1
|
Wang Z, Qiao XH, Xu YJ, Liu XY, Huang RT, Xue S, Qiu HY, Yang YQ. SMAD1 Loss-of-Function Variant Responsible for Congenital Heart Disease. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9916325. [PMID: 35281600 PMCID: PMC8913148 DOI: 10.1155/2022/9916325] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/16/2022] [Indexed: 12/22/2022]
Abstract
As the most common form of developmental malformation affecting the heart and endothoracic great vessels, congenital heart disease (CHD) confers substantial morbidity and mortality as well as socioeconomic burden on humans globally. Aggregating convincing evidence highlights the genetic origin of CHD, and damaging variations in over 100 genes have been implicated with CHD. Nevertheless, the genetic basis underpinning CHD remains largely elusive. In this study, via whole-exosome sequencing analysis of a four-generation family inflicted with autosomal-dominant CHD, a heterozygous SMAD1 variation, NM_005900.3: c.264C > A; p.(Tyr88∗), was detected and validated by Sanger sequencing analysis to be in cosegregation with CHD in the whole family. The truncating variation was not observed in 362 unrelated healthy volunteers employed as control persons. Dual-luciferase reporter gene assay in cultured COS7 cells demonstrated that Tyr88∗-mutant SMAD1 failed to transactivate the genes TBX20 and NKX2.5, two already well-established CHD-causative genes. Additionally, the variation nullified the synergistic transcriptional activation between SMAD1 and MYOCD, another recognized CHD-causative gene. These data indicate SMAD1 as a new gene responsible for CHD, which provides new insight into the genetic mechanism underlying CHD, suggesting certain significance for genetic risk assessment and precise antenatal prevention of the family members inflicted with CHD.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hai-Yan Qiu
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
2
|
Zheng M, Erhardt S, Ai D, Wang J. Bmp Signaling Regulates Hand1 in a Dose-Dependent Manner during Heart Development. Int J Mol Sci 2021; 22:ijms22189835. [PMID: 34576009 PMCID: PMC8465227 DOI: 10.3390/ijms22189835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 01/22/2023] Open
Abstract
The bone morphogenetic protein (Bmp) signaling pathway and the basic helix–loop–helix (bHLH) transcription factor Hand1 are known key regulators of cardiac development. In this study, we investigated the Bmp signaling regulation of Hand1 during cardiac outflow tract (OFT) development. In Bmp2 and Bmp4loss-of-function embryos with varying levels of Bmp in the heart, Hand1 is sensitively decreased in response to the dose of Bmp expression. In contrast, Hand1 in the heart is dramatically increased in Bmp4 gain-of-function embryos. We further identified and characterized the Bmp/Smad regulatory elements in Hand1. Combined transfection assays and chromatin immunoprecipitation (ChIP) experiments indicated that Hand1 is directly activated and bound by Smads. In addition, we found that upon the treatment of Bmp2 and Bmp4, P19 cells induced Hand1 expression and favored cardiac differentiation. Together, our data indicated that the Bmp signaling pathway directly regulates Hand1 expression in a dose-dependent manner during heart development.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Di Ai
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (M.Z.); (S.E.)
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
3
|
Zhou Q, Chen W, Fan Z, Chen Z, Liang J, Zeng G, Liu L, Liu W, Yang T, Cao X, Yu B, Xu M, Chen YG, Chen L. Targeting hyperactive TGFBR2 for treating MYOCD deficient lung cancer. Theranostics 2021; 11:6592-6606. [PMID: 33995678 PMCID: PMC8120205 DOI: 10.7150/thno.59816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
Purpose: Clinical success of cancer therapy is severely limited by drug resistance, attributed in large part to the loss of function of tumor suppressor genes (TSGs). Developing effective strategies to treat those tumors is challenging, but urgently needed in clinic. Experimental Design: MYOCD is a clinically relevant TSG in lung cancer patients. Our in vitro and in vivo data confirm its tumor suppressive function. Further analysis reveals that MYOCD potently inhibits stemness of lung cancer stem cells. Mechanistically, MYOCD localizes to TGFBR2 promoter region and thereby recruits PRMT5/MEP50 complex to epigenetically silence its transcription. Conclusions: NSCLC cells deficient of MYOCD are particularly sensitive to TGFBR kinase inhibitor (TGFBRi). TGFBRi and stemness inhibitor synergize with existing drugs to treat MYOCD deficient lung cancers. Our current work shows that loss of function of MYOCD creates Achilles' heels in lung cancer cells, which might be exploited in clinic.
Collapse
|
4
|
Bhuvanalakshmi G, Arfuso F, Kumar AP, Dharmarajan A, Warrier S. Epigenetic reprogramming converts human Wharton's jelly mesenchymal stem cells into functional cardiomyocytes by differential regulation of Wnt mediators. Stem Cell Res Ther 2017; 8:185. [PMID: 28807014 PMCID: PMC5557557 DOI: 10.1186/s13287-017-0638-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/14/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022] Open
Abstract
Background Lineage commitment of mesenchymal stem cells (MSCs) to cardiac differentiation is controlled by transcription factors that are regulated by epigenetic events, mainly histone deacetylation and promoter DNA methylation. Here, we studied the differentiation of human Wharton’s jelly MSCs (WJMSCs) into the cardiomyocyte lineage via epigenetic manipulations. Methods We introduced these changes using inhibitors of DNA methyl transferase and histone deacetylase, DC301, DC302, and DC303, in various combinations. We characterized for cardiogenic differentiation by assessing the expression of cardiac-specific markers by immunolocalization, quantitative RT-PCR, and flow cytometry. Cardiac functional studies were performed by FURA2AM staining and Greiss assay. The role of Wnt signaling during cardiac differentiation was analyzed by quantitative RT-PCR. In-vivo studies were performed in a doxorubicin-induced cardiotoxic mouse model by injecting cardiac progenitor cells. Promoter methylation status of the cardiac transcription factor Nkx2.5 and the Wnt antagonist, secreted frizzled-related protein 4 (sFRP4), after cardiac differentiation was studied by bisulfite sequencing. Results By induction with DC301 and DC302, WJMSCs differentiated into cardiomyocyte-like structures with an upregulation of Wnt antagonists, sFRP3 and sFRP4, and Dickkopf (Dkk)1 and Dkk3. The cardiac function enhancer, vinculin, and DDX20, a DEAD-box RNA helicase, were also upregulated in differentiated cardiomyocytes. Additionally, bisulfite sequencing revealed, for the first time in cardiogenesis, that sFRP4 is activated by promoter CpG island demethylation. In vivo, these MSC-derived cardiac progenitors could not only successfully engraft to the site of cardiac injury in mice with doxorubicin-induced cardiac injury, but also form functional cardiomyocytes and restore cardiac function. Conclusion The present study unveils a link between Wnt inhibition and epigenetic modification to initiate cardiac differentiation, which could enhance the efficacy of stem cell therapy for ischemic heart disorders. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0638-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- G Bhuvanalakshmi
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal University, Bangalore, 560 065, India
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6845, Australia.,School of Anatomy, Physiology and Human Biology, Faculty of Science, The University of Western Australia, 35 Stirling Highway, Crawley, WA, 6009, Australia
| | - Alan Prem Kumar
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal University, Bangalore, 560 065, India.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia.,National University Cancer Institute, Singapore, 119074, Singapore.,Department of Biological Sciences, University of North Texas, Denton, TX, 76203-5017, USA
| | - Arun Dharmarajan
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6845, Australia
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal University, Bangalore, 560 065, India. .,School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6102, Australia. .,Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, WA, 6875, Australia.
| |
Collapse
|
5
|
|
6
|
Xia XD, Zhou Z, Yu XH, Zheng XL, Tang CK. Myocardin: A novel player in atherosclerosis. Atherosclerosis 2017; 257:266-278. [DOI: 10.1016/j.atherosclerosis.2016.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022]
|
7
|
Zhu S, Hu X, Yu Z, Peng Y, Zhu J, Liu X, Li M, Han S, Zhu C. Effect of miR-20b on Apoptosis, Differentiation, the BMP Signaling Pathway and Mitochondrial Function in the P19 Cell Model of Cardiac Differentiation In Vitro. PLoS One 2015; 10:e0123519. [PMID: 25898012 PMCID: PMC4405592 DOI: 10.1371/journal.pone.0123519] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 02/19/2015] [Indexed: 12/29/2022] Open
Abstract
Objective To explore the effect of miR-20b on apoptosis, differentiation, the BMP signaling pathway and mitochondrial function in the P19 cell model of cardiac differentiation in vitro. Methods A miR-20b over-expression vector, a miR-20b silencing vector and their corresponding empty vectors were constructed and transfected into P19 cells, separately. Stably miR-20b overexpressing and silenced P19 cell lines were successfully selected by blasticidin and puromycin, separately. The cells were induced to undergo apoptosis in FBS-free-α-MEM. The induced cells were examined by flow cytometry and measurement of their caspase-3 activities. Quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) was used to evaluate the relative expression of marker genes of cardiomyocytes during differentiation, such as cTnT, GATA4 and ANP. QRT-PCR was also used to detect the mitochondrial DNA (mtDNA) copy number. We investigated the cellular ATP production using a luciferase-based luminescence assay. The reactive oxygen species (ROS) was determined by DCFDA (2’, 7’-Dichlorofluorescein diacetate) and the mitochondrial membrane potential (MMP) was elucidated by a JC-1 fluorescent probe, both using fluorescence microscopy and flow cytometer. The expression of BMP signaling pathway-related proteins were analyzed by Western blotting. Results Stably miR-20b overexpressing and silenced P19 cell lines were successfully obtained. MiR-20b overexpression increased apoptosis and promoted differentiation in P19 cells by promoting the activation of the BMP signaling pathway. In addition, miR-20b overexpression induced mitochondrial impairment in P19 cells during differentiation, which was characterized by lower MMP, raised ATP synthesis and increased ROS levels. The effects of miR-20b silencing were the exact opposite to those of overexpression. Conclusion Collectively, these results suggested that miR-20b was very important in apoptosis, differentiation and mitochondrial function of P19 cells. MiR-20b may represent a new therapeutic target for congenital heart diseases and provide new insights into the mechanisms of cardiac diseases.
Collapse
Affiliation(s)
- Shasha Zhu
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiaoshan Hu
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Zhangbin Yu
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Yuzhu Peng
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Jingai Zhu
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Xuehua Liu
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Mengmeng Li
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Shuping Han
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
- * E-mail: (SPH); (CZ)
| | - Chun Zhu
- State key Laboratory of Reproductive Medicine, Department of Pediatrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
- * E-mail: (SPH); (CZ)
| |
Collapse
|
8
|
Rahman MS, Akhtar N, Jamil HM, Banik RS, Asaduzzaman SM. TGF-β/BMP signaling and other molecular events: regulation of osteoblastogenesis and bone formation. Bone Res 2015; 3:15005. [PMID: 26273537 PMCID: PMC4472151 DOI: 10.1038/boneres.2015.5] [Citation(s) in RCA: 398] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/26/2015] [Accepted: 02/27/2015] [Indexed: 02/08/2023] Open
Abstract
Transforming growth factor-beta (TGF-β)/bone morphogenetic protein (BMP) plays a fundamental role in the regulation of bone organogenesis through the activation of receptor serine/threonine kinases. Perturbations of TGF-β/BMP activity are almost invariably linked to a wide variety of clinical outcomes, i.e., skeletal, extra skeletal anomalies, autoimmune, cancer, and cardiovascular diseases. Phosphorylation of TGF-β (I/II) or BMP receptors activates intracellular downstream Smads, the transducer of TGF-β/BMP signals. This signaling is modulated by various factors and pathways, including transcription factor Runx2. The signaling network in skeletal development and bone formation is overwhelmingly complex and highly time and space specific. Additive, positive, negative, or synergistic effects are observed when TGF-β/BMP interacts with the pathways of MAPK, Wnt, Hedgehog (Hh), Notch, Akt/mTOR, and miRNA to regulate the effects of BMP-induced signaling in bone dynamics. Accumulating evidence indicates that Runx2 is the key integrator, whereas Hh is a possible modulator, miRNAs are regulators, and β-catenin is a mediator/regulator within the extensive intracellular network. This review focuses on the activation of BMP signaling and interaction with other regulatory components and pathways highlighting the molecular mechanisms regarding TGF-β/BMP function and regulation that could allow understanding the complexity of bone tissue dynamics.
Collapse
Affiliation(s)
- Md Shaifur Rahman
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Naznin Akhtar
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Hossen Mohammad Jamil
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| | - Rajat Suvra Banik
- Lab of Network Biology, Biotechnology and Genetic Engineering Discipline, Khulna University , Khulna 9208, Bangladesh
| | - Sikder M Asaduzzaman
- Tissue Banking and Biomaterial Research Unit, Atomic Energy Research Establishment , Dhaka 1349, Bangladesh
| |
Collapse
|
9
|
Yang B, He K, Zheng F, Wan L, Yu X, Wang X, Zhao D, Bai Y, Chu W, Sun Y, Lu Y. Over-expression of hypoxia-inducible factor-1 alpha in vitro protects the cardiac fibroblasts from hypoxia-induced apoptosis. J Cardiovasc Med (Hagerstown) 2015; 15:579-86. [PMID: 24583668 DOI: 10.2459/jcm.0b013e3283629c52] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES A great number of studies indicate that cardiac fibroblasts are essential for maintaining the structure and function of heart. Hypoxia-inducible factor-1 alpha (HIF-1α) is a central transcriptional regulator of hypoxic response. The present study examined whether over-expression of HIF-1α could prevent hypoxia-induced injury in neonatal rat cardiac fibroblasts and, if so, its possible molecular targets. METHODS Western blotting was used to detect protein level. MTT, electron microscopy, TUNEL staining and confocal microscopy were used to identify cell viability, cell apoptosis and intracellular calcium ([Ca]i) in cardiac fibroblasts, respectively. RESULTS When cardiac fibroblasts were exposed to hypoxia, HIF-1α protein in nuclei was transiently accumulated at 1 h, and then gradually degraded within 24 h of hypoxia exposure. Over-expression of HIF-1α enhanced nucleus expression of HIF-1α in cardiac fibroblasts, and significantly abolished the decrease of cell viability and cell apoptosis caused by 24-h hypoxia. Accordingly, hypoxia-induced Bax up-regulation, Bcl-2 down-regulation, caspase-3 activation and overload of [Ca]i in cardiac fibroblasts were reversed by HIF-1α over-expression, but were promoted by 30 μmol/l SC205346, a specific HIF-1α blocker. CONCLUSIONS Our results indicate that HIF-1α may act as a protective factor in the apoptotic process of cardiac fibroblasts and represent a potential therapeutic target for heart remodeling after hypoxia injury.
Collapse
Affiliation(s)
- Baofeng Yang
- aDepartment of Pharmacy, the Daqing Oilfield General Hospital, Daqing, Heilongjiang bDepartment of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education) cDepartment of Surgery, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, P.R. China *These authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Tokola H, Rysä J, Pikkarainen S, Hautala N, Leskinen H, Kerkelä R, Ilves M, Aro J, Vuolteenaho O, Ritvos O, Ruskoaho H. Bone morphogenetic protein-2--a potential autocrine/paracrine factor in mediating the stretch activated B-type and atrial natriuretic peptide expression in cardiac myocytes. Mol Cell Endocrinol 2015; 399:9-21. [PMID: 25218476 DOI: 10.1016/j.mce.2014.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Revised: 08/25/2014] [Accepted: 09/03/2014] [Indexed: 12/31/2022]
Abstract
Hemodynamic overload exposes the heart to variety of neural, humoral and mechanical stresses. Even without the neurohumoral control of the entire organism cardiac myocytes have the ability to sense mechanical stretch and convert it into adaptive intracellular signals. This process is controlled by several growth factors. Here we show that mechanical stretch in vitro and hemodynamic overload in vivo activated the expression of bone morphogenetic protein-2 (BMP-2), while expression of BMP-4 was temporarily attenuated by stretch. BMP-2 and BMP-4 alone stimulated B-type and atrial natriuretic peptide (BNP and ANP) expression and protein synthesis, and activated transcription factor GATA-4 resembling the effects of mechanical stretch of cultured cardiac myocytes. Further, BMP antagonist Noggin was able to inhibit stretch and hypertrophic agonist induced BNP and ANP expression. Together these data provide evidence for BMP-2 as a new autocrine/paracrine factor that regulates cardiomyocyte mechanotransduction and adaptation to increased mechanical stretch.
Collapse
Affiliation(s)
- Heikki Tokola
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Pathology, Institute of Diagnostics, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Pathology, Oulu University Hospital, P.O. BOX 50, Oulu FI-90029 OYS, Finland
| | - Jaana Rysä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio FIN-70211, Finland
| | - Sampsa Pikkarainen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Medicine, Division of Gastroenterology, Helsinki University Central Hospital, P.O. BOX 340, Helsinki FI-00029 HUS, Finland
| | - Nina Hautala
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Department of Ophthalmology, Oulu University Hospital, P.O. BOX 21, Oulu FI-90029 OYS, Finland
| | - Hanna Leskinen
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Risto Kerkelä
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Mika Ilves
- Department of Physiology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Jani Aro
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Olli Vuolteenaho
- Department of Physiology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland
| | - Olli Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki FI-00014, Finland
| | - Heikki Ruskoaho
- Department of Pharmacology and Toxicology, Institute of Biomedicine, University of Oulu, P.O. BOX 5000, Oulu FI-90014, Finland; Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, P.O. BOX 56, Viikinkaari 5E, Helsinki FI-00014, Finland.
| |
Collapse
|
11
|
Zheng XL. Myocardin and smooth muscle differentiation. Arch Biochem Biophys 2014; 543:48-56. [DOI: 10.1016/j.abb.2013.12.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/15/2013] [Accepted: 12/18/2013] [Indexed: 01/08/2023]
|
12
|
Luo XG, Zhang CL, Zhao WW, Liu ZP, Liu L, Mu A, Guo S, Wang N, Zhou H, Zhang TC. Histone methyltransferase SMYD3 promotes MRTF-A-mediated transactivation of MYL9 and migration of MCF-7 breast cancer cells. Cancer Lett 2013; 344:129-137. [PMID: 24189459 DOI: 10.1016/j.canlet.2013.10.026] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/20/2013] [Accepted: 10/24/2013] [Indexed: 01/06/2023]
Abstract
Myocardin-related transcription factor-A (MRTF-A) is a Rho signal-responsive transcriptional coactivator of serum response factor (SRF). Recent studies indicated that MRTF-A might be an important regulator of mammary gland and be involved in cancer metastasis. However, the roles of histone modification in the MRTF-A-dependent signal pathway and tumor migration are still not very clear. Here, we report that histone methylation is required for the MRTF-A-mediated upregulation of myosin regulatory light chain 9 (MYL9), an important cytoskeletal component which is implicated in cell migration. Furthermore, we demonstrate that SET and MYND domain containing protein 3 (SMYD3), a hitone methyltransferase (HMT) associated with carcinogenesis, might be the one which is responsible for the histone methylation occurred in the MRTF-A-mediated- transactivation of MYL9 and migration of breast cancer cells. Overexpression of SMYD3 promotes MRTF-A-mediated upregulation of MYL9 and migration of MCF-7 breast cancer cells, while contrary results were observed when the endogenous MRTF-A and SMYD3 were suppressed with specific siRNAs. In addition, the mutation analysis suggested that this cooperative transactivation is mainly mediated via the proximal binding element of MRTF-A in the promoter of MYL9, and the HMT activity of SMYD3 is required as well. Our findings reveal a new mechanism by which MRTF-A and SMYD3 functions in transcriptional regulation and cell migration, and provide a better understanding for metastasis of breast cancer.
Collapse
Affiliation(s)
- Xue-Gang Luo
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China.
| | - Chun-Ling Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Wen-Wen Zhao
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Zhi-Peng Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Lei Liu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Ai Mu
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Shu Guo
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Nan Wang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Hao Zhou
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China; Tianjin Key Laboratory of Industrial Microbiology, Tianjin 300457, China; School of Medicine, Wuhan University of Science and Technology, Wuhan 430081, China.
| |
Collapse
|
13
|
Treatment with bone morphogenetic protein 2 limits infarct size after myocardial infarction in mice. Shock 2013; 39:353-60. [PMID: 23376954 DOI: 10.1097/shk.0b013e318289728a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Various strategies have been devised to reduce the clinical consequences of myocardial infarction, including acute medical care, revascularization, stem cell transplantations, and more recently, prevention of cardiomyocyte cell death. Activation of embryonic signaling pathways is a particularly interesting option to complement these strategies and to improve the functional performance and survival rate of cardiomyocytes. Here, we have concentrated on bone morphogenetic protein 2 (BMP-2), which induces ectopic formation of beating cardiomyocytes during development in the mesoderm and protects neonatal cardiomyocytes from ischemia-reperfusion injury. In a mouse model of acute myocardial infarction, an i.v. injection of BMP-2 reduced infarct size in mice when given after left anterior descending artery ligation. Mice treated with BMP-2 are characterized by a reduced rate of apoptotic cardiomyocytes both in the border zone of the infarcts and in the remote myocardium. In vitro, BMP-2 increases the frequency of spontaneously beating neonatal cardiomyocytes and the contractile performance under electrical pacing at 2 Hz, preserves cellular adenosine triphosphate stores, and decreases the rate of apoptosis despite the increased workload. In addition, BMP-2 specifically induced phosphorylation of Smad1/5/8 proteins and protected adult cardiomyocytes from long-lasting hypoxia-induced cellular damage and oxidative stress without activation of the cardiodepressant transforming growth factor-β pathway. Our data suggest that BMP-2 treatment may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by improving the contractility of cardiomyocytes and preventing cardiomyocyte cell death.
Collapse
|
14
|
Cao D, Wang C, Tang R, Chen H, Zhang Z, Tatsuguchi M, Wang DZ. Acetylation of myocardin is required for the activation of cardiac and smooth muscle genes. J Biol Chem 2012; 287:38495-504. [PMID: 23007391 DOI: 10.1074/jbc.m112.353649] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myocardin belongs to the SAF-A/B, Acinus, PIAS (SAP) domain family of transcription factors and is specifically expressed in cardiac and smooth muscle. Myocardin functions as a transcriptional coactivator of SRF and is sufficient and necessary for smooth muscle gene expression. We have previously found that myocardin induces the acetylation of nucleosomal histones surrounding SRF-binding sites in the control regions of cardiac and smooth muscle genes through recruiting chromatin-modifying enzyme p300, yet no studies have determined whether myocardin itself is similarly modified. In this study, we show that myocardin is a direct target for p300-mediated acetylation. p300 acetylates lysine residues at the N terminus of the myocardin protein. Interestingly, a direct interaction between p300 and myocardin, which is mediated by the C terminus of myocardin, is required for the acetylation event. Acetylation of myocardin by p300 enhances the association of myocardin and SRF as well as the formation of the myocardin-SRF-CArG box ternary complex. Conversely, acetylation of myocardin decreases the binding of histone deacetylase 5 (HDAC5) to myocardin. Acetylation of myocardin is required for myocardin to activate smooth muscle genes. Our study demonstrates that acetylation plays a key role in modulating myocardin function in controlling cardiac and smooth muscle gene expression.
Collapse
Affiliation(s)
- Dongsun Cao
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Wang Y, Sun A, Xue J, Jiang Y. Adenovirus-mediated expression of hypoxia-inducible factor 1α double mutant converts neonatal cardiac fibroblasts into (cardio)myocyte phenotype. Cell Biochem Funct 2011; 30:24-32. [PMID: 22006794 DOI: 10.1002/cbf.1813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 08/16/2011] [Accepted: 09/05/2011] [Indexed: 02/04/2023]
Abstract
Adenovirus-mediated expression of hypoxia-inducible factor 1α double mutant (pAd-HIF-1α-Ala564-Ala803) can be effectively transfected into bone marrow stem cells (MSCs) in the MSCs and cardiomyocytes co-culture system at normoxia to regulate the expression of downstream target genes of hypoxia-inducible factor 1α (HIF-1α), which in turn can promote MSC differentiation into cardiomyocytes. Fibroblasts share common characteristics with MSCs such as the morphology, phenotype and differentiation potential. Therefore, we further studied whether the pAd-HIF-1α-Ala564-Ala803 also can convert neonatal rat cardiac fibroblasts (NCFs) into (cardio)myocyte phenotype via regulating the downstream target genes of HIF-1α at normoxia. The immunostaining analysis showed that NCFs treated with pAd-HIF-1α-Ala564-Ala803 exhibited higher protein expression levels of smooth muscle α-actin (SMA, myocyte marker) and cardiac troponin T (cTnT, cardiomyocyte marker), compared with phosphate-buffered saline and pAd-LacZ treatments. The reverse transcription-polymerase chain reaction results showed that NCFs transfected with pAd-HIF-1α-Ala564-Ala803 augmented messenger RNA (mRNA) expression of transforming growth factor-β1 (TGF-β1), Smad4, NKx2.5, GATA4, myocardin, SMA and cTnT. The effects of HIF-1α-Ala564-Ala803 on NCFs were attenuated by pre-transfection of TGF-β1 or myocardin small interference RNAs. Adult CFs transfected with pAd-HIF-1α-Ala564-Ala803 showed a lower protein expression of SMA but not cTnT without any change in the mRNA expression level of NKx2.5, myocardin. Therefore, NCFs but not adult CFs possess a similar differentiation potential to MSCs as evidenced by the fact that pAd-HIF-1α-Ala564-Ala803 can convert NCFs into (cardio)myocyte phenotype via regulating its downstream target genes.
Collapse
Affiliation(s)
- Yesong Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.
| | | | | | | |
Collapse
|
16
|
Yook JY, Kim MJ, Son MJ, Lee S, Nam Y, Han YM, Cho YS. Combinatorial Activin Receptor-Like Kinase/Smad and Basic Fibroblast Growth Factor Signals Stimulate the Differentiation of Human Embryonic Stem Cells into the Cardiac Lineage. Stem Cells Dev 2011; 20:1479-90. [DOI: 10.1089/scd.2010.0392] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Jin-Yong Yook
- Development and Differentiation Research Center, KRIBB, Daejeon, Republic of Korea
| | - Min-Jeong Kim
- Development and Differentiation Research Center, KRIBB, Daejeon, Republic of Korea
- University of Science and Technology, Daejeon, Republic of Korea
| | - Myung Jin Son
- Development and Differentiation Research Center, KRIBB, Daejeon, Republic of Korea
| | - Seokyoung Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yoonkey Nam
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yong-Mahn Han
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Yee Sook Cho
- Development and Differentiation Research Center, KRIBB, Daejeon, Republic of Korea
- University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
17
|
Wang C, Cao D, Wang Q, Wang DZ. Synergistic activation of cardiac genes by myocardin and Tbx5. PLoS One 2011; 6:e24242. [PMID: 21897873 PMCID: PMC3163680 DOI: 10.1371/journal.pone.0024242] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/03/2011] [Indexed: 11/18/2022] Open
Abstract
Myocardial differentiation is associated with the activation and expression of an array of cardiac specific genes. However, the transcriptional networks that control cardiac gene expression are not completely understood. Myocardin is a cardiac and smooth muscle-specific expressed transcriptional coactivator of Serum Response Factor (SRF) and is able to potently activate cardiac and smooth muscle gene expression during development. We hypothesize that myocardin discriminates between cardiac and smooth muscle specific genes by associating with distinct co-factors. Here, we show that myocardin directly interacts with Tbx5, a member of the T-box family of transcription factors involved in the Holt-Oram syndrome. Tbx5 synergizes with myocardin to activate expression of the cardiac specific genes atrial natriuretic factor (ANF) and alpha myosin heavy chain (α-MHC), but not that of smooth muscle specific genes SM22 or smooth muscle myosin heavy chain (SM-MHC). We found that this synergistic activation of shared target genes is dependent on the binding sites for Tbx5, T-box factor-Binding Elements (TBEs). Myocardin and Tbx5 physically interact and their interaction domains were mapped to the basic domain and the coil domain of myocardin and Tbx5, respectively. Our analysis demonstrates that the Tbx5G80R mutation, which leads to the Holt-Oram syndrome in humans, failed to synergize with myocardin to activate cardiac gene expression. These data uncover a key role for Tbx5 and myocardin in establishing the transcriptional foundation for cardiac gene activation and suggest that the interaction of myocardin and Tbx5 maybe involved in cardiac development and diseases.
Collapse
Affiliation(s)
- Chunbo Wang
- UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Dongsun Cao
- UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Qing Wang
- Department of Molecular Cardiology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Da-Zhi Wang
- Department of Cardiology, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, United States of America
- UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
18
|
Mack CP. Signaling mechanisms that regulate smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol 2011; 31:1495-505. [PMID: 21677292 DOI: 10.1161/atvbaha.110.221135] [Citation(s) in RCA: 189] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Extensive studies over the last 30 years have demonstrated that vascular smooth muscle cell (SMC) differentiation and phenotypic modulation is controlled by a dynamic array of environmental cues. The identification of the signaling mechanisms by which these environmental cues regulate SMC phenotype has been more difficult because of our incomplete knowledge of the transcription mechanisms that regulate SMC-specific gene expression. However, recent advances in this area have provided significant insight, and the goal of this review is to summarize the signaling mechanisms by which extrinsic cues control SMC differentiation.
Collapse
Affiliation(s)
- Christopher P Mack
- Department of Pathology, University of North Carolina, Chapel Hill, NC 27599-7525, USA.
| |
Collapse
|
19
|
Chu W, Li X, Li C, Wan L, Shi H, Song X, Liu X, Chen X, Zhang C, Shan H, Lu Y, Yang B. TGFBR3, a potential negative regulator of TGF-β signaling, protects cardiac fibroblasts from hypoxia-induced apoptosis. J Cell Physiol 2011; 226:2586-94. [DOI: 10.1002/jcp.22604] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
20
|
Ilagan RM, Genheimer CW, Quinlan SF, Guthrie KI, Sangha N, Ramachandrannair S, Kelley RW, Presnell SC, Basu J, Ludlow JW. Smooth muscle phenotypic diversity is mediated through alterations in Myocardin gene splicing. J Cell Physiol 2011; 226:2702-11. [DOI: 10.1002/jcp.22622] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
21
|
Airik R, Trowe MO, Foik A, Farin HF, Petry M, Schuster-Gossler K, Schweizer M, Scherer G, Kist R, Kispert A. Hydroureternephrosis due to loss of Sox9-regulated smooth muscle cell differentiation of the ureteric mesenchyme. Hum Mol Genet 2010; 19:4918-29. [PMID: 20881014 DOI: 10.1093/hmg/ddq426] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Congenital ureter anomalies, including hydroureter, affect up to 1% of the newborn children. Despite the prevalence of these developmental abnormalities in young children, the underlying molecular causes are only poorly understood. Here, we show that the high mobility group domain transcription factor Sox9 plays an important role in ureter development in the mouse. Transient Sox9 expression was detected in the undifferentiated ureteric mesenchyme and inactivation of Sox9 in this domain resulted in strong proximal hydroureter formation due to functional obstruction. Loss of Sox9 did not affect condensation, proliferation and apoptosis of the undifferentiated mesenchyme, but perturbed cyto-differentiation into smooth muscle cells (SMCs). Expression of genes encoding extracellular matrix (ECM) components was strongly reduced, suggesting that deficiency in ECM composition and/or signaling may underlie the observed defects. Prolonged expression of Sox9 in the ureteric mesenchyme led to increased deposition of ECM components and SMC dispersal. Furthermore, Sox9 genetically interacts with the T-box transcription factor 18 gene (Tbx18) during ureter development at two levels--as a downstream mediator of Tbx18 function and in a converging pathway. Together, our results argue that obstructive uropathies in campomelic dysplasia patients that are heterozygous for mutations in and around SOX9 arise from a primary requirement of Sox9 in the development of the ureteric mesenchyme.
Collapse
Affiliation(s)
- Rannar Airik
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, D-30625 Hannover, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mandel EM, Kaltenbrun E, Callis TE, Zeng XXI, Marques SR, Yelon D, Wang DZ, Conlon FL. The BMP pathway acts to directly regulate Tbx20 in the developing heart. Development 2010; 137:1919-29. [PMID: 20460370 DOI: 10.1242/dev.043588] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
TBX20 has been shown to be essential for vertebrate heart development. Mutations within the TBX20 coding region are associated with human congenital heart disease, and the loss of Tbx20 in a wide variety of model systems leads to cardiac defects and eventually heart failure. Despite the crucial role of TBX20 in a range of cardiac cellular processes, the signal transduction pathways that act upstream of Tbx20 remain unknown. Here, we have identified and characterized a conserved 334 bp Tbx20 cardiac regulatory element that is directly activated by the BMP/SMAD1 signaling pathway. We demonstrate that this element is both necessary and sufficient to drive cardiac-specific expression of Tbx20 in Xenopus, and that blocking SMAD1 signaling in vivo specifically abolishes transcription of Tbx20, but not that of other cardiac factors, such as Tbx5 and MHC, in the developing heart. We further demonstrate that activation of Tbx20 by SMAD1 is mediated by a set of novel, non-canonical, high-affinity SMAD-binding sites located within this regulatory element and that phospho-SMAD1 directly binds a non-canonical SMAD1 site in vivo. Finally, we show that these non-canonical sites are necessary and sufficient for Tbx20 expression in Xenopus, and that reporter constructs containing these sites are expressed in a cardiac-specific manner in zebrafish and mouse. Collectively, our findings define Tbx20 as a direct transcriptional target of the BMP/SMAD1 signaling pathway during cardiac maturation.
Collapse
Affiliation(s)
- Elizabeth M Mandel
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Callis TE, Pandya K, Seok HY, Tang RH, Tatsuguchi M, Huang ZP, Chen JF, Deng Z, Gunn B, Shumate J, Willis MS, Selzman CH, Wang DZ. MicroRNA-208a is a regulator of cardiac hypertrophy and conduction in mice. J Clin Invest 2009; 119:2772-86. [PMID: 19726871 DOI: 10.1172/jci36154] [Citation(s) in RCA: 623] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 06/10/2009] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small noncoding RNAs that have gained status as important regulators of gene expression. Here, we investigated the function and molecular mechanisms of the miR-208 family of miRNAs in adult mouse heart physiology. We found that miR-208a, which is encoded within an intron of alpha-cardiac muscle myosin heavy chain gene (Myh6), was actually a member of a miRNA family that also included miR-208b, which was determined to be encoded within an intron of beta-cardiac muscle myosin heavy chain gene (Myh7). These miRNAs were differentially expressed in the mouse heart, paralleling the expression of their host genes. Transgenic overexpression of miR-208a in the heart was sufficient to induce hypertrophic growth in mice, which resulted in pronounced repression of the miR-208 regulatory targets thyroid hormone-associated protein 1 and myostatin, 2 negative regulators of muscle growth and hypertrophy. Studies of the miR-208a Tg mice indicated that miR-208a expression was sufficient to induce arrhythmias. Furthermore, analysis of mice lacking miR-208a indicated that miR-208a was required for proper cardiac conduction and expression of the cardiac transcription factors homeodomain-only protein and GATA4 and the gap junction protein connexin 40. Together, our studies uncover what we believe are novel miRNA-dependent mechanisms that modulate cardiac hypertrophy and electrical conduction.
Collapse
Affiliation(s)
- Thomas E Callis
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Qin H, Chan MWY, Liyanarachchi S, Balch C, Potter D, Souriraj IJ, Cheng ASL, Agosto-Perez FJ, Nikonova EV, Yan PS, Lin HJ, Nephew KP, Saltz JH, Showe LC, Huang THM, Davuluri RV. An integrative ChIP-chip and gene expression profiling to model SMAD regulatory modules. BMC SYSTEMS BIOLOGY 2009; 3:73. [PMID: 19615063 PMCID: PMC2724489 DOI: 10.1186/1752-0509-3-73] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Accepted: 07/17/2009] [Indexed: 12/24/2022]
Abstract
Background The TGF-β/SMAD pathway is part of a broader signaling network in which crosstalk between pathways occurs. While the molecular mechanisms of TGF-β/SMAD signaling pathway have been studied in detail, the global networks downstream of SMAD remain largely unknown. The regulatory effect of SMAD complex likely depends on transcriptional modules, in which the SMAD binding elements and partner transcription factor binding sites (SMAD modules) are present in specific context. Results To address this question and develop a computational model for SMAD modules, we simultaneously performed chromatin immunoprecipitation followed by microarray analysis (ChIP-chip) and mRNA expression profiling to identify TGF-β/SMAD regulated and synchronously coexpressed gene sets in ovarian surface epithelium. Intersecting the ChIP-chip and gene expression data yielded 150 direct targets, of which 141 were grouped into 3 co-expressed gene sets (sustained up-regulated, transient up-regulated and down-regulated), based on their temporal changes in expression after TGF-β activation. We developed a data-mining method driven by the Random Forest algorithm to model SMAD transcriptional modules in the target sequences. The predicted SMAD modules contain SMAD binding element and up to 2 of 7 other transcription factor binding sites (E2F, P53, LEF1, ELK1, COUPTF, PAX4 and DR1). Conclusion Together, the computational results further the understanding of the interactions between SMAD and other transcription factors at specific target promoters, and provide the basis for more targeted experimental verification of the co-regulatory modules.
Collapse
Affiliation(s)
- Huaxia Qin
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Yang L, Jiang J, Drouin LM, Agbandje-Mckenna M, Chen C, Qiao C, Pu D, Hu X, Wang DZ, Li J, Xiao X. A myocardium tropic adeno-associated virus (AAV) evolved by DNA shuffling and in vivo selection. Proc Natl Acad Sci U S A 2009; 106:3946-51. [PMID: 19234115 PMCID: PMC2656185 DOI: 10.1073/pnas.0813207106] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Indexed: 01/11/2023] Open
Abstract
To engineer gene vectors that target striated muscles after systemic delivery, we constructed a random library of adeno-associated virus (AAV) by shuffling the capsid genes of AAV serotypes 1 to 9, and screened for muscle-targeting capsids by direct in vivo panning after tail vein injection in mice. After 2 rounds of in vivo selection, a capsid gene named M41 was retrieved mainly based on its high frequency in the muscle and low frequency in the liver. Structural analyses revealed that the AAVM41 capsid is a recombinant of AAV1, 6, 7, and 8 with a mosaic capsid surface and a conserved capsid interior. AAVM41 was then subjected to a side-by-side comparison to AAV9, the most robust AAV for systemic heart and muscle gene delivery; to AAV6, a parental AAV with strong muscle tropism. After i.v. delivery of reporter genes, AAVM41 was found more efficient than AAV6 in the heart and muscle, and was similar to AAV9 in the heart but weaker in the muscle. In fact, the myocardium showed the highest gene expression among all tissues tested in mice and hamsters after systemic AAVM41 delivery. However, gene transfer in non-muscle tissues, mainly the liver, was dramatically reduced. AAVM41 was further tested in a genetic cardiomyopathy hamster model and achieved efficient long-term delta-sarcoglycan gene expression and rescue of cardiac functions. Thus, direct in vivo panning of capsid libraries is a simple tool for the de-targeting and retargeting of viral vector tissue tropisms facilitated by acquisition of desirable sequences and properties.
Collapse
Affiliation(s)
- Lin Yang
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Jiangang Jiang
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Lauren M. Drouin
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610; and
| | - Mavis Agbandje-Mckenna
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610; and
| | - Chunlian Chen
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Chunping Qiao
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Dongqiuye Pu
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Xiaoyun Hu
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599
| | - Da-Zhi Wang
- Carolina Cardiovascular Biology Center, University of North Carolina, Chapel Hill, NC 27599
| | - Juan Li
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| | - Xiao Xiao
- Division of Molecular Pharmaceutics, University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC 27599
| |
Collapse
|
26
|
Bessa PC, Casal M, Reis RL. Bone morphogenetic proteins in tissue engineering: the road from the laboratory to the clinic, part I (basic concepts). J Tissue Eng Regen Med 2008; 2:1-13. [PMID: 18293427 DOI: 10.1002/term.63] [Citation(s) in RCA: 230] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Discovered in 1965, bone morphogenetic proteins (BMPs) are a group of cytokines from the transforming growth factor-beta (TGFbeta) superfamily with significant roles in bone and cartilage formation. BMPs are used as powerful osteoinductive components of diverse tissue-engineering products for the healing of bone. Several BMPs with different physiological roles have been identified in humans. The purpose of this review is to cover the biological function of the main members of BMP family, the latest research on BMPs signalling pathways and advances in the production of recombinant BMPs for tissue engineering purposes.
Collapse
Affiliation(s)
- P C Bessa
- 3Bs Research Group, Biomaterials, Biodegradables and Biomimetics, Department of Polymer Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | | | | |
Collapse
|
27
|
Myocardin inhibits cellular proliferation by inhibiting NF-kappaB(p65)-dependent cell cycle progression. Proc Natl Acad Sci U S A 2008; 105:3362-7. [PMID: 18296632 DOI: 10.1073/pnas.0705842105] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We previously reported the importance of the serum response factor (SRF) cofactor myocardin in controlling muscle gene expression as well as the fundamental role for the inflammatory transcription factor NF-kappaB in governing cellular fate. Inactivation of myocardin has been implicated in malignant tumor growth. However, the underlying mechanism of myocardin regulation of cellular growth remains unclear. Here we show that NF-kappaB(p65) represses myocardin activation of cardiac and smooth muscle genes in a CArG-box-dependent manner. Consistent with their functional interaction, p65 directly interacts with myocardin and inhibits the formation of the myocardin/SRF/CArG ternary complex in vitro and in vivo. Conversely, myocardin decreases p65-mediated target gene activation by interfering with p65 DNA binding and abrogates LPS-induced TNF-alpha expression. Importantly, myocardin inhibits cellular proliferation by interfering with NF-kappaB-dependent cell-cycle regulation. Cumulatively, these findings identify a function for myocardin as an SRF-independent transcriptional repressor and cell-cycle regulator and provide a molecular mechanism by which interaction between NF-kappaB and myocardin plays a central role in modulating cellular proliferation and differentiation.
Collapse
|
28
|
Erickson BP, Pierce AR, Simpson AK, Nash J, Grauer JN. 125I-labeled OP-1 is locally retained in a rabbit lumbar fusion model. Clin Orthop Relat Res 2008; 466:210-5. [PMID: 18196395 PMCID: PMC2505306 DOI: 10.1007/s11999-007-0017-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 09/10/2007] [Indexed: 01/31/2023]
Abstract
Osteogenic protein-1 is evolving as a potential bone graft alternative. Surgical site retention is important to maximize local osteoinduction and to limit peripheral effects. An established rabbit lumbar posterolateral fusion model was used to evaluate the systemic distribution and pharmacokinetics of locally applied osteogenic protein-1 delivered on a collagen carrier. L5-L6 intertransverse process fusions were performed on 27 New Zealand White rabbits. Radiolabeled (125)I-osteogenic protein-1 collagen putty was implanted. At intervals, whole blood, plasma, and excreta were analyzed for radioactivity with liquid scintillation counting. Surgical site and tissue radioactivity also were assessed by quantitative whole-body autoradioluminography of animals euthanized at times ranging from 6 hours to 35 days. Animals remaining at the final time were assessed for fusion with manual palpation, radiography, and histology. Limited distribution of radioactivity was observed in the blood, plasma, and tissues apart from at the surgical site and in the urinary bladder and thyroid. The mean residence time for osteogenic protein-1 collagen putty was 10.4 +/- 2.7 days. These excretion profiles and kinetic properties are similar to those described for recombinant human bone morphogenetic protein-2 in the rabbit model (mean residence times of 7.6 days and 10.2 days with different carriers).
Collapse
Affiliation(s)
- Benjamin P. Erickson
- Department of Orthopaedics, Rehabilitation, Yale University School of Medicine, PO Box 208071, New Haven, CT 06520-8071 USA
| | | | - Andrew K. Simpson
- Department of Orthopaedics, Rehabilitation, Yale University School of Medicine, PO Box 208071, New Haven, CT 06520-8071 USA
| | - John Nash
- Charles River Laboratories Preclinical Services, Montreal, Quebec Canada
| | - Jonathan N. Grauer
- Department of Orthopaedics, Rehabilitation, Yale University School of Medicine, PO Box 208071, New Haven, CT 06520-8071 USA
| |
Collapse
|
29
|
Myocardin A Enhances Telomerase Activities in Adipose Tissue Mesenchymal Cells and Embryonic Stem Cells Undergoing Cardiovascular Myogenic Differentiation. Stem Cells 2008; 26:202-11. [DOI: 10.1634/stemcells.2007-0490] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
30
|
Morita T, Mayanagi T, Sobue K. Dual roles of myocardin-related transcription factors in epithelial mesenchymal transition via slug induction and actin remodeling. ACTA ACUST UNITED AC 2007; 179:1027-42. [PMID: 18056415 PMCID: PMC2099179 DOI: 10.1083/jcb.200708174] [Citation(s) in RCA: 221] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epithelial–mesenchymal transition (EMT) is a critical process occurring during embryonic development and in fibrosis and tumor progression. Dissociation of cell–cell contacts and remodeling of the actin cytoskeleton are major events of the EMT. Here, we show that myocardin-related transcription factors (MRTFs; also known as MAL and MKL) are critical mediators of transforming growth factor β (TGF-β) 1–induced EMT. In all epithelial cell lines examined here, TGF-β1 triggers the nuclear translocation of MRTFs. Ectopic expression of constitutive-active MRTF-A induces EMT, whereas dominant-negative MRTF-A or knockdown of MRTF-A and -B prevents the TGF-β1–induced EMT. MRTFs form complexes with Smad3. Via Smad3, the MRTF–Smad3 complexes bind to a newly identified cis-element GCCG-like motif in the promoter region of Canis familiaris and the human slug gene, which activates slug transcription and thereby dissociation of cell–cell contacts. MRTFs also increase the expression levels of actin cytoskeletal proteins via serum response factor, thereby triggering reorganization of the actin cytoskeleton. Thus, MRTFs are important mediators of TGF-β1–induced EMT.
Collapse
Affiliation(s)
- Tsuyoshi Morita
- Department of Neuroscience, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | | | | |
Collapse
|
31
|
Kontaraki JE, Parthenakis FI, Patrianakos AP, Karalis IK, Vardas PE. Myocardin gene regulatory variants as surrogate markers of cardiac hypertrophy - study in a genetically homogeneous population. Clin Genet 2007; 73:71-8. [PMID: 18028454 DOI: 10.1111/j.1399-0004.2007.00932.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Myocardin is thought to contribute to heart hypertrophy as assessed in animal models. The aim of this study was to identify polymorphisms on the myocardin gene and investigate possible relationships with left ventricular structure in human hypertrophic cardiomyopathy (HCM). Eighty-four native Cretan individuals (36 patients with HCM and 48 healthy controls) were examined by direct sequencing and subsequent restriction fragment length polymorphism analysis and six polymorphisms were identified in the promoter region at positions -435T>C (rs758187), -629A>T (rs8071072), -1030C>G (rs1233851), -1069A>G, -1166A>G and -1406G>A (rs976906). Allele and haplotype frequencies were not significantly different between patients and controls. However, patients carrying the [-435C;-629T] allelic variant had decreased left ventricular wall thickness (LVWT, p = 0.020) and left ventricular mass (p = 0.006) as compared with the wild-type genotype. Carrier status of this myocardin promoter allelic variant was also associated with significant lower myocardin mRNA levels in peripheral blood (p = 0.039). Thus, a myocardin promoter allelic variant existing in the normal Cretan population was associated with decreased left ventricular mass in HCM patients and decreased myocardin mRNA levels in peripheral blood. Our results may be limited by the limited sample size, but are strengthened by the genetic homogeneity of the Cretan population. Our data suggest that functional natural myocardin promoter variation might be a genetic factor contributing to inter-individual differences in the development of cardiac hypertrophy.
Collapse
Affiliation(s)
- J E Kontaraki
- Molecular Cardiology Laboratory, Division of Internal Medicine, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece.
| | | | | | | | | |
Collapse
|
32
|
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: 8.2] [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
|
33
|
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: 4.8] [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
|
34
|
Warkman AS, Yatskievych TA, Hardy KM, Krieg PA, Antin PB. Myocardin expression during avian embryonic heart development requires the endoderm but is independent of BMP signaling. Dev Dyn 2007; 237:216-21. [DOI: 10.1002/dvdy.21393] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
35
|
Oka T, Xu J, Molkentin JD. Re-employment of developmental transcription factors in adult heart disease. Semin Cell Dev Biol 2006; 18:117-31. [PMID: 17161634 PMCID: PMC1855184 DOI: 10.1016/j.semcdb.2006.11.012] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A finite number of transcription factors constitute a combinatorial code that orchestrates cardiac development and the specification and differentiation of myocytes. Many, if not all of these same transcription factors are re-employed in the adult heart in response to disease stimuli that promote hypertrophic enlargement and/or dilated cardiomyopathy, as part of the so-called "fetal gene program". This review will discuss the transcription factors that regulate the hypertrophic growth response of the adult heart, with a special emphasis on those regulators that participate in cardiac development.
Collapse
|
36
|
van Tuyn J, Atsma DE, Winter EM, van der Velde-van Dijke I, Pijnappels DA, Bax NAM, Knaän-Shanzer S, Gittenberger-de Groot AC, Poelmann RE, van der Laarse A, van der Wall EE, Schalij MJ, de Vries AAF. Epicardial cells of human adults can undergo an epithelial-to-mesenchymal transition and obtain characteristics of smooth muscle cells in vitro. Stem Cells 2006; 25:271-8. [PMID: 16990583 DOI: 10.1634/stemcells.2006-0366] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Myocardial and coronary development are both critically dependent on epicardial cells. During cardiomorphogenesis, a subset of epicardial cells undergoes an epithelial-to-mesenchymal transition (EMT) and invades the myocardium to differentiate into various cell types, including coronary smooth muscle cells and perivascular and cardiac interstitial fibroblasts. Our current knowledge of epicardial EMT and the ensuing epicardium-derived cells (EPDCs) comes primarily from studies of chick and mouse embryonic development. Due to the absence of an in vitro culture system, very little is known about human EPDCs. Here, we report for the first time the establishment of cultures of primary epicardial cells from human adults and describe their immunophenotype, transcriptome, transducibility, and differentiation potential in vitro. Changes in morphology and beta-catenin staining pattern indicated that human epicardial cells spontaneously undergo EMT early during ex vivo culture. The surface antigen profile of the cells after EMT closely resembles that of subepithelial fibroblasts; however, only EPDCs express the cardiac marker genes GATA4 and cardiac troponin T. After infection with an adenovirus vector encoding the transcription factor myocardin or after treatment with transforming growth factor-beta1 or bone morphogenetic protein-2, EPDCs obtain characteristics of smooth muscle cells. Moreover, EPDCs can undergo osteogenesis but fail to form adipocytes or endothelial cells in vitro. Cultured epicardial cells from human adults recapitulate at least part of the differentiation potential of their embryonic counterparts and represent an excellent model system to explore the biological properties and therapeutic potential of these cells.
Collapse
Affiliation(s)
- John van Tuyn
- Department of Cardiology, Leiden University Medical Center, Leiden, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Pipes GCT, Creemers EE, Olson EN. The myocardin family of transcriptional coactivators: versatile regulators of cell growth, migration, and myogenesis. Genes Dev 2006; 20:1545-56. [PMID: 16778073 DOI: 10.1101/gad.1428006] [Citation(s) in RCA: 377] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The association of transcriptional coactivators with sequence-specific DNA-binding proteins provides versatility and specificity to gene regulation and expands the regulatory potential of individual cis-regulatory DNA sequences. Members of the myocardin family of coactivators activate genes involved in cell proliferation, migration, and myogenesis by associating with serum response factor (SRF). The partnership of myocardin family members and SRF also controls genes encoding components of the actin cytoskeleton and confers responsiveness to extracellular growth signals and intracellular changes in the cytoskeleton, thereby creating a transcriptional-cytoskeletal regulatory circuit. These functions are reflected in defects in smooth muscle differentiation and function in mice with mutations in myocardin family members. This article reviews the functions and mechanisms of action of the myocardin family of coactivators and the physiological significance of transcriptional coactivation in the context of signal-dependent and cell-type-specific gene regulation.
Collapse
Affiliation(s)
- G C Teg Pipes
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | |
Collapse
|
38
|
Gude N, Muraski J, Rubio M, Kajstura J, Schaefer E, Anversa P, Sussman MA. Akt promotes increased cardiomyocyte cycling and expansion of the cardiac progenitor cell population. Circ Res 2006; 99:381-8. [PMID: 16840722 DOI: 10.1161/01.res.0000236754.21499.1c] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of Akt is associated with enhanced cell cycling and cellular proliferation in nonmyocytes, but this effect of nuclear Akt accumulation has not been explored in the context of the myocardium. Cardiac-specific expression of nuclear-targeted Akt (Akt/nuc) in transgenics prolongs postnatal cell cycling as evidenced by increased numbers of Ki67+ cardiomyocytes at 2 to 3 weeks after birth. Similarly, nuclear-targeting of Akt promotes expansion of the presumptive cardiac progenitor cell population as assessed by immunolabeling for c-kit in combination with myocyte-specific markers Nkx 2.5 or MEF 2C. Increases in pro-proliferative cytokines, including tumor-necrosis superfamily 8, interleukin-17e, and hepatocyte growth factor, were found in nuclear-targeted Akt myocardial samples. Concurrent signaling mediated by paracrine factors downstream of Akt/nuc expression may be responsible for phenotypic effects of nuclear-targeted Akt in the myocardium, including enhanced cell proliferation and expansion of the stem cell population.
Collapse
Affiliation(s)
- Natalie Gude
- San Diego State University Heart Institute and Department of Biology, San Diego State University, San Diego, CA 92182, USA
| | | | | | | | | | | | | |
Collapse
|