence and mechanical conditioning on smooth muscle cell (SMC) responses by combining bilayered poly(ethylene glycol) diacrylate (PEGDA) hydrogels with a pulsatile flow bioreactor. Each construct was composed of an outer PEGDA layer containing SMC and an inner PEGDA layer, either with or without EC. After an initial 3 days of static culture, EC(+) and EC(-) constructs were each further divided into two subgroups, half of which received mechanical conditioning mimetic of late gestation (mean pressures of approximately 50 mmHg and peak-to-trough pressure differentials of approximately 20 mmHg at approximately 140-180 beats/min) and half of which were cultured statically. After 18 additional days of culture, the SMC-containing layer of each construct was harvested, and western blots and quantitative histology were conducted to compare collagen type I, collagen type III, and elastin levels among treatment groups. SMC differentiation was evaluated by focusing on SMC marker calponin h1 and direct regulators of its gene expression-the transcription factor serum response factor (SRF) and two of its binding partners, myocardin and Elk-1. Combined EC and pulsatile flow conditioning increased elastin production, but decreased collagen type I deposition. Further, combined EC presence and mechanical stimulation increased SRF levels and the ratio of myocardin to active, phosphorylated Elk-1. This modulation of SRF and its binding partners appeared to result in a net increase in SMC differentiation, as evidenced by an associated increase in calponin h1 levels.
Collapse
Affiliation(s)
- Allen S Bulick
- Department of Chemical Engineering, Texas A&M University , College Station, Texas
| | | | | | | | | | | | | |
Collapse
|
43
|
Neuman NA, Ma S, Schnitzler GR, Zhu Y, Lagna G, Hata A. The four-and-a-half LIM domain protein 2 regulates vascular smooth muscle phenotype and vascular tone. J Biol Chem 2009; 284:13202-12. [PMID: 19265191 PMCID: PMC2676052 DOI: 10.1074/jbc.m900282200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 02/23/2009] [Indexed: 01/14/2023] Open
Abstract
In response to vascular injury, differentiated vascular smooth muscle cells (vSMCs) undergo a unique process known as "phenotype modulation," transitioning from a quiescent, "contractile" phenotype to a proliferative, "synthetic" state. We have demonstrated previously that the signaling pathway of bone morphogenetic proteins, members of the transforming growth factor beta family, play a role in the induction and maintenance of a contractile phenotype in human primary pulmonary artery smooth muscle cells. In this study, we show that a four-and-a-half LIM domain protein 2 (FHL2) inhibits transcriptional activation of vSMC-specific genes mediated by the bone morphogenetic protein signaling pathway through the CArG box-binding proteins, such as serum response factor and members of the myocardin (Myocd) family. Interestingly, FHL2 does not affect recruitment of serum response factor or Myocd, however, it inhibits recruitment of a component of the SWI/SNF chromatin remodeling complex, Brg1, and RNA polymerase II, which are essential for the transcriptional activation. This is a novel mechanism of regulation of SMC-specific contractile genes by FHL2. Finally, aortic rings from homozygous FHL2-null mice display abnormalities in both endothelial-dependent and -independent relaxation, suggesting that FHL2 is essential for the regulation of vasomotor tone.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Aorta, Thoracic/cytology
- Aorta, Thoracic/metabolism
- Blotting, Western
- Bone Morphogenetic Protein Receptors, Type II/genetics
- Bone Morphogenetic Protein Receptors, Type II/metabolism
- COS Cells
- Cells, Cultured
- Chlorocebus aethiops
- Chromatin Assembly and Disassembly
- Chromatin Immunoprecipitation
- DNA Helicases/genetics
- DNA Helicases/metabolism
- Fluorescent Antibody Technique
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Homeodomain Proteins/physiology
- Humans
- LIM-Homeodomain Proteins
- Mice
- Mice, Knockout
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle Proteins/physiology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Promoter Regions, Genetic
- Pulmonary Artery/cytology
- Pulmonary Artery/metabolism
- RNA Polymerase II/genetics
- RNA Polymerase II/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serum Response Factor/genetics
- Serum Response Factor/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription Factors/physiology
- Transcriptional Activation
Collapse
Affiliation(s)
- Nicole A Neuman
- Department of Biochemistry, Tufts University School of Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
44
|
Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A. Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem 2009; 284:3728-38. [PMID: 19088079 PMCID: PMC2635044 DOI: 10.1074/jbc.m808788200] [Citation(s) in RCA: 259] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 12/15/2008] [Indexed: 01/13/2023] Open
Abstract
The platelet-derived growth factor (PDGF) signaling pathway is a critical regulator of animal development and homeostasis. Activation of the PDGF pathway leads to neointimal proliferative responses to artery injury; it promotes a switch of vascular smooth muscle cells (vSMC) to a less contractile phenotype by inhibiting the SMC-specific gene expression and increasing the rate of proliferation and migration. The molecular mechanism for these pleiotropic effects of PDGFs has not been fully described. Here, we identify the microRNA-221 (miR-221), a small noncoding RNA, as a modulator of the phenotypic change of vSMCs in response to PDGF signaling. We demonstrate that miR-221 is transcriptionally induced upon PDGF treatment in primary vSMCs, leading to down-regulation of the targets c-Kit and p27Kip1. Down-regulation of p27Kip1 by miR-221 is critical for PDGF-mediated induction of cell proliferation. Additionally, decreased c-Kit causes inhibition of SMC-specific contractile gene transcription by reducing the expression of Myocardin (Myocd), a potent SMC-specific nuclear coactivator. Our study demonstrates that PDGF signaling, by modulating the expression of miR-221, regulates two critical determinants of the vSMC phenotype; they are SMC gene expression and cell proliferation.
Collapse
Affiliation(s)
- Brandi N Davis
- Department of Biochemistry, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
45
|
Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A. Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem 2008. [PMID: 19088079 DOI: 10.1074/jbc.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The platelet-derived growth factor (PDGF) signaling pathway is a critical regulator of animal development and homeostasis. Activation of the PDGF pathway leads to neointimal proliferative responses to artery injury; it promotes a switch of vascular smooth muscle cells (vSMC) to a less contractile phenotype by inhibiting the SMC-specific gene expression and increasing the rate of proliferation and migration. The molecular mechanism for these pleiotropic effects of PDGFs has not been fully described. Here, we identify the microRNA-221 (miR-221), a small noncoding RNA, as a modulator of the phenotypic change of vSMCs in response to PDGF signaling. We demonstrate that miR-221 is transcriptionally induced upon PDGF treatment in primary vSMCs, leading to down-regulation of the targets c-Kit and p27Kip1. Down-regulation of p27Kip1 by miR-221 is critical for PDGF-mediated induction of cell proliferation. Additionally, decreased c-Kit causes inhibition of SMC-specific contractile gene transcription by reducing the expression of Myocardin (Myocd), a potent SMC-specific nuclear coactivator. Our study demonstrates that PDGF signaling, by modulating the expression of miR-221, regulates two critical determinants of the vSMC phenotype; they are SMC gene expression and cell proliferation.
Collapse
Affiliation(s)
- Brandi N Davis
- Department of Biochemistry, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
46
|
Kawai-Kowase K, Ohshima T, Matsui H, Tanaka T, Shimizu T, Iso T, Arai M, Owens GK, Kurabayashi M. PIAS1 mediates TGFbeta-induced SM alpha-actin gene expression through inhibition of KLF4 function-expression by protein sumoylation. Arterioscler Thromb Vasc Biol 2008; 29:99-106. [PMID: 18927467 DOI: 10.1161/atvbaha.108.172700] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE TGFbeta and proliferation/phenotypic switching of smooth muscle cells (SMCs) play a pivotal role in pathogenesis of atherosclerotic and restenotic lesions after angioplasty. We have previously shown that the protein inhibitor of activated STAT (PIAS)1 activates expression of SMC differentiation marker genes including smooth muscle (SM) alpha-actin by interacting with serum response factor (SRF) and class I bHLH proteins. Here, we tested the hypothesis that TGFbeta activates SM alpha-actin through PIAS1. METHODS AND RESULTS An siRNA specific for PIAS1 and ubc9, an E2-ligase for sumoylation, inhibited TGFbeta-induced expression of SM alpha-actin in cultured SMCs as determined by real-time RT-PCR. Overexpression of PIAS1 increased SM alpha-actin promoter activity in a TGFbeta control element (TCE)-dependent manner. Because the TCE within the SM alpha-actin promoter could mediate repression through interaction with KLF4, we tested whether PIAS1 regulates the function of KLF4 for SMC gene expression. PIAS1 interacted with KLF4 in mammalian two-hybrid and coimmunoprecipitation assays, and overexpression of PIAS1 inhibited KLF4-repression of SM alpha-actin promoter activity. Moreover, PIAS1 promoted degradation of KLF4 through sumoylation. CONCLUSIONS These results provide evidence that PIAS1 promotes TGFbeta-induced activation of SM alpha-actin gene expression at least in part by promoting sumoylation and degradation of the TCE repressor protein, KLF4.
Collapse
Affiliation(s)
- Keiko Kawai-Kowase
- Department of Medicine and Biological Science, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lockman K, Taylor JM, Mack CP. The histone demethylase, Jmjd1a, interacts with the myocardin factors to regulate SMC differentiation marker gene expression. Circ Res 2007; 101:e115-23. [PMID: 17991879 DOI: 10.1161/circresaha.107.164178] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We and others have previously shown that the myocardin transcription factors play critical roles in the regulation of smooth muscle cell (SMC) differentiation marker gene expression. In a yeast 2-hybrid screen for proteins that interact with myocardin-related transcription factor-A (MRTF-A), we identified the histone 3 lysine 9 (H3K9)-specific demethylase, Jmjd1a. GST pull-down assays demonstrated that Jmjd1a bound all 3 myocardin family members, and further mapping studies showed that the jumonjiC domain of Jmjd1a was sufficient to mediate this interaction. Overexpression of Jmjd1a in multipotential 10T1/2 cells decreased global levels of di-methyl H3K9, stimulated the SM alpha-actin and SM22 promoters, and synergistically enhanced MRTF-A- and myocardin-dependent transactivation. Using chromatin immunoprecipitation assays, we also demonstrated that TGF-beta-mediated upregulation of SMC differentiation marker gene expression in 10T1/2 cells was associated with decreased H3K9 dimethylation at the CArG-containing regions of the SMC differentiation marker gene promoters. Importantly, knockdown of Jmjd1a in 10T1/2 cells and primary rat aortic SMCs by retroviral delivery of siRNA attenuated TGF-beta-induced upregulation of endogenous SM myosin heavy chain expression. These effects were concomitant with increased H3K9 dimethylation at the SMC differentiation marker gene promoters and with inhibition of MRTF-A-dependent transactivation of the SMC-specific transcription. These results suggest, for the first time, that SMC differentiation marker gene expression is regulated by H3K9 methylation and that the effects of the myocardin factors on SMC-specific transcription may involve the recruitment of Jmjd1a to the SMC-specific promoters.
Collapse
Affiliation(s)
- Kashelle Lockman
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA
| | | | | |
Collapse
|
48
|
Lagna G, Ku MM, Nguyen PH, Neuman NA, Davis BN, Hata A. Control of phenotypic plasticity of smooth muscle cells by bone morphogenetic protein signaling through the myocardin-related transcription factors. J Biol Chem 2007; 282:37244-55. [PMID: 17947237 DOI: 10.1074/jbc.m708137200] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs), unlike other muscle cells, do not terminally differentiate. In response to injury, VSMCs change phenotype, proliferate, and migrate as part of the repair process. Dysregulation of this plasticity program contributes to the pathogenesis of several vascular disorders, such as atherosclerosis, restenosis, and hypertension. The discovery of mutations in the gene encoding BMPRII, the type II subunit of the receptor for bone morphogenetic proteins (BMPs), in patients with pulmonary arterial hypertension (PAH) provided an indication that BMP signaling may affect the homeostasis of VSMCs and their phenotype modulation. Here we report that BMP signaling potently induces SMC-specific genes in pluripotent cells and prevents dedifferentiation of arterial SMCs. The BMP-induced phenotype switch requires intact RhoA/ROCK signaling but is not blocked by inhibitors of the TGFbeta and PI3K/Akt pathways. Furthermore, nuclear localization and recruitment of the myocardin-related transcription factors (MRTF-A and MRTF-B) to a smooth muscle alpha-actin promoter is observed in response to BMP treatment. Thus, BMP signaling modulates VSMC phenotype via cross-talk with the RhoA/MRTFs pathway, and may contribute to the development of the pathological characteristics observed in patients with PAH and other obliterative vascular diseases.
Collapse
Affiliation(s)
- Giorgio Lagna
- Molecular Cardiology Research Institute, Tufts-New England Medical Center, Boston, MA 02111, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Hinson JS, Medlin MD, Lockman K, Taylor JM, Mack CP. Smooth muscle cell-specific transcription is regulated by nuclear localization of the myocardin-related transcription factors. Am J Physiol Heart Circ Physiol 2007; 292:H1170-80. [PMID: 16997888 DOI: 10.1152/ajpheart.00864.2006] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
On the basis of our previous studies on RhoA signaling in smooth muscle cells (SMC), we hypothesized that RhoA-mediated nuclear translocalization of the myocardin-related transcription factors (MRTFs) was important for regulating SMC phenotype. MRTF-A protein and MRTF-B message were detected in aortic SMC and in many adult mouse organs that contain a large SMC component. Both MRTFs upregulated SMC-specific promoter activity as well as endogenous SM22α expression in multipotential 10T1/2 cells, although to a lesser extent than myocardin. We used enhanced green fluorescent protein (EGFP) fusion proteins to demonstrate that the myocardin factors have dramatically different localization patterns and that the stimulation of SMC-specific transcription by certain RhoA-dependent agonists was likely mediated by increased nuclear translocation of the MRTFs. Importantly, a dominant-negative form of MRTF-A (ΔB1/B2) that traps endogenous MRTFs in the cytoplasm inhibited the SM α-actin, SM22α, and SM myosin heavy chain promoters in SMC and attenuated the effects of sphingosine 1-phosphate and transforming growth factor (TGF)-β on SMC-specific transcription. Our data confirmed the importance of the NH2-terminal RPEL domains for regulating MRTF localization, but our analysis of MRTF-A/myocardin chimeras and myocardin RPEL2 mutations indicated that the myocardin B1/B2 region can override this signal. Gel shift assays demonstrated that myocardin factor activity correlated well with ternary complex formation at the SM α-actin CArGs and that MRTF-serum response factor interactions were partially dependent on CArG sequence. Taken together, our results indicate that the MRTFs regulate SMC-specific gene expression in at least some SMC subtypes and that regulation of MRTF nuclear localization may be important for the effects of selected agonists on SMC phenotype.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Aorta, Thoracic/metabolism
- Cell Differentiation
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cells, Cultured
- Lysophospholipids/pharmacology
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Mutation
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Phenotype
- Platelet-Derived Growth Factor/pharmacology
- Promoter Regions, Genetic/drug effects
- RNA, Messenger/metabolism
- Rats
- Serum Response Factor/metabolism
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- Time Factors
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic/drug effects
- Transfection
- Transforming Growth Factor beta/pharmacology
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Jeremiah S Hinson
- Department of Pathology and Laboratory Medicine and the Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|
50
|
Mondin M, Moreau V, Genot E, Combe C, Ripoche J, Dubus I. Alterations in cytoskeletal protein expression by mycophenolic acid in human mesangial cells requires Rac inactivation. Biochem Pharmacol 2006; 73:1491-8. [PMID: 17258688 DOI: 10.1016/j.bcp.2006.12.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 12/20/2006] [Accepted: 12/20/2006] [Indexed: 11/26/2022]
Abstract
In response to glomerular injury, mesangial cells are activated into myofibroblasts, which contribute to the physiopathology of glomerulosclerosis. We have previously shown that chronic treatment of cultured human mesangial cells with mycophenolic acid (MPA), a specific inhibitor of guanosine nucleotide synthesis, prevents their activation and alters cytoskeleton protein expression and associated functions, such as contractility and migratory capacity. The aim of the present study was to explore the mechanisms underlying MPA-induced mesangial cytoskeleton alterations. We therein show that coincubation with guanosine (100 microM) compensates for the effects of MPA on mesangial cell proliferation and migration, and prevents MPA-induced overexpression of alpha-smooth muscle actin (SMA) and basic calponin (b-calp), indicating that guanylates are involved in mesangial responses to MPA. MPA decreased the GTP-bound (active) form of both RhoA, Rac1 and Cdc42, and specifically altered the expression level of Rac1. Pharmacological inhibition of RhoA activity reduced expression of both SMA and calponin, whereas overexpression of a dominant-negative form of Rac1 increased SMA expression. Conversely, overexpression of constitutively active Rac1 resulted in SMA and b-calp down-regulation, and fully prevented their stimulation by MPA, indicating that Rac inactivation is responsible for MPA effects on mesangial cytoskeletal expression. These results show that in human mesangial cells, RhoA and Rac1 exert opposite effects on the expression of two major cytoskeletal proteins: SMA and basic calponin. Moreover, these data highlight for the first time an integrated mechanism whereby MPA regulates mesangial phenotype, which is mediated by loss of Rac activity.
Collapse
|