1
|
Wang X, Sun H, Yu H, Du B, Fan Q, Jia B, Zhang Z. Bone morphogenetic protein 10, a rising star in the field of diabetes and cardiovascular disease. J Cell Mol Med 2024; 28:e18324. [PMID: 38760897 PMCID: PMC11101671 DOI: 10.1111/jcmm.18324] [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: 10/16/2023] [Revised: 12/12/2023] [Accepted: 04/03/2024] [Indexed: 05/20/2024] Open
Abstract
Early research suggested that bone morphogenetic protein 10 (BMP10) is primarily involved in cardiac development and congenital heart disease processes. BMP10 is a newly identified cardiac-specific protein. In recent years, reports have emphasized the effects of BMP10 on myocardial apoptosis, fibrosis and immune response, as well as its synergistic effects with BMP9 in vascular endothelium and role in endothelial dysfunction. We believe that concentrating on this aspect of the study will enhance our knowledge of the pathogenesis of diabetes and the cardiovascular field. However, there have been no reports of any reviews discussing the role of BMP10 in diabetes and cardiovascular disease. In addition, the exact pathogenesis of diabetic cardiomyopathy is not fully understood, including myocardial energy metabolism disorders, microvascular changes, abnormal apoptosis of cardiomyocytes, collagen structural changes and myocardial fibrosis, all of which cause cardiac function impairment directly or indirectly and interact with one another. This review summarizes the research results of BMP10 in cardiac development, endothelial function and cardiovascular disease in an effort to generate new ideas for future research into diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Xueyin Wang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabolismAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| | - Helin Sun
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Haomiao Yu
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Bingyu Du
- Teaching and Research Section of Internal Medicine, College of MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Qi Fan
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Baoxue Jia
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
| | - Zhongwen Zhang
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabologyThe Third Affiliated Hospital of Shandong First Medical UniversityJinanChina
- Department of Endocrinology and MetabolismAffiliated Hospital of Shandong Second Medical UniversityWeifangChina
| |
Collapse
|
2
|
Gu JN, Yang CX, Ding YY, Qiao Q, Di RM, Sun YM, Wang J, Yang L, Xu YJ, Yang YQ. Identification of BMP10 as a Novel Gene Contributing to Dilated Cardiomyopathy. Diagnostics (Basel) 2023; 13:diagnostics13020242. [PMID: 36673052 PMCID: PMC9857772 DOI: 10.3390/diagnostics13020242] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Dilated cardiomyopathy (DCM), characterized by left ventricular or biventricular enlargement with systolic dysfunction, is the most common type of cardiac muscle disease. It is a major cause of congestive heart failure and the most frequent indication for heart transplantation. Aggregating evidence has convincingly demonstrated that DCM has an underlying genetic basis, though the genetic defects responsible for DCM in a larger proportion of cases remain elusive, motivating the ongoing research for new DCM-causative genes. In the current investigation, a multigenerational family affected with autosomal-dominant DCM was recruited from the Chinese Han population. By whole-exome sequencing and Sanger sequencing analyses of the DNAs from the family members, a new BMP10 variation, NM_014482.3:c.166C > T;p.(Gln56*), was discovered and verified to be in co-segregation with the DCM phenotype in the entire family. The heterozygous BMP10 variant was not detected in 268 healthy volunteers enrolled as control subjects. The functional measurement via dual-luciferase reporter assay revealed that Gln56*-mutant BMP10 lost the ability to transactivate its target genes NKX2.5 and TBX20, two genes that had been causally linked to DCM. The findings strongly indicate BMP10 as a new gene contributing to DCM in humans and support BMP10 haploinsufficiency as an alternative pathogenic mechanism underpinning DCM, implying potential implications for the early genetic diagnosis and precision prophylaxis of DCM.
Collapse
Affiliation(s)
- Jia-Ning Gu
- Department of Cardiology, Shanghai Fifth People′s Hospital, Fudan University, Shanghai 200240, China
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People′s Hospital, Fudan University, Shanghai 200240, China
| | - Yuan-Yuan Ding
- Shanghai Health Development Research Center, Shanghai Medical Information Center, Shanghai 200031, China
| | - Qi Qiao
- Department of Cardiology, Shanghai Fifth People′s Hospital, Fudan University, Shanghai 200240, China
| | - Ruo-Min Di
- Department of Cardiology, Shanghai Fifth People′s Hospital, Fudan University, Shanghai 200240, China
| | - Yu-Min Sun
- Department of Cardiology, Shanghai Jing’an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Jun Wang
- Department of Cardiology, Shanghai Jing’an District Central Hospital, Fudan University, Shanghai 200040, China
| | - Ling Yang
- Department of Ultrasound, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People′s Hospital, Fudan University, Shanghai 200240, China
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| | - 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
- Correspondence: (Y.-J.X.); (Y.-Q.Y.)
| |
Collapse
|
3
|
An P, Fan D, Guo Z, Liu FY, Li CF, Yang D, Wang MY, Yang Z, Tang QZ. Bone morphogenetic protein 10 alleviates doxorubicin-induced cardiac injury via signal transducer and activator of transcription 3 signaling pathway. Bioengineered 2022; 13:7471-7484. [PMID: 35293279 PMCID: PMC9208532 DOI: 10.1080/21655979.2022.2048994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Doxorubicin (DOX) has limited antitumor applications owing to its association with life-threatening cardiac injury. Oxidative damage and cardiac apoptosis are crucial in DOX-induced cardiac injury. Bone morphogenetic protein 10 (BMP10) is predominantly distributed in the heart and acts as a cardioprotective factor that preserves cardiac function. However, the role of BMP10 in DOX-induced cardiac injury has not yet been explored. The current study aimed to examine the function and mechanism of action of BMP10 in DOX-induced cardiac injury. An adeno-associated viral system was used for the overexpression or silencing of cardiac-specific BMP10, and subsequently, a single dose of DOX was intraperitoneally injected to induce cardiac injury. Results showed that DOX exposure decreased BMP10 expression in the heart. Cardiac-specific overexpression of BMP10 alleviated the oxidative stress and apoptosis and improved cardiac function. Conversely, cardiac-specific silencing of BMP10 aggravated the redox disorder and apoptosis and worsened the cardiac dysfunction caused by DOX. Exogenous BMP10 supplementation amelioratesd the DOX-induced cardiac contractile dysfunction. Mechanistically, we found that phosphorylation of signal transducer and activator of transcription 3 (STAT3) is reduced in DOX-induced cardiotoxicity, and, BMP10 activated impaired STAT3 via a non-canonical pathway. BMP10 lost its cardioprotective function in cardiomyocyte-specific STAT3 knockout (STAT3-cKO) mice. Based on our findings, we suggested that BMP10 is a potential therapeutic agent against DOX-induced cardiac injury and that the cardioprotective effects of BMP10 are dependent on the activation of STAT3.
Collapse
Affiliation(s)
- Peng An
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Di Fan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Fang-Yuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Chen-Fei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Ming-Yu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, RP China.,Hubei Key Laboratory of Metabolic And Chronic Diseases, Wuhan, RP China.,Cardiovascular Research Institute of Wuhan University, Wuhan 430060, RP China
| |
Collapse
|
4
|
Herbal Extract from Codonopsis pilosula (Franch.) Nannf. Enhances Cardiogenic Differentiation and Improves the Function of Infarcted Rat Hearts. Life (Basel) 2021; 11:life11050422. [PMID: 34063127 PMCID: PMC8148170 DOI: 10.3390/life11050422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/01/2021] [Accepted: 05/02/2021] [Indexed: 11/25/2022] Open
Abstract
Background: The roots of Codonopsis pilosula (Franch.) Nannf. have been used in traditional Chinese medicine for treating cardiovascular disease. In the current study, we aimed to discover herbal extracts from C. pilosula that are capable of improving cardiac function of infarcted hearts to develop a potential therapeutic approach. Methods: A mouse embryonic stem (ES) cell-based model with an enhanced green fluorescent protein (eGFP) reporter driven by a cardiomyocyte-specific promoter, the α-myosin heavy chain, was constructed to evaluate the cardiogenic activity of herbal extracts. Then, herbal extracts from C. pilosula with cardiogenic activity based on an increase in eGFP expression during ES cell differentiation were further tested in a rat myocardial infarction model with left anterior descending artery (LAD) ligation. Cardiac function assessments were performed using echocardiography, 1, 3, and 6 weeks post LAD ligation. Results: The herbal extract 417W from C. pilosula was capable of enhancing cardiogenic differentiation in mouse ES cells in vitro. Echocardiography results in the LAD-ligated rat model revealed significant improvements in the infarcted hearts at least 6 weeks after 417W treatment that were determined based on left ventricle fractional shortening (FS), fractional area contraction (FAC), and ejection fraction (EF). Conclusions: The herbal extract 417W can enhance the cardiogenic differentiation of ES cells and improve the cardiac function of infarcted hearts.
Collapse
|
5
|
Meng Y, Zhong K, Xiao J, Huang Y, Wei Y, Tang L, Chen S, Wu J, Ma J, Cao Z, Liao X, Lu H. Exposure to pyrimethanil induces developmental toxicity and cardiotoxicity in zebrafish. CHEMOSPHERE 2020; 255:126889. [PMID: 32388256 DOI: 10.1016/j.chemosphere.2020.126889] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 05/27/2023]
Abstract
Pyrimethanil is a broad-spectrum fungicide commonly used in the prevention and treatment of Botrytis cinerea. However, little information is available in the literature to show the toxicity of Pyrimethanil to cardiac development. In this study, we used an experimental animal model to explore the developmental and cardiac toxicity of Pyrimethanil in aquatic vertebrates; we exposed zebrafish embryos to Pyrimethanil at concentrations of 2, 4, and 6 mg/L from 5.5 to 72 h post fertilisation. We found that Pyrimethanil caused a decrease in the hatching rate, heart rate, and survival rate of zebrafish embryos. Pyrimethanil exposure also resulted in pericardial and yolk sac edema, spinal deformity, and heart loop failure. Moreover, Pyrimethanil increased reactive oxygen stress levels and heightened the activity of superoxide dismutase and catalase. Alterations were induced in the transcription of apoptosis-related genes (p53, Bax, Bcl2, Casp 9, and Casp6l1) and heart development-related genes (Tbx2b, Gata4, Myh6, Vmhc, Nppa, Bmp2b, Bpm 4, and Bpm 10). Our data showed that the activation of Wnt signalling by BML-284 could partially rescue the malformed phenotype caused by Pyrimethanil. Our results provide new evidence for Pyrimethanil's toxicity and the danger of its residues in the environment and agricultural products.
Collapse
Affiliation(s)
- Yunlong Meng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Lin Tang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Suping Chen
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juan Wu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China.
| |
Collapse
|
6
|
Qu X, Liu Y, Cao D, Chen J, Liu Z, Ji H, Chen Y, Zhang W, Zhu P, Xiao D, Li X, Shou W, Chen H. BMP10 preserves cardiac function through its dual activation of SMAD-mediated and STAT3-mediated pathways. J Biol Chem 2019; 294:19877-19888. [PMID: 31712309 DOI: 10.1074/jbc.ra119.010943] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic protein 10 (BMP10) is a cardiac peptide growth factor belonging to the transforming growth factor β superfamily that critically controls cardiovascular development, growth, and maturation. It has been shown that BMP10 elicits its intracellular signaling through a receptor complex of activin receptor-like kinase 1 with morphogenetic protein receptor type II or activin receptor type 2A. Previously, we generated and characterized a transgenic mouse line expressing BMP10 from the α-myosin heavy chain gene promoter and found that these mice have normal cardiac hypertrophic responses to both physiological and pathological stimuli. In this study, we report that these transgenic mice exhibit significantly reduced levels of cardiomyocyte apoptosis and cardiac fibrosis in response to a prolonged administration of the β-adrenoreceptor agonist isoproterenol. We further confirmed this cardioprotective function with a newly generated conditional Bmp10 transgenic mouse line, in which Bmp10 was activated in adult hearts by tamoxifen. Moreover, the intraperitoneal administration of recombinant human BMP10 was found to effectively protect hearts from injury, suggesting potential therapeutic utility of using BMP10 to prevent heart failure. Gene profiling and biochemical analyses indicated that BMP10 activates the SMAD-mediated canonical pathway and, unexpectedly, also the signal transducer and activator of transcription 3 (STAT3)-mediated signaling pathway both in vivo and in vitro Additional findings further supported the notion that BMP10's cardioprotective function likely is due to its dual activation of SMAD- and STAT3-regulated signaling pathways, promoting cardiomyocyte survival and suppressing cardiac fibrosis.
Collapse
Affiliation(s)
- Xiuxia Qu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China.,Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Ying Liu
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Dayan Cao
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Jinghai Chen
- Department of Cardiology, the Second Affiliate Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Zhejiang 310029, China
| | - Zhuo Liu
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hongrui Ji
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202.,School of Chemical and Environmental Engineering, Harbin University of Science and Technology, Heilongjiang 150040, China
| | - Yuwen Chen
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Wenjun Zhang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Ping Zhu
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong 510100, China
| | - Deyong Xiao
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202.,Fountain Valley Institute of Life Sciences and Fountain Valley Biotechnology Inc., Dalian Hi-Tech Industrial Zone, Liaoning 116023, China
| | - Xiaohui Li
- Institute of Materia Medica and Center of Translational Medicine, College of Pharmacy, Army Medical University, Chongqing 400038, China
| | - Weinian Shou
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Hanying Chen
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| |
Collapse
|
7
|
Tatebe S, Sugimura K, Aoki T, Yamamoto S, Yaoita N, Suzuki H, Sato H, Kozu K, Konno R, Satoh K, Fukuda K, Adachi O, Saito R, Nakanishi N, Morisaki H, Oyama K, Saiki Y, Okada Y, Shimokawa H. The Efficacy of a Genetic Analysis of the BMPR2 Gene in a Patient with Severe Pulmonary Arterial Hypertension and an Atrial Septal Defect Treated with Bilateral Lung Transplantation. Intern Med 2017; 56:3193-3197. [PMID: 29021450 PMCID: PMC5742392 DOI: 10.2169/internalmedicine.8686-16] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Severe pulmonary arterial hypertension (PAH) rarely develops in children with an atrial septal defect (ASD), even those with a large defect. We herein report the case of a 27-year-old man with a moderate-sized secundum ASD and right ventricular failure due to severe PAH, which developed in his early teens. He was diagnosed as having a genetic mutation of the bone morphogenetic protein receptor-2 (BMPR2) gene and was successfully treated with bilateral lung transplantation with ASD path closure. In patients with congenital heart disease, a genetic analysis may provide information about the lifetime risk of developing PAH.
Collapse
Affiliation(s)
- Shunsuke Tatebe
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Koichiro Sugimura
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Tatsuo Aoki
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Saori Yamamoto
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Nobuhiro Yaoita
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Hideaki Suzuki
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Haruka Sato
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Katsuya Kozu
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Ryo Konno
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Koji Fukuda
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| | - Osamu Adachi
- Department of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Ryoko Saito
- Department of Pathology, Tohoku University School of Medicine, Japan
| | - Norifumi Nakanishi
- Division of Pulmonary Circulation, Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Japan
| | - Hiroko Morisaki
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Japan
- Department of Medical Genetics, Sakakibara Heart Institute, Japan
| | - Kotaro Oyama
- Department of Pediatrics, Iwate Medical University, Japan
| | - Yoshikatsu Saiki
- Department of Cardiovascular Surgery, Tohoku University Graduate School of Medicine, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Japan
| |
Collapse
|
8
|
Fakoya AOJ, Otohinoyi DA, Marcelle T, Yusuf J. The Palm-Heart Diameter: A Prospective Simple Screening Tool for Identifying Heart Enlargement. Open Access Maced J Med Sci 2017; 5:818-824. [PMID: 29362603 PMCID: PMC5771279 DOI: 10.3889/oamjms.2017.181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 09/05/2017] [Accepted: 09/02/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND: Several speculations have linked the size of the fist to be equal to the size of the heart. However, the substantial scientific report still lacks to support this theory. AIM: This study aims to provide the validity of the fist-heart assumption by correlating the palm and heart diameters while benchmarking it as a reference tool for determining the normal heart size. MATERIALS AND METHOD: Volunteers from the public were recruited during a health fair organised by the school. A self-administered questionnaire for necessary information was distributed after the volunteers signed the consent forms. The palm of both hands was measured in duplicates using a flexible ruler. Ultrasound examination was used in measuring the diameter of the heart with the landmark being from the anterior fibrous pericardium to the lowest part of the posterior fibrous pericardium. The level of significance was kept at P < 0.05. RESULTS: A total of 275 people, consisting of 123 males and 152 females participated in the study. The age range was from 15 to 80 years with a mean age of 28.16 ± 16.18. The measurement showed that the size of both palms correlated with the heart diameter, p < 0.05. Other factors such as age and height showed a substantial level of correlation. However, this correlation ceased with older participants. Palm size did not correlate among participants with previously diagnosed prehypertension. However, participants with previously diagnosed hypertension with good medication compliance maintained the correlation. CONCLUSION: This study establishes the correlation between the palm and heart diameters. Since the heart tissue and the upper limb share a similar embryonic origin, being the mesoderm, this study prospects the fact that heart enlargement could be preliminarily identified by measuring the size of the hand.
Collapse
Affiliation(s)
| | - David Adeiza Otohinoyi
- Department of Anatomical Sciences, All Saints University School of Medicine Dominica, Dominica
| | - Terrence Marcelle
- Department of Anatomical Sciences, All Saints University School of Medicine Dominica, Dominica
| | - Joshua Yusuf
- Department of Anatomical Sciences, All Saints University School of Medicine Dominica, Dominica
| |
Collapse
|
9
|
Mesenchymal Stem Cells for Cardiac Regenerative Therapy: Optimization of Cell Differentiation Strategy. Stem Cells Int 2015; 2015:524756. [PMID: 26339251 PMCID: PMC4539177 DOI: 10.1155/2015/524756] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/28/2015] [Accepted: 03/11/2015] [Indexed: 01/25/2023] Open
Abstract
With the high mortality rate, coronary heart disease (CHD) has currently become a major life-threatening disease. The main pathological change of myocardial infarction (MI) is the induction of myocardial necrosis in infarction area which finally causes heart failure. Conventional treatments cannot regenerate the functional cell efficiently. Recent researches suggest that mesenchymal stem cells (MSCs) are able to differentiate into multiple lineages, including cardiomyocyte-like cells in vitro and in vivo, and they have been used for the treatment of MI to repair the injured myocardium and improve cardiac function. In this review, we will focus on the recent progress on MSCs derived cardiomyocytes for cardiac regeneration after MI.
Collapse
|
10
|
Fiebig JE, Weidauer SE, Qiu LY, Bauer M, Schmieder P, Beerbaum M, Zhang JL, Oschkinat H, Sebald W, Mueller TD. The clip-segment of the von Willebrand domain 1 of the BMP modulator protein Crossveinless 2 is preformed. Molecules 2013; 18:11658-82. [PMID: 24071977 PMCID: PMC6270503 DOI: 10.3390/molecules181011658] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 11/19/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are secreted protein hormones that act as morphogens and exert essential roles during embryonic development of tissues and organs. Signaling by BMPs occurs via hetero-oligomerization of two types of serine/threonine kinase transmembrane receptors. Due to the small number of available receptors for a large number of BMP ligands ligand-receptor promiscuity presents an evident problem requiring additional regulatory mechanisms for ligand-specific signaling. Such additional regulation is achieved through a plethora of extracellular antagonists, among them members of the Chordin superfamily, that modulate BMP signaling activity by binding. The key-element in Chordin-related antagonists for interacting with BMPs is the von Willebrand type C (VWC) module, which is a small domain of about 50 to 60 residues occurring in many different proteins. Although a structure of the VWC domain of the Chordin-member Crossveinless 2 (CV2) bound to BMP-2 has been determined by X-ray crystallography, the molecular mechanism by which the VWC domain binds BMPs has remained unclear. Here we present the NMR structure of the Danio rerio CV2 VWC1 domain in its unbound state showing that the key features for high affinity binding to BMP-2 is a pre-oriented peptide loop.
Collapse
Affiliation(s)
- Juliane E. Fiebig
- Julius-von-Sachs Institut für Biowissenschaften der Universität Würzburg, Julius-von-Sachs Platz 2, Würzburg D-97082, Germany; E-Mails: (J.E.F.); (S.E.W.); (M.B.)
| | - Stella E. Weidauer
- Julius-von-Sachs Institut für Biowissenschaften der Universität Würzburg, Julius-von-Sachs Platz 2, Würzburg D-97082, Germany; E-Mails: (J.E.F.); (S.E.W.); (M.B.)
| | - Li-Yan Qiu
- Lehrstuhl für Physiologische Chemie II, Biozentrum der Universität Würzburg, Am Hubland, Würzburg D-97074, Germany; E-Mails: (L.-Y.Q.); (J.-L.Z.); (W.S.)
| | - Markus Bauer
- Julius-von-Sachs Institut für Biowissenschaften der Universität Würzburg, Julius-von-Sachs Platz 2, Würzburg D-97082, Germany; E-Mails: (J.E.F.); (S.E.W.); (M.B.)
| | - Peter Schmieder
- Leibnizinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Robert-Roessle Str. 10, Berlin D-13125, Germany; E-Mails: (P.S.); (M.B.); (H.O.)
| | - Monika Beerbaum
- Leibnizinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Robert-Roessle Str. 10, Berlin D-13125, Germany; E-Mails: (P.S.); (M.B.); (H.O.)
| | - Jin-Li Zhang
- Lehrstuhl für Physiologische Chemie II, Biozentrum der Universität Würzburg, Am Hubland, Würzburg D-97074, Germany; E-Mails: (L.-Y.Q.); (J.-L.Z.); (W.S.)
| | - Hartmut Oschkinat
- Leibnizinstitut für Molekulare Pharmakologie (FMP), Campus Berlin-Buch, Robert-Roessle Str. 10, Berlin D-13125, Germany; E-Mails: (P.S.); (M.B.); (H.O.)
| | - Walter Sebald
- Lehrstuhl für Physiologische Chemie II, Biozentrum der Universität Würzburg, Am Hubland, Würzburg D-97074, Germany; E-Mails: (L.-Y.Q.); (J.-L.Z.); (W.S.)
| | - Thomas D. Mueller
- Julius-von-Sachs Institut für Biowissenschaften der Universität Würzburg, Julius-von-Sachs Platz 2, Würzburg D-97082, Germany; E-Mails: (J.E.F.); (S.E.W.); (M.B.)
| |
Collapse
|
11
|
Iwata JI, Suzuki A, Pelikan RC, Ho TV, Chai Y. Noncanonical transforming growth factor β (TGFβ) signaling in cranial neural crest cells causes tongue muscle developmental defects. J Biol Chem 2013; 288:29760-70. [PMID: 23950180 DOI: 10.1074/jbc.m113.493551] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Microglossia is a congenital birth defect in humans and adversely impacts quality of life. In vertebrates, tongue muscle derives from the cranial mesoderm, whereas tendons and connective tissues in the craniofacial region originate from cranial neural crest (CNC) cells. Loss of transforming growth factor β (TGFβ) type II receptor in CNC cells in mice (Tgfbr2(fl/fl);Wnt1-Cre) causes microglossia due to a failure of cell-cell communication between cranial mesoderm and CNC cells during tongue development. However, it is still unclear how TGFβ signaling in CNC cells regulates the fate of mesoderm-derived myoblasts during tongue development. Here we show that activation of the cytoplasmic and nuclear tyrosine kinase 1 (ABL1) cascade in Tgfbr2(fl/fl);Wnt1-Cre mice results in a failure of CNC-derived cell differentiation followed by a disruption of TGFβ-mediated induction of growth factors and reduction of myogenic cell proliferation and differentiation activities. Among the affected growth factors, the addition of fibroblast growth factor 4 (FGF4) and neutralizing antibody for follistatin (FST; an antagonist of bone morphogenetic protein (BMP)) could most efficiently restore cell proliferation, differentiation, and organization of muscle cells in the tongue of Tgfbr2(fl/fl);Wnt1-Cre mice. Thus, our data indicate that CNC-derived fibroblasts regulate the fate of mesoderm-derived myoblasts through TGFβ-mediated regulation of FGF and BMP signaling during tongue development.
Collapse
Affiliation(s)
- Jun-ichi Iwata
- From the Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California 90033
| | | | | | | | | |
Collapse
|
12
|
Complete and unidirectional conversion of human embryonic stem cells to trophoblast by BMP4. Proc Natl Acad Sci U S A 2013; 110:E1212-21. [PMID: 23493551 DOI: 10.1073/pnas.1303094110] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human ES cells (hESC) exposed to bone morphogenic protein 4 (BMP4) in the absence of FGF2 have become widely used for studying trophoblast development, but the soundness of this model has been challenged by others, who concluded that differentiation was primarily toward mesoderm rather than trophoblast. Here we confirm that hESC grown under the standard conditions on a medium conditioned by mouse embryonic fibroblasts in the presence of BMP4 and absence of FGF2 on a Matrigel substratum rapidly convert to an epithelium that is largely KRT7(+) within 48 h, with minimal expression of mesoderm markers, including T (Brachyury). Instead, they begin to express a series of trophoblast markers, including HLA-G, demonstrate invasive properties that are independent of the continued presence of BMP4 in the medium, and, over time, produce extensive amounts of human chorionic gonadotropin, progesterone, placental growth factor, and placental lactogen. This process of differentiation is not dependent on conditioning of the medium by mouse embryonic fibroblasts and is accelerated in the presence of inhibitors of Activin and FGF2 signaling, which at day 2 provide colonies that are entirely KRT7(+) and in which the majority of cells are transiently CDX2(+). Colonies grown on two chemically defined media, including the one in which BMP4 was reported to drive mesoderm formation, also differentiate at least partially to trophoblast in response to BMP4. The experiments demonstrate that the in vitro BMP4/hESC model is valid for studying the emergence and differentiation of trophoblasts.
Collapse
|
13
|
Huang J, Elicker J, Bowens N, Liu X, Cheng L, Cappola TP, Zhu X, Parmacek MS. Myocardin regulates BMP10 expression and is required for heart development. J Clin Invest 2012; 122:3678-91. [PMID: 22996691 DOI: 10.1172/jci63635] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Accepted: 07/26/2012] [Indexed: 01/12/2023] Open
Abstract
Myocardin is a muscle lineage-restricted transcriptional coactivator that has been shown to transduce extracellular signals to the nucleus required for SMC differentiation. We now report the discovery of a myocardin/BMP10 (where BMP10 indicates bone morphogenetic protein 10) signaling pathway required for cardiac growth, chamber maturation, and embryonic survival. Myocardin-null (Myocd) embryos and embryos harboring a cardiomyocyte-restricted mutation in the Myocd gene exhibited myocardial hypoplasia, defective atrial and ventricular chamber maturation, heart failure, and embryonic lethality. Cardiac hypoplasia was caused by decreased cardiomyocyte proliferation accompanied by a dramatic increase in programmed cell death. Defective chamber maturation and the block in cardiomyocyte proliferation were caused in part by a block in BMP10 signaling. Myocardin transactivated the Bmp10 gene via binding of a serum response factor-myocardin protein complex to a nonconsensus CArG element in the Bmp10 promoter. Expression of p57kip2, a BMP10-regulated cyclin-dependent kinase inhibitor, was induced in Myocd-/- hearts, while BMP10-activated cardiogenic transcription factors, including NKX2.5 and MEF2c, were repressed. Remarkably, when embryonic Myocd-/- hearts were cultured ex vivo in BMP10-conditioned medium, the defects in cardiomyocyte proliferation and p57kip2 expression were rescued. Taken together, these data identify a heretofore undescribed myocardin/BMP10 signaling pathway that regulates cardiomyocyte proliferation and apoptosis in the embryonic heart.
Collapse
Affiliation(s)
- Jianhe Huang
- University of Pennsylvania, Cardiovascular Institute, Department of Medicine, Philadelphia, PA 19104-5159, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Zhang Z, Li H, Ma Z, Feng J, Gao P, Dong H, Zhang Z. Efficient cardiomyogenic differentiation of bone marrow mesenchymal stromal cells by combination of Wnt11 and bone morphogenetic protein 2. Exp Biol Med (Maywood) 2012; 237:768-76. [PMID: 22829700 DOI: 10.1258/ebm.2012.011291] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wnt11 and bone morphogenetic protein 2 (BMP-2) are key signaling factors for stem cell differentiation into functional cardiomyocytes (CMs). In this study, we elucidate the biological effect of BMP-2 and Wnt11 on bone marrow mesenchymal stromal cells (BM-MSCs) that differentiate into myocardial-like cells in a simulated myocardial microenvironment in vitro. A cell co-culture system was established with recombinant Wnt11 treatment of NIH/3T3 cells and CMs. BMP-2 was added in a diverse schedule to induce cardiomyogenic differentiation of BM-MSCs co-cultured under various conditions. The levels of cardiac-specific markers Nkx2.5, α-myosin heavy chain ( α-MHC), β-myosin heavy chain ( β-MHC) and cardiac troponin I (cTnI) were determined by reverse transcriptase polymerase chain reaction and immunocytochemistry to evaluate cardiomyogenic differentiation. Wnt11 or BMP-2 used on their own to differentiate BM-MSCs resulted in no expression of α-MHC and cTnI. Wnt11 alone in a myocardial microenvironment enhanced cardiomyogenic differentiation. BMP-2 demonstrated a dose-dependent effect on BM-MSC differentiation into myocardial-like cells. Addition of BMP to BM-MSCs at various time points resulted in varying effects on cardiomyogenic differentiation. The combination of Wnt11 and BMP-2 treatment in a temporal manner significantly enhanced cardiomyogenic differentiation of BM-MSCs, with high expressions of α-MHC, β-MHC, Nkx2.5 and cTnI upon co-culture with CMs. Our study demonstrates that the combination of Wnt11 and BMP-2 effectively promotes cardiomyogenic differentiation of BM-MSCs in vitro. The synergistic effect of Wnt11 and BMP-2 on the cardiomyogenic differentiation of BM-MSCs is further enhanced in a myocardial microenvironment.
Collapse
Affiliation(s)
| | - Huixian Li
- Institute of Cardiovascular Diseases, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Zhifeng Ma
- Institute of Cardiovascular Diseases, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Junqing Feng
- Institute of Cardiovascular Diseases, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | - Pan Gao
- Institute of Cardiovascular Diseases, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| | | | - Zhongming Zhang
- Institute of Cardiovascular Diseases, Affiliated Hospital of Xuzhou Medical College, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
15
|
Goligorsky MS. BuMPy road of delayed graft function. Am J Physiol Renal Physiol 2012; 303:F21-2. [PMID: 22513849 DOI: 10.1152/ajprenal.00210.2012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
16
|
Lin H, Wang L, Jiang M, Huang J, Qi L. P-glycoprotein(ABCB1)inhibited network of mitochondrion transport along microtubule and BMP signal-induced cell shape in chimpanzee left cerebrum by systems-theoretical analysis. Cell Biochem Funct 2012; 30:582-7. [DOI: 10.1002/cbf.2837] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2011] [Revised: 03/01/2012] [Accepted: 04/02/2012] [Indexed: 12/20/2022]
Affiliation(s)
- Hong Lin
- Biomedical Center, School of Electronic Engineering; Beijing University of Posts and Telecommunications; Beijing; China
| | - Lin Wang
- Biomedical Center, School of Electronic Engineering; Beijing University of Posts and Telecommunications; Beijing; China
| | - Minghu Jiang
- Lab of Computational Linguistics, School of Humanities and Social Sciences; Tsinghua University; Beijing; China
| | - Juxiang Huang
- Biomedical Center, School of Electronic Engineering; Beijing University of Posts and Telecommunications; Beijing; China
| | - Lianxiu Qi
- Biomedical Center, School of Electronic Engineering; Beijing University of Posts and Telecommunications; Beijing; China
| |
Collapse
|
17
|
Torres J, Prieto J, Durupt FC, Broad S, Watt FM. Efficient differentiation of embryonic stem cells into mesodermal precursors by BMP, retinoic acid and Notch signalling. PLoS One 2012; 7:e36405. [PMID: 22558462 PMCID: PMC3340340 DOI: 10.1371/journal.pone.0036405] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 04/09/2012] [Indexed: 12/31/2022] Open
Abstract
The ability to direct differentiation of mouse embryonic stem (ES) cells into specific lineages not only provides new insights into the pathways that regulate lineage selection but also has translational applications, for example in drug discovery. We set out to develop a method of differentiating ES cells into mesodermal cells at high efficiency without first having to induce embryoid body formation. ES cells were plated on a feeder layer of PA6 cells, which have membrane-associated stromal-derived inducing activity (SDIA), the molecular basis of which is currently unknown. Stimulation of ES/PA6 co-cultures with Bone Morphogenetic Protein 4 (BMP4) both favoured self-renewal of ES cells and induced differentiation into a Desmin and Nestin double positive cell population. Combined stimulation with BMP4 and all-trans Retinoic Acid (RA) inhibited self-renewal and resulted in 90% of cells expressing Desmin and Nestin. Quantitative reverse transcription-polymerase chain reaction (qPCR) analysis confirmed that the cells were of mesodermal origin and expressed markers of mesenchymal and smooth muscle cells. BMP4 activation of a MAD-homolog (Smad)-dependent reporter in undifferentiated ES cells was attenuated by co-stimulation with RA and co-culture with PA6 cells. The Notch ligand Jag1 was expressed in PA6 cells and inhibition of Notch signalling blocked the differentiation inducing activity of PA6 cells. Our data suggest that mesodermal differentiation is regulated by the level of Smad activity as a result of inputs from BMP4, RA and the Notch pathway.
Collapse
Affiliation(s)
- Josema Torres
- Departamento de Biología Celular, Universidad de Valencia, Burjassot, Comunidad Valenciana, Spain
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (JT); (FMW)
| | - Javier Prieto
- Departamento de Biología Celular, Universidad de Valencia, Burjassot, Comunidad Valenciana, Spain
| | - Fabrice C. Durupt
- Departamento de Biología Celular, Universidad de Valencia, Burjassot, Comunidad Valenciana, Spain
| | - Simon Broad
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, United Kingdom
| | - Fiona M. Watt
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (JT); (FMW)
| |
Collapse
|
18
|
Bernardo AS, Faial T, Gardner L, Niakan KK, Ortmann D, Senner CE, Callery EM, Trotter MW, Hemberger M, Smith JC, Bardwell L, Moffett A, Pedersen RA. BRACHYURY and CDX2 mediate BMP-induced differentiation of human and mouse pluripotent stem cells into embryonic and extraembryonic lineages. Cell Stem Cell 2012; 9:144-55. [PMID: 21816365 PMCID: PMC3567433 DOI: 10.1016/j.stem.2011.06.015] [Citation(s) in RCA: 291] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 04/14/2011] [Accepted: 06/30/2011] [Indexed: 11/01/2022]
Abstract
BMP is thought to induce hESC differentiation toward multiple lineages including mesoderm and trophoblast. The BMP-induced trophoblast phenotype is a long-standing paradox in stem cell biology. Here we readdressed BMP function in hESCs and mouse epiblast-derived cells. We found that BMP4 cooperates with FGF2 (via ERK) to induce mesoderm and to inhibit endoderm differentiation. These conditions induced cells with high levels of BRACHYURY (BRA) that coexpressed CDX2. BRA was necessary for and preceded CDX2 expression; both genes were essential for expression not only of mesodermal genes but also of trophoblast-associated genes. Maximal expression of the latter was seen in the absence of FGF but these cells coexpressed mesodermal genes and moreover they differed in cell surface and epigenetic properties from placental trophoblast. We conclude that BMP induces human and mouse pluripotent stem cells primarily to form mesoderm, rather than trophoblast, acting through BRA and CDX2.
Collapse
Affiliation(s)
- Andreia S Bernardo
- The Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge CB2 0SZ, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Cheung C, Bernardo AS, Trotter MWB, Pedersen RA, Sinha S. Generation of human vascular smooth muscle subtypes provides insight into embryological origin-dependent disease susceptibility. Nat Biotechnol 2012; 30:165-73. [PMID: 22252507 PMCID: PMC3272383 DOI: 10.1038/nbt.2107] [Citation(s) in RCA: 292] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 12/19/2011] [Indexed: 12/28/2022]
Abstract
Heterogeneity of embryological origins is a hallmark of vascular smooth muscle cells (SMCs) and may influence the development of vascular disease. Differentiation of human pluripotent stem cells (hPSCs) into developmental origin-specific SMC subtypes remains elusive. Here we describe a chemically defined protocol in which hPSCs were initially induced to form neuroectoderm, lateral plate mesoderm or paraxial mesoderm. These intermediate populations were further differentiated toward SMCs (>80% MYH11(+) and ACTA2(+)), which displayed contractile ability in response to vasoconstrictors and invested perivascular regions in vivo. Derived SMC subtypes recapitulated the unique proliferative and secretory responses to cytokines previously documented in studies using aortic SMCs of distinct origins. Notably, this system predicted increased extracellular matrix degradation by SMCs derived from lateral plate mesoderm, which was confirmed using rat aortic SMCs from corresponding origins. This differentiation approach will have broad applications in modeling origin-dependent disease susceptibility and in developing bioengineered vascular grafts for regenerative medicine.
Collapse
Affiliation(s)
- Christine Cheung
- The Anne McLaren Laboratory for Regenerative Medicine, University of Cambridge, Cambridge, UK
| | | | | | | | | |
Collapse
|
20
|
Conidi A, Cazzola S, Beets K, Coddens K, Collart C, Cornelis F, Cox L, Joke D, Dobreva MP, Dries R, Esguerra C, Francis A, Ibrahimi A, Kroes R, Lesage F, Maas E, Moya I, Pereira PNG, Stappers E, Stryjewska A, van den Berghe V, Vermeire L, Verstappen G, Seuntjens E, Umans L, Zwijsen A, Huylebroeck D. Few Smad proteins and many Smad-interacting proteins yield multiple functions and action modes in TGFβ/BMP signaling in vivo. Cytokine Growth Factor Rev 2011; 22:287-300. [PMID: 22119658 DOI: 10.1016/j.cytogfr.2011.11.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Signaling by the many ligands of the TGFβ family strongly converges towards only five receptor-activated, intracellular Smad proteins, which fall into two classes i.e. Smad2/3 and Smad1/5/8, respectively. These Smads bind to a surprisingly high number of Smad-interacting proteins (SIPs), many of which are transcription factors (TFs) that co-operate in Smad-controlled target gene transcription in a cell type and context specific manner. A combination of functional analyses in vivo as well as in cell cultures and biochemical studies has revealed the enormous versatility of the Smad proteins. Smads and their SIPs regulate diverse molecular and cellular processes and are also directly relevant to development and disease. In this survey, we selected appropriate examples on the BMP-Smads, with emphasis on Smad1 and Smad5, and on a number of SIPs, i.e. the CPSF subunit Smicl, Ttrap (Tdp2) and Sip1 (Zeb2, Zfhx1b) from our own research carried out in three different vertebrate models.
Collapse
Affiliation(s)
- Andrea Conidi
- Laboratory of Molecular Biology (Celgen) of Center for Human Genetics, University of Leuven, Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Heart failure is an important cause of morbidity and mortality in individuals of all ages. The many-faceted nature of the clinical heart failure syndrome has historically frustrated attempts to develop an overarching explanative theory. However, much useful information has been gained by basic and clinical investigation, even though a comprehensive understanding of heart failure has been elusive. Heart failure is a growing problem, in both adult and pediatric populations, for which standard medical therapy, as of 2010, can have positive effects, but these are usually limited and progressively diminish with time in most patients. If we want curative or near-curative therapy that will return patients to a normal state of health at a feasible cost, much better diagnostic and therapeutic technologies need to be developed. This review addresses the vexing group of heart failure etiologies that include cardiomyopathies and other ventricular dysfunctions of various types, for which current therapy is only modestly effective. Although there are many unique aspects to heart failure in patients with pediatric and congenital heart disease, many of the innovative approaches that are being developed for the care of adults with heart failure will be applicable to heart failure in childhood.
Collapse
Affiliation(s)
- Daniel J Penny
- Section of Pediatric Cardiology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, 6621 Fannin Street, Houston, TX 77030, USA
| | | |
Collapse
|
22
|
The amiloride derivative phenamil attenuates pulmonary vascular remodeling by activating NFAT and the bone morphogenetic protein signaling pathway. Mol Cell Biol 2010; 31:517-30. [PMID: 21135135 DOI: 10.1128/mcb.00884-10] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pulmonary artery hypertension (PAH) is characterized by elevated pulmonary artery resistance and increased medial thickness due to deregulation of vascular remodeling. Inactivating mutations of the BMPRII gene, which encodes a receptor for bone morphogenetic proteins (BMPs), are identified in ∼60% of familial PAH (FPAH) and ∼30% of idiopathic PAH (IPAH) patients. It has been hypothesized that constitutive reduction in BMP signal by BMPRII mutations may cause abnormal vascular remodeling by promoting dedifferentiation of vascular smooth muscle cells (vSMCs). Here, we demonstrate that infusion of the amiloride analog phenamil during chronic-hypoxia treatment in rat attenuates development of PAH and vascular remodeling. Phenamil induces Tribbles homolog 3 (Trb3), a positive modulator of the BMP pathway that acts by stabilizing the Smad family signal transducers. Through induction of Trb3, phenamil promotes the differentiated, contractile vSMC phenotype characterized by elevated expression of contractile genes and reduced cell growth and migration. Phenamil activates the Trb3 gene transcription via activation of the calcium-calcineurin-nuclear factor of activated T cell (NFAT) pathway. These results indicate that constitutive elevation of Trb3 by phenamil is a potential therapy for IPAH and FPAH.
Collapse
|
23
|
Yang D, Lai D, Huang X, Shi X, Gao Z, Huang F, Zhou X, Geng YJ. The defects in development and apoptosis of cardiomyocytes in mice lacking the transcriptional factor Pax-8. Int J Cardiol 2010; 154:43-51. [PMID: 20851479 DOI: 10.1016/j.ijcard.2010.08.057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2009] [Revised: 05/31/2010] [Accepted: 08/19/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND Cardiac-specific deletion of ALK3 is lethal in mid-gestation with ventricular septum malformations (VSM). This study was designed to define the Pax-8's role in heart development and cardiomyocyte apoptosis. METHODS Pathologic changes in the hearts of Pax-8 or ALK3 knockout and wild type control mice were determined by light and electron microscopy. Analysis of cardiomyocyte apoptosis was performed by TUNEL. The effect of Pax-8 gene deficiency on caspase-3 activity was examined after transfecting Pax-8 siRNA into cultured myoblast cell line. RESULTS Mice with ALK3 or Pax-8 gene knockout but not wild type control animals showed the development of VSM. Increased cardiomyocyte apoptosis was found in homozygotes. Echocardiography showed that Pax-8 homozygote mice developed malfunction of the heart. Furthermore, the caspase-3 activity was significantly higher in the cells treated with Pax-8 siRNA as compared to those treated with negative control siRNA in H9C2 (2-1) cell line. CONCLUSIONS The Pax-8 gene may play a crucial role in heart development and regulating cardiocyte apoptosis. Knockout of Pax-8 may exert a similar effect on myocardial morphology and apoptosis as those seen in ALK3 knockouts. Furthermore, the ventricular septum malformations could be partially attributed to accelerated cardiomyocyte apoptosis.
Collapse
Affiliation(s)
- Deye Yang
- The First Affiliated Hospital of Wenzhou Medical College, Wenzhou 325000, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Chan MC, Hilyard AC, Wu C, Davis BN, Hill NS, Lal A, Lieberman J, Lagna G, Hata A. Molecular basis for antagonism between PDGF and the TGFbeta family of signalling pathways by control of miR-24 expression. EMBO J 2009; 29:559-73. [PMID: 20019669 DOI: 10.1038/emboj.2009.370] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 11/06/2009] [Indexed: 12/12/2022] Open
Abstract
Modulation of the vascular smooth-muscle-cell (vSMC) phenotype from a quiescent 'contractile' phenotype to a proliferative 'synthetic' phenotype has been implicated in vascular injury repair, as well as pathogenesis of vascular proliferative diseases. Both bone morphogenetic protein (BMP) and transforming growth factor-beta (TGFbeta)-signalling pathways promote a contractile phenotype, while the platelet-derived growth factor-BB (PDGF-BB)-signalling pathway promotes a switch to the synthetic phenotype. Here we show that PDGF-BB induces microRNA-24 (miR-24), which in turn leads to downregulation of Tribbles-like protein-3 (Trb3). Repression of Trb3 coincides with reduced expression of Smad proteins and decrease in BMP and TGFbeta signalling, promoting a synthetic phenotype in vSMCs. Inhibition of miR-24 by antisense oligonuclotides abrogates the downregulation of Trb3 as well as pro-synthetic activity of the PDGF-signalling pathway. Thus, this study provides a molecular basis for the antagonism between the PDGF and TGFbeta pathways, and its effect on the control of the vSMC phenotype.
Collapse
Affiliation(s)
- Mun Chun Chan
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Beppu H, Malhotra R, Beppu Y, Lepore JJ, Parmacek MS, Bloch KD. BMP type II receptor regulates positioning of outflow tract and remodeling of atrioventricular cushion during cardiogenesis. Dev Biol 2009; 331:167-75. [PMID: 19409885 PMCID: PMC2745439 DOI: 10.1016/j.ydbio.2009.04.032] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 03/20/2009] [Accepted: 04/25/2009] [Indexed: 12/20/2022]
Abstract
Signaling of bone morphogenetic protein (BMP) via type I and type II receptors is involved in multiple processes contributing to cardiogenesis. To investigate the role of the BMP type II receptor (BMPRII) in heart development, the BMPRII gene was deleted throughout the embryo during gastrulation using a Mox2-Cre transgene. BMPRII(flox/-);Mox2-Cre mice exhibited cardiac defects including double-outlet right ventricle, ventricular septal defect (VSD), atrioventricular (AV) cushion defects, and thickened valve leaflets. To characterize the tissue-specific functions of BMPRII in cardiogenesis, a series of Cre transgenes (alphaMHC-, Tie2-, Wnt1-, and SM22alpha-Cre) was employed. Interestingly, myocardial development was normal when the BMPRII gene was deleted in myocardial cells using Mox2-Cre, alphaMHC-Cre, or SM22alpha-Cre transgenes, suggesting that signaling by other BMP type II receptors may compensate for the absence of BMPRII in the myocardial cells. AV cushion defects including atrial septal defect, membranous VSD, and thickened valve leaflets were found in BMPRII(flox/-);Tie2-Cre mice. Abnormal positioning of the aorta was observed in BMPRII(flox/-);Wnt1-Cre and BMPRII(flox/-);SM22alpha-Cre mice. Taken together, these results demonstrate that endocardial BMPRII expression is required for septal formation and valvulogenesis. Moreover, mesenchymal BMPRII expression in the outflow tract cushion is required for proper positioning of the aorta.
Collapse
Affiliation(s)
- Hideyuki Beppu
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, 02129, USA.
| | | | | | | | | | | |
Collapse
|
26
|
McCulley DJ, Kang JO, Martin JF, Black BL. BMP4 is required in the anterior heart field and its derivatives for endocardial cushion remodeling, outflow tract septation, and semilunar valve development. Dev Dyn 2008; 237:3200-9. [PMID: 18924235 PMCID: PMC2728547 DOI: 10.1002/dvdy.21743] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The endocardial cushions play a critical role in septation of the four-chambered mammalian heart and in the formation of the valve leaflets that control blood flow through the heart. Within the outflow tract (OFT), both cardiac neural crest and endocardial-derived mesenchymal cells contribute to the endocardial cushions. Bone morphogenetic protein 4 (BMP4) is required for endocardial cushion development and for normal septation of the OFT. In the present study, we show that anterior heart field (AHF)-derived myocardium is an essential source of BMP4 required for normal endocardial cushion expansion and remodeling. Loss of BMP4 from the AHF in mice results in an insufficient number of cells in the developing OFT endocardial cushions, defective cushion remodeling, ventricular septal defects, persistent truncus arteriosus, and abnormal semilunar valve formation.
Collapse
Affiliation(s)
- David J. McCulley
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-2240
| | - Ji-One Kang
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-2240
| | - James F. Martin
- Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-2240
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA 94143-2240
| |
Collapse
|
27
|
Perino MG, Yamanaka S, Li J, Wobus AM, Boheler KR. Cardiomyogenic stem and progenitor cell plasticity and the dissection of cardiopoiesis. J Mol Cell Cardiol 2008; 45:475-94. [PMID: 18565538 PMCID: PMC2597345 DOI: 10.1016/j.yjmcc.2008.05.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 04/04/2008] [Accepted: 05/02/2008] [Indexed: 12/13/2022]
Abstract
Cell-based therapies hold promise of repairing an injured heart, and the description of stem and progenitor cells with cardiomyogenic potential is critical to its realization. At the vanguard of these efforts are analyses of embryonic stem cells, which clearly have the capacity to generate large numbers of cardiomyocytes in vitro. Through the use of this model system, a number of signaling mechanisms have been worked out that describes at least partially the process of cardiopoiesis. Studies on adult stem and on progenitor cells with cardiomyogenic potential are still in their infancy, and much less is known about the molecular signals that are required to induce the differentiation to cardiomyocytes. It is also unclear whether the pathways are similar or different between embryonic and adult cell-induced cardiomyogenesis, partly because of the continued controversies that surround the stem cell theory of cardiac self-renewal. Irrespective of any perceived or actual limitations, the study of stem and progenitor cells has provided important insights into the process of cardiomyogenesis, and it is likely that future research in this area will turn the promise of repairing an injured heart into a reality.
Collapse
Affiliation(s)
- Maria Grazia Perino
- From the Laboratory of Cardiovascular Sciences, National Institute on Aging, NIH, Baltimore MD 21224, U.S.A. and In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Satoshi Yamanaka
- From the Laboratory of Cardiovascular Sciences, National Institute on Aging, NIH, Baltimore MD 21224, U.S.A. and In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Jinliang Li
- From the Laboratory of Cardiovascular Sciences, National Institute on Aging, NIH, Baltimore MD 21224, U.S.A. and In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Anna M. Wobus
- From the Laboratory of Cardiovascular Sciences, National Institute on Aging, NIH, Baltimore MD 21224, U.S.A. and In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Kenneth R. Boheler
- From the Laboratory of Cardiovascular Sciences, National Institute on Aging, NIH, Baltimore MD 21224, U.S.A. and In Vitro Differentiation Group, Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| |
Collapse
|
28
|
Yuasa S, Fukuda K. Recent advances in cardiovascular regenerative medicine: the induced pluripotent stem cell era. Expert Rev Cardiovasc Ther 2008; 6:803-10. [PMID: 18570618 DOI: 10.1586/14779072.6.6.803] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Induced pluripotent stem (iPS) cells have recently been established by transfecting mouse and human fibroblasts with the transcription factors Oct3/4, Sox2, Klf4 and c-Myc, known to be expressed at high levels in embryonic stem (ES) cells. These cells have great potential in regenerative medicine as they have the capacity to differentiate into all three germ layer-derived cells and are syngeneic. The differentiation of ES cells into cardiomyocytes mimics the early processes involved in heart development. Recent studies describe the contribution of various growth factors and corresponding inhibitors to heart development during embryogenesis. Bone morphogenetic proteins, Wnt protein and Notch signals play critical roles in heart development in a context- and time-dependent manner. Consistent with ES cells, the exposure of iPS cells to such growth factors is hypothesized to augment differentiation into cardiomyocytes. The combination of iPS cells and appropriate developmental signal information has the potential for providing the foundations for future regenerative medicine.
Collapse
Affiliation(s)
- Shinsuke Yuasa
- Cardiology Division, Department of Medicine, Keio University School of Medicine, 35-Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | | |
Collapse
|
29
|
Yang D, Zhang J, Chen C, Xie M, Sperling S, Fang F, Chen B, Li X, Zhang H. BMPR IA downstream genes related to VSD. Pediatr Res 2008; 63:602-6. [PMID: 18543407 DOI: 10.1203/pdr.0b013e31813cbe9f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cardiac-specific deletion of the receptor IA of bone morphogenetic protein (BMP) (ALK3) by Cre recombinase driven under the [alpha]-MHC promoter is lethal in mid-gestation with defects in the interventricular septum [ventricular septum defect (VSD)]. Analysis of expression of the ALK3 downstream genes is important to identify the signaling pathway for interventricular septum development. The mRNA expression level of a control group was compared with that of a test group. ALK3 downstream genes were screened using polymerase chain reaction (PCR)-select cDNA subtraction and microarray. It was found that the mice with an ALK3 knockout gene produced a VSD. The expression of some genes such as platelet-activating factor acetylhydrolase (PAF) and Pax-8 was down-regulated in the test group. Pax-8 gene expression was down-regulated by 7.1 times in the test group and expressed specifically in the 11.5-d embryonic (E11.5) heart. Furthermore, the expression of the protein-tyrosine kinase of the focal adhesion kinase subfamily (PTK) and [beta] subtype protein 14-3-3 was up-regulated in the test group. PTK gene expression was up-regulated by 3.7 times in the test group. These data provided support that the ALK3 gene plays an important role during heart development. The PAF and Pax-8 genes could be important ALK3 downstream genes in the BMP signaling pathway during interventricular septum development. PTK and [beta] subtype protein 14-3-3 might be regulatory factors in this pathway.
Collapse
Affiliation(s)
- Deye Yang
- Department of Cardiology, The First Affiliated Hospital, Institute ofor Cardiovascular Biology and Gene, Wenzhou Medical College, Wenzhou 325000, PR China.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Filipczyk AA, Passier R, Rochat A, Mummery CL. Regulation of cardiomyocyte differentiation of embryonic stem cells by extracellular signalling. Cell Mol Life Sci 2007; 64:704-18. [PMID: 17380311 PMCID: PMC2778649 DOI: 10.1007/s00018-007-6523-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Investigating the signalling pathways that regulate heart development is essential if stem cells are to become an effective source of cardiomyocytes that can be used for studying cardiac physiology and pharmacology and eventually developing cell-based therapies for heart repair. Here, we briefly describe current understanding of heart development in vertebrates and review the signalling pathways thought to be involved in cardiomyogenesis in multiple species. We discuss how this might be applied to stem cells currently thought to have cardiomyogenic potential by considering the factors relevant for each differentiation step from the undifferentiated cell to nascent mesoderm, cardiac progenitors and finally a fully determined cardiomyocyte. We focus particularly on how this is being applied to human embryonic stem cells and provide recent examples from both our own work and that of others.
Collapse
Affiliation(s)
- A. A. Filipczyk
- Hubrecht Laboratory, Netherlands Institute of Developmental Biology, Utrecht, The Netherlands
| | - R. Passier
- Hubrecht Laboratory, Netherlands Institute of Developmental Biology, Utrecht, The Netherlands
| | - A. Rochat
- Hubrecht Laboratory, Netherlands Institute of Developmental Biology, Utrecht, The Netherlands
- Mouse Molecular Genetics Group, Faculté de Médecine Pitié-Salpêtriére, 105, boulevard de l’Hôpital, 75364 Paris Cedex 13, France
| | - C. L. Mummery
- Hubrecht Laboratory, Netherlands Institute of Developmental Biology, Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands and Heart Lung Center, University Medical Centre, Utrecht, The Netherlands
| |
Collapse
|
31
|
Kim KH, Antkiewicz DS, Yan L, Eliceiri KW, Heideman W, Peterson RE, Lee Y. Lrrc10 is required for early heart development and function in zebrafish. Dev Biol 2007; 308:494-506. [PMID: 17601532 PMCID: PMC2048587 DOI: 10.1016/j.ydbio.2007.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/15/2007] [Accepted: 06/06/2007] [Indexed: 11/15/2022]
Abstract
Leucine-rich Repeat Containing protein 10 (LRRC10) has recently been identified as a cardiac-specific factor in mice. However, the function of this factor remains to be elucidated. In this study, we investigated the developmental roles of Lrrc10 using zebrafish as an animal model. Knockdown of Lrrc10 in zebrafish embryos (morphants) using morpholinos caused severe cardiac morphogenic defects including a cardiac looping failure accompanied by a large pericardial edema, and embryonic lethality between day 6 and 7 post fertilization. The Lrrc10 morphants exhibited cardiac functional defects as evidenced by a decrease in ejection fraction and cardiac output. Further investigations into the underlying mechanisms of the cardiac defects revealed that the number of cardiomyocyte was reduced in the morphants. Expression of two cardiac genes was deregulated in the morphants including an increase in atrial natriuretic factor, a hallmark for cardiac hypertrophy and failure, and a decrease in cardiac myosin light chain 2, an essential protein for cardiac contractility in zebrafish. Moreover, a reduced fluorescence intensity from NADH in the morphant heart was observed in live zebrafish embryos as compared to control. Taken together, the present study demonstrates that Lrrc10 is necessary for normal cardiac development and cardiac function in zebrafish embryos, which will enhance our understanding of congenital heart defects and heart disease.
Collapse
Affiliation(s)
- Ki-Hyun Kim
- Department of Anatomy, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Dagmara S. Antkiewicz
- Molecular and Environmental Toxicology Center, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Long Yan
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, USA
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI 53706, USA
| | - Kevin W. Eliceiri
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin, Madison, WI 53706, USA
| | - Warren Heideman
- Molecular and Environmental Toxicology Center, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Richard E. Peterson
- Molecular and Environmental Toxicology Center, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Youngsook Lee
- Department of Anatomy, School of Medicine and Public Health, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
32
|
Abstract
Congenital heart diseases are the most commonly observed human birth defects and are the leading cause of infant morbidity and mortality. Accumulating evidence indicates that transforming growth factor-beta/bone morphogenetic protein signaling pathways play critical roles during cardiogenesis. Smad4 encodes the only common Smad protein in mammals, which is a critical nuclear mediator of transforming growth factor-beta/bone morphogenetic protein signaling. The aim of this work was to investigate the roles of Smad4 during heart development. To overcome the early embryonic lethality of Smad4(-/-) mice, we specifically disrupted Smad4 in the myocardium using a Cre/loxP system. We show that myocardial-specific inactivation of Smad4 caused heart failure and embryonic lethality at midgestation. Histological analysis revealed that mutant mice displayed a hypocellular myocardial wall defect, which is likely the primary cause for heart failure. Both decreased cell proliferation and increased apoptosis contributed to the myocardial wall defect in mutant mice. Data presented in this article contradict a previous report showing that Smad4 is dispensable for heart development. Our further molecular characterization showed that expression of Nmyc and its downstream targets, including cyclin D1, cyclin D2, and Id2, were downregulated in mutant embryos. Reporter analysis indicated that the transcriptional activity of the 351-bp Nmyc promoter can be positively regulated by bone morphogenetic protein stimulation and negatively regulated by transforming growth factor-beta stimulation. Chromatin immunoprecipitation analysis revealed that the Nmyc promoter can form a complex with Smad4, suggesting that Nmyc is a direct downstream target of Smad4. In conclusion, this study provides the first mouse model showing that Smad4 plays essential roles during cardiogenesis.
Collapse
Affiliation(s)
- Lanying Song
- Department of Genetics, Division of Genetic and Translational Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
33
|
Kim KH, Kim TG, Micales BK, Lyons GE, Lee Y. Dynamic expression patterns of leucine-rich repeat containing protein 10 in the heart. Dev Dyn 2007; 236:2225-34. [PMID: 17626279 PMCID: PMC2002521 DOI: 10.1002/dvdy.21225] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Leucine-rich repeat containing protein 10 (LRRC10) is a heart-specific factor whose function remains unknown. Examination of the intracellular location of the gene products is a critical step in determining the biological functions of the protein. Our expression analyses in mice indicate that LRRC10 is exclusively expressed from the precardiac region in early embryos to the adult heart. LRRC10 expression is markedly elevated upon birth, suggesting its role in the embryonic as well as adult hearts. Of interest, LRRC10 exhibits dynamic intracellular expression patterns in cardiomyocytes. Cardiomyocytes from embryos and newborns show diffuse cytoplasmic and nuclear staining of LRRC10. In contrast, striking striations are observed in adult cardiomyocytes, which are colocalized with the markers for the Z-line, sarcoplasmic reticulum (SR), and transverse (T)-tubule by double immunostaining. Further investigation by electron micrographs places LRRC10 in a diad region where the SR interacts with the T-tubule that locates along the Z-line.
Collapse
Affiliation(s)
- Ki-Hyun Kim
- Department of Anatomy, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | | | | | | | |
Collapse
|
34
|
van Bezooijen RL, Deruiter MC, Vilain N, Monteiro RM, Visser A, van der Wee-Pals L, van Munsteren CJ, Hogendoorn PCW, Aguet M, Mummery CL, Papapoulos SE, Ten Dijke P, Löwik CWGM. SOST expression is restricted to the great arteries during embryonic and neonatal cardiovascular development. Dev Dyn 2007; 236:606-12. [PMID: 17195180 DOI: 10.1002/dvdy.21054] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Spatial-temporal regulation of bone morphogenetic protein (BMP) and Wnt activity is essential for normal cardiovascular development, and altered activity of these growth factors causes maldevelopment of the cardiac outflow tract and great arteries. In the present study, we show that SOST, a Dan family member reported to antagonize BMP and Wnt activity, is expressed within the medial vessel wall of the great arteries containing smooth muscle cells. The ascending aorta, aortic arch, brachiocephalic artery, common carotids, and pulmonary trunk were all associated with SOST expressing smooth muscle cells, while the heart itself, including the valves, and more distal arteries, that is, pulmonary arteries, subclavian arteries, and descending aorta, were negative. SOST was expressed from embryonic day 15.5 up to the neonatal period. SOST expression, however, did not correspond with inhibition of Smad-dependent BMP activity or beta-catenin-dependent Wnt activity in the great arteries. Activity of both signaling pathways was already down-regulated before induction of SOST expression.
Collapse
Affiliation(s)
- Rutger L van Bezooijen
- Department of Endocrinology and Metabolic Diseases, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Wang WD, Huang CJ, Lu YF, Hsin JP, Prabhakar VR, Cheng CF, Hwang SPL. Heart-targeted overexpression of Nip3a in zebrafish embryos causes abnormal heart development and cardiac dysfunction. Biochem Biophys Res Commun 2006; 347:979-87. [PMID: 16854375 DOI: 10.1016/j.bbrc.2006.06.174] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2006] [Accepted: 06/29/2006] [Indexed: 11/30/2022]
Abstract
We transiently expressed a proapoptotic protein, Nip3a, by a heart-specific BMP4 promoter in zebrafish embryos and generated two variants of embryos with abnormal heart phenotypes (A and B). Embryos with phenotype A heart defects showed hypoplastic or elongated ventricles, elongated or enlarged atriums with no normal cardiac looping resulting a significant longer SV-BA distance, and bradycardia at 48 h post-fertilization (hpf). Embryos with phenotype B heart defects showed an enlarged fluid-filled pericardium, severe hypoplasia, non-contracting ventricles, and elongated or enlarged slowly beating atriums with no normal looping. Histological sections further revealed the absence of a proper atrioventricular boundary and no endocardial cells lining this region in both 48- and 72-hpf Nip3a-overexpressing embryos, implicating defective endocardial cushion formation. These phenotypes are reminiscent of atrioventricular canal defects in humans. In addition, induced apoptotic myocardium cells were clustered in the presumptive atrioventricular boundary as well as in the adjacent ventricle and atrium of 48- and 72-hpf Nip3a-overexpressing embryos. Nip3a expression was readily detected in 80% epiboly BMP4-Nip3a-injected embryos, and defects in heart development were observed in both the linear heart tube and subsequent chamber formation stages. These results showed that myocyte apoptosis is a universal pathogenic factor for congenital heart failure using zebrafish as a model organism.
Collapse
Affiliation(s)
- Wen-Der Wang
- Vanderbilt University, LH529, 2215 Garland Avenue, Nashville, TN 37232-0275, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Yang L, Cai CL, Lin L, Qyang Y, Chung C, Monteiro RM, Mummery CL, Fishman GI, Cogen A, Evans S. Isl1Cre reveals a common Bmp pathway in heart and limb development. Development 2006; 133:1575-85. [PMID: 16556916 PMCID: PMC5576437 DOI: 10.1242/dev.02322] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A number of human congenital disorders present with both heart and limb defects, consistent with common genetic pathways. We have recently shown that the LIM homeodomain transcription factor islet 1 (Isl1) marks a subset of cardiac progenitors. Here, we perform lineage studies with an Isl1Cre mouse line to demonstrate that Isl1 also marks a subset of limb progenitors. In both cardiac and limb progenitors, Isl1 expression is downregulated as progenitors migrate in to form either heart or limb. To investigate common heart-limb pathways in Isl1-expressing progenitors, we ablated the Type I Bmp receptor, Bmpr1a utilizing Isl1Cre/+. Analysis of consequent heart and limb phenotypes has revealed novel requirements for Bmp signaling. Additionally, we find that Bmp signaling in Isl1-expressing progenitors is required for expression of T-box transcription factors Tbx2 and Tbx3 in heart and limb. Tbx3 is required for heart and limb formation, and is mutated in ulnar-mammary syndrome. We provide evidence that the Tbx3 promoter is directly regulated by Bmp Smads in vivo.
Collapse
Affiliation(s)
- Lei Yang
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Chen-Leng Cai
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Lizhu Lin
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yibing Qyang
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, 185 Cambridge Street, MA 02114, USA
| | - Christine Chung
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Rui M. Monteiro
- Hubrecht Laboratory (Netherlands Institute for Developmental Biology), Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Christine L. Mummery
- Hubrecht Laboratory (Netherlands Institute for Developmental Biology), Utrecht, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Glenn I. Fishman
- Leon H. Charney Division of Cardiology, New York University School of Medicine, New York, NY 10016, USA
| | - Anna Cogen
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sylvia Evans
- Skaggs School of Pharmacy, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Author for correspondence ()
| |
Collapse
|
37
|
Wei H, Juhasz O, Li J, Tarasova YS, Boheler KR. Embryonic stem cells and cardiomyocyte differentiation: phenotypic and molecular analyses. J Cell Mol Med 2006; 9:804-17. [PMID: 16364192 PMCID: PMC6740270 DOI: 10.1111/j.1582-4934.2005.tb00381.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Embryonic stem (ES) cell lines, derived from the inner cell mass (ICM) of blastocyst-stage embryos, are pluripotent and have a virtually unlimited capacity for self-renewal and differentiation into all cell types of an embryoproper. Both human and mouse ES cell lines are the subject of intensive investigation for potential applications in developmental biology and medicine. ES cells from both sources differentiate in vitro into cells of ecto-, endoand meso-dermal lineages, and robust cardiomyogenic differentiation is readily observed in spontaneously differentiating ES cells when cultured under appropriate conditions. Molecular, cellular and physiologic analyses demonstrate that ES cell-derived cardiomyocytes are functionally viable and that these cell derivatives exhibit characteristics typical of heart cells in early stages of cardiac development. Because terminal heart failure is characterized by a significant loss of cardiomyocytes, the use of human ES cell-derived progeny represents one possible source for cell transplantation therapies. With these issues in mind, this review will focus on the differentiation of pluripotent embryonic stem cells into cardiomyocytes as a developmental model, and the possible use of ES cell-derived cardiomyocytes as source of donor cells.
Collapse
Affiliation(s)
- Hong Wei
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
38
|
de Velasco B, Mandal L, Mkrtchyan M, Hartenstein V. Subdivision and developmental fate of the head mesoderm in Drosophila melanogaster. Dev Genes Evol 2006; 216:39-51. [PMID: 16249873 DOI: 10.1007/s00427-005-0029-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Accepted: 09/04/2005] [Indexed: 10/25/2022]
Abstract
In this paper, we define temporal and spatial subdivisions of the embryonic head mesoderm and describe the fate of the main lineages derived from this tissue. During gastrulation, only a fraction of the head mesoderm (primary head mesoderm; PHM) invaginates as the anterior part of the ventral furrow. The PHM can be subdivided into four linearly arranged domains, based on the expression of different combinations of genetic markers (tinman, heartless, snail, serpent, mef-2, zfh-1). The anterior domain (PHMA) produces a variety of cell types, among them the neuroendocrine gland (corpus cardiacum). PHMB, forming much of the "T-bar" of the ventral furrow, migrates anteriorly and dorsally and gives rise to the dorsal pharyngeal musculature. PHMC is located behind the T-bar and forms part of the anterior endoderm, besides contributing to hemocytes. The most posterior domain, PHMD, belongs to the anterior gnathal segments and gives rise to a few somatic muscles, but also to hemocytes. The procephalic region flanking the ventral furrow also contributes to head mesoderm (secondary head mesoderm, SHM) that segregates from the surface after the ventral furrow has invaginated, indicating that gastrulation in the procephalon is much more protracted than in the trunk. We distinguish between an early SHM (eSHM) that is located on either side of the anterior endoderm and is the major source of hemocytes, including crystal cells. The eSHM is followed by the late SHM (lSHM), which consists of an anterior and posterior component (lSHMa, lSHMp). The lSHMa, flanking the stomodeum anteriorly and laterally, produces the visceral musculature of the esophagus, as well as a population of tinman-positive cells that we interpret as a rudimentary cephalic aorta ("cephalic vascular rudiment"). The lSHM contributes hemocytes, as well as the nephrocytes forming the subesophageal body, also called garland cells.
Collapse
Affiliation(s)
- Begona de Velasco
- Department of Molecular Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
39
|
Beppu H, Lei H, Bloch KD, Li E. Generation of a floxed allele of the mouse BMP type II receptor gene. Genesis 2005; 41:133-7. [PMID: 15736264 DOI: 10.1002/gene.20099] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Bone morphogenetic proteins (BMPs) regulate a wide range of cellular functions that contribute to embryonic development from mesoderm formation to organogenesis. BMP type II receptor (BMPR-II) transduces BMP signals by forming heteromeric complexes with and phosphorylating BMP type I receptors. Heterozygous germline mutations of BMPR-II gene have been identified in patients with familial and sporadic primary pulmonary hypertension, indicating that BMPR-II may contribute to the maintenance of normal pulmonary vascular structure and function. Since embryos homozygous for a null BMPR-II allele died during gastrulation, precluding further studies of BMPR-II function in organ formation and in adult tissues, we generated mice carrying a conditional mutant BMPR-II allele in which exons 4 and 5 were flanked by loxP sequences. We anticipate that studies of mice carrying a floxed BMPR-II allele and a Cre transgene (under the control of a tissue-specific promoter) will enable characterization of the role of BMPR-II in specific cell types during development and in the pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Hideyuki Beppu
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | |
Collapse
|
40
|
Morse JH. Defining the Role and Clinical Relevance of BMPR2 Mutations In Pulmonary Arterial Hypertension. ACTA ACUST UNITED AC 2005. [DOI: 10.21693/1933-088x-4.1.5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Jane H. Morse
- Professor Emerita of Clinical Medicine and Special Lecturer, Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
41
|
Monteiro RM, de Sousa Lopes SMC, Korchynskyi O, ten Dijke P, Mummery CL. Spatio-temporal activation of Smad1 and Smad5 in vivo: monitoring transcriptional activity of Smad proteins. J Cell Sci 2004; 117:4653-63. [PMID: 15331632 DOI: 10.1242/jcs.01337] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Signaling by bone morphogenetic proteins is essential for a wide variety of developmental processes. Receptor-regulated Smad proteins, Smads 1 and 5, are intracellular mediators of bone morphogenetic protein signaling. Together with Smad4, these proteins translocate to the nucleus and modulate transcription by binding to specific sequences on the promoters of target genes. We sought to map transcriptional Smad1/5 activity in development by generating embryonic stem cell lines carrying a Smad1/5-specific response element derived from the Id1 promoter coupled to beta-galactosidase or luciferase as reporters. Three independent lines (BRE-lac1, BRE-lac2 and BRE-luc) have shown the existence of an autocrine bone morphogenetic protein signaling pathway in mouse embryonic stem cells. Reporter activity was detected in chimeric embryos, suggesting sensitivity to physiological concentrations of bone morphogenetic protein. Reporter activity in embryos from transgenic mouse lines was detected in tissues where an essential role for active bone morphogenetic protein signaling via Smads 1 or 5 had been previously established. We have thus generated, for the first time, an in vivo readout for studying the role of Smad1/5-mediated transcriptional activity in development.
Collapse
Affiliation(s)
- Rui M Monteiro
- Hubrecht Laboratory, Netherlands Institute for Developmental Biology, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | | | | | | | | |
Collapse
|
42
|
Abstract
Cardiovascular calcification is a common consequence of aging, diabetes, hypercholesterolemia, mechanically abnormal valve function, and chronic renal insufficiency. Although vascular calcification may appear to be a uniform response to vascular insult, it is a heterogenous disorder, with overlapping yet distinct mechanisms of initiation and progression. A minimum of four histoanatomic variants-atherosclerotic (fibrotic) calcification, cardiac valve calcification, medial artery calcification, and vascular calciphylaxis-arise in response to metabolic, mechanical, infectious, and inflammatory injuries. Common to the first three variants is a variable degree of vascular infiltration by T cells and macrophages. Once thought benign, the deleterious clinical consequences of calcific vasculopathy are now becoming clear; stroke, amputation, ischemic heart disease, and increased mortality are portended by the anatomy and extent of calcific vasculopathy. Along with dystrophic calcium deposition in dying cells and lipoprotein deposits, active endochondral and intramembranous (nonendochondral) ossification processes contribute to vascular calcium load. Thus vascular calcification is subject to regulation by osteotropic hormones and skeletal morphogens in addition to key inhibitors of passive tissue mineralization. In response to oxidized lipids, inflammation, and mechanical injury, the microvascular smooth muscle cell becomes activated. Orthotopically, proliferating stromal myofibroblasts provide osteoprogenitors for skeletal growth and fracture repair; however, in valves and arteries, vascular myofibroblasts contribute to cardiovascular ossification. Current data suggest that paracrine signals are provided by bone morphogenetic protein-2, Wnts, parathyroid hormone-related polypeptide, osteopontin, osteoprotegerin, and matrix Gla protein, all entrained to endocrine, metabolic, inflammatory, and mechanical cues. In end-stage renal disease, a "perfect storm" of vascular calcification often occurs, with hyperglycemia, hyperphosphatemia, hypercholesterolemia, hypertension, parathyroid hormone resistance, and iatrogenic calcitriol excess contributing to severe calcific vasculopathy. This brief review recounts emerging themes in the pathobiology of vascular calcification and highlights some fundamental deficiencies in our understanding of vascular endocrinology and metabolism that are immediately relevant to human health and health care.
Collapse
Affiliation(s)
- Radhika Vattikuti
- Washington Univ. School of Medicine, Dept. of Internal Medicine, Division of Bone and Mineral Diseases, Campus Box 8301, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | |
Collapse
|
43
|
Chen H, Shi S, Acosta L, Li W, Lu J, Bao S, Chen Z, Yang Z, Schneider MD, Chien KR, Conway SJ, Yoder MC, Haneline LS, Franco D, Shou W. BMP10 is essential for maintaining cardiac growth during murine cardiogenesis. Development 2004; 131:2219-31. [PMID: 15073151 PMCID: PMC2628765 DOI: 10.1242/dev.01094] [Citation(s) in RCA: 388] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
During cardiogenesis, perturbation of a key transition at mid-gestation from cardiac patterning to cardiac growth and chamber maturation often leads to diverse types of congenital heart disease, such as ventricular septal defect (VSD), myocardium noncompaction, and ventricular hypertrabeculation. This transition, which occurs at embryonic day (E) 9.0-9.5 in murine embryos and E24-28 in human embryos, is crucial for the developing heart to maintain normal cardiac growth and function in response to an increasing hemodynamic load. Although, ventricular trabeculation and compaction are key morphogenetic events associated with this transition, the molecular and cellular mechanisms are currently unclear. Initially, cardiac restricted cytokine bone morphogenetic protein 10 (BMP10) was identified as being upregulated in hypertrabeculated hearts from mutant embryos deficient in FK506 binding protein 12 (FKBP12). To determine the biological function of BMP10 during cardiac development, we generated BMP10-deficient mice. Here we describe an essential role of BMP10 in regulating cardiac growth and chamber maturation. BMP10 null mice display ectopic and elevated expression of p57(kip2) and a dramatic reduction in proliferative activity in cardiomyocytes at E9.0-E9.5. BMP10 is also required for maintaining normal expression levels of several key cardiogenic factors (e.g. NKX2.5 and MEF2C) in the developing myocardium at mid-gestation. Furthermore, BMP10-conditioned medium is able to rescue BMP10-deficient hearts in culture. Our data suggest an important pathway that involves a genetic interaction between BMP10, cell cycle regulatory proteins and several major cardiac transcription factors in orchestrating this transition in cardiogenesis at mid-gestation. This may provide an underlying mechanism for understanding the pathogenesis of both structural and functional congenital heart defects.
Collapse
Affiliation(s)
- Hanying Chen
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Brown CO, Chi X, Garcia-Gras E, Shirai M, Feng XH, Schwartz RJ. The cardiac determination factor, Nkx2-5, is activated by mutual cofactors GATA-4 and Smad1/4 via a novel upstream enhancer. J Biol Chem 2003; 279:10659-69. [PMID: 14662776 DOI: 10.1074/jbc.m301648200] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mammalian homologue of Drosophila tinman, Nkx2-5, plays an early role in regulating cardiac genes and morphogenesis. Bone morphogenetic proteins (BMPs), members of the transforming growth factor (TGF)-beta family of signaling molecules, are involved in numerous developmental processes. BMP signaling is crucial in the regulation of Nkx2-5 expression and specification of the cardiac lineage. Constitutively active BMP type I receptor or the downstream pathway components and DNA-binding transcription factors, Smad1/4 directly activated Nkx2-5 gene transcription. We identified and characterized a novel upstream Nkx2-5 enhancer, composed of clustered repeats of Smad and GATA DNA binding sites. This composite Nkx2-5 enhancer was a direct target of BMP signaling via cooperative interactions between the downstream transducers Smad1/4 and GATA-4. In mammalian two hybrid assays, Smad factors recruited the hybrid gene GATA4-VP16 to strongly drive transcription of a reporter gene containing multimerized Smad binding sites These cofactors interacted through the second zinc finger and adjacent basic domain of GATA-4 and the N-terminal domain of Smads. Smad4 and GATA4 were also found to bind in vivo with the Nkx2-5 composite enhancer, as revealed by chromatin immunoprecipitation analysis of differentiated P19 cells. Finally, transgenic mice containing the Smad/GATA composite enhancer recapitulated early murine Nkx2-5 cardiac expression and deletion of this enhancer within a 10-kb transgene pBS-Nkx2-5 LacZ significantly reduced expression in the cardiac crescent. Thus, integration of GATA transcription factors with BMP signaling, through co-association with Smads factors, may initiate early Nkx2-5 expression; suggesting a vital role for the combination of these factors in the specification of cardiac progenitors.
Collapse
Affiliation(s)
- Carl O Brown
- Department of Molecular and Cellular Biology, The Center for Cardiovascular Development, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
45
|
Azhar M, Schultz JEJ, Grupp I, Dorn GW, Meneton P, Molin DGM, Gittenberger-de Groot AC, Doetschman T. Transforming growth factor beta in cardiovascular development and function. Cytokine Growth Factor Rev 2003; 14:391-407. [PMID: 12948523 PMCID: PMC3855389 DOI: 10.1016/s1359-6101(03)00044-3] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Transforming growth factor betas (TGFbetas) are pleiotropic cytokines involved in many biological processes. Genetic engineering and tissue explanation studies have revealed specific non-overlapping roles for TGFbeta ligands and their signaling molecules in development and in normal function of the cardiovascular system in the adult. In the embryo, TGFbetas appear to be involved in epithelial-mesenchymal transformations (EMT) during endocardial cushion formation, and in epicardial epithelial-mesenchymal transformations essential for coronary vasculature, ventricular myocardial development and compaction. In the adult, TGFbetas are involved in cardiac hypertrophy, vascular remodeling and regulation of the renal renin-angiotensin system. The evidence for TGFbeta activities during cardiovascular development and physiologic function will be given and areas which need further investigation will be discussed.
Collapse
Affiliation(s)
- Mohamad Azhar
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Jiao K, Kulessa H, Tompkins K, Zhou Y, Batts L, Baldwin HS, Hogan BLM. An essential role of Bmp4 in the atrioventricular septation of the mouse heart. Genes Dev 2003; 17:2362-7. [PMID: 12975322 PMCID: PMC218073 DOI: 10.1101/gad.1124803] [Citation(s) in RCA: 300] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Proper septation and valvulogenesis during cardiogenesis depend on interactions between the myocardium and the endocardium. By combining use of a hypomorphic Bone morphogenetic protein 4 (Bmp4) allele with conditional gene inactivation, we here identify Bmp4 as a signal from the myocardium directly mediating atrioventricular septation. Defects in this process cause one of the most common human congenital heart abnormalities, atrioventricular canal defect (AVCD). The spectrum of defects obtained through altering Bmp4 expression in the myocardium recapitulates the range of AVCDs diagnosed in patients, thus providing a useful genetic model with AVCD as the primary defect.
Collapse
Affiliation(s)
- Kai Jiao
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Affiliation(s)
- Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, USA.
| | | |
Collapse
|
48
|
Latinkić BV, Kotecha S, Mohun TJ. Induction of cardiomyocytes by GATA4 in Xenopus ectodermal explants. Development 2003; 130:3865-76. [PMID: 12835401 DOI: 10.1242/dev.00599] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The earliest step in heart formation in vertebrates occurs during gastrulation, when cardiac tissue is specified. Dorsoanterior endoderm is thought to provide a signal that induces adjacent mesodermal cells to adopt a cardiac fate. However, the nature of this signalling and the precise role of endoderm are unknown because of the close proximity and interdependence of mesoderm and endoderm during gastrulation. To better define the molecular events that underlie cardiac induction, we have sought to develop a simple means of inducing cardiac tissue. We show that the transcription factor GATA4, which has been implicated in regulating cardiac gene expression, is sufficient to induce cardiac differentiation in Xenopus embryonic ectoderm (animal pole) explants, frequently resulting in beating tissue. Lineage labelling experiments demonstrate that GATA4 can trigger cardiac differentiation not only in cells in which it is present, but also in neighbouring cells. Surprisingly, cardiac differentiation can occur without any stable differentiation of anterior endoderm and is in fact enhanced under conditions in which endoderm formation is inhibited. Remarkably, cardiac tissue is formed even when GATA4 activity is delayed until long after explants have commenced differentiation into epidermal tissue. These findings provide a simple assay system for cardiac induction that may allow elucidation of pathways leading to cardiac differentiation. Better knowledge of the pathways governing this process may help develop procedures for efficient generation of cardiomyocytes from pluripotent stem cells.
Collapse
Affiliation(s)
- Branko V Latinkić
- Division of Developmental Biology, National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | | | | |
Collapse
|
49
|
Nakamura T, Schneider MD. The way to a human's heart is through the stomach: visceral endoderm-like cells drive human embryonic stem cells to a cardiac fate. Circulation 2003; 107:2638-9. [PMID: 12782614 DOI: 10.1161/01.cir.0000074240.87740.be] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|