1
|
Yamaguchi H, Tran LT, Bi J, Urayama A, Yutzey KE, Mishina Y, Komatsu Y. Enhanced BMP signaling in cranial neural crest cells induces aberrant chondrogenesis by upregulating Tbx20 expression during craniofacial development. Biochem Biophys Res Commun 2025; 765:151834. [PMID: 40273622 DOI: 10.1016/j.bbrc.2025.151834] [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: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Bone morphogenetic proteins (BMPs) are critical for craniofacial development. We previously reported that cranial neural crest cell (CNCC)-specific enhanced BMP signaling through the ALK2 receptor causes ectopic cartilage formation in the face during mouse embryonic development. However, the downstream effectors triggering this ectopic chondrogenesis remain unclear. Here, we investigated the targets of BMP signaling responsible for ectopic cartilage formation. A microarray analysis using CNC-derived ectomesenchymal cells from the first branchial arches identified T-box transcription factor 20 (Tbx20) as the top candidate gene in CNC-specific gain-of-function ALK2 mouse embryos. This prompted us to hypothesize that enhanced BMP signaling increases Tbx20 expression, which triggers ectopic cartilage formation in the craniofacial region. To examine whether Tbx20 overexpression in CNCCs alters craniofacial development, we utilized a Cre-LoxP system to augment Tbx20 expression in a neural crest-specific manner in mice. CNCC-specific overexpression of Tbx20 led to neonatal death with severe craniofacial defects, such as orofacial clefts and exencephaly. Interestingly, aberrant chondrogenesis was observed in the posterior frontal (PF) suture, a structure derived from CNCCs, suggesting that augmented Tbx20 expression triggers ectopic cartilage formation in the PF suture. This study reveals that enhanced BMP-Tbx20 signaling in CNCCs causes aberrant chondrogenesis in the PF suture during craniofacial development.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Lauren T Tran
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiarui Bi
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Periodontics & Dental Hygiene, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX, USA
| | - Akihiko Urayama
- Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Katherine E Yutzey
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA; The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics and Epigenetics, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.
| |
Collapse
|
2
|
Li YJ, Zou S, Bian YZ, Liu XY, Yang CX, Li L, Qiu XB, Xu YJ, Yang YQ, Huang RT. Chromosomal Location and Identification of TBX20 as a New Gene Responsible for Familial Bicuspid Aortic Valve. Diagnostics (Basel) 2025; 15:600. [PMID: 40075846 PMCID: PMC11898536 DOI: 10.3390/diagnostics15050600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Congenital bicuspid aortic valve (BAV) signifies the most frequent category of congenital cardiovascular anomaly globally, occurring in approximately 0.5-2% of the general population worldwide. BAV is a major cause of thoracic aortopathy, encompassing aortic stenosis, aortic root dilation with regurgitation, aortic dissection, and aortic aneurysms, consequently leading to substantial late-onset morbidity and mortality. Accumulating evidence convincingly demonstrates the strong genetic basis underpinning BAV, though the inheritable reasons responsible for BAV in most patients remain largely obscure. Methods: A genome-wide genotyping with 400 polymorphic genetic markers followed by linkage analysis, haplotype assay, and sequencing analysis of candidate genes was conducted in a 4-generation BAV kindred of 47 individuals. Biochemical assays were performed to evaluate the functional effect of the identified mutation on TBX20. Results: A novel BAV-causative locus was mapped to chromosome 7p14. A sequencing assay of the genes within the mapped chromosomal region (locus) unveiled that only the c.656T>G (p.Ile219Arg) variation of TBX20 was in co-segregation with BAV in the entire pedigree. The missense mutation was not uncovered in 322 healthy persons employed as control individuals. Functional deciphers revealed that the mutation significantly decreased the transcriptional activation of the representative target gene ANP and the binding ability to the ANP promoter and impaired the intranuclear distribution of TBX20. Conclusions: This investigation maps a new genetic locus (chromosome 7p14) linked to BAV and uncovers TBX20 as a novel causative gene for familial BAV, adding more insight into the mechanisms underlying BAV and providing a molecular target for the individualized management of BAV.
Collapse
Affiliation(s)
- Yan-Jie Li
- Department of Cardiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.-J.L.); (X.-B.Q.)
| | - Su Zou
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China; (S.Z.); (Y.-Z.B.); (C.-X.Y.); (Y.-J.X.)
| | - Yi-Zhe Bian
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China; (S.Z.); (Y.-Z.B.); (C.-X.Y.); (Y.-J.X.)
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China;
| | - Chen-Xi Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China; (S.Z.); (Y.-Z.B.); (C.-X.Y.); (Y.-J.X.)
| | - Li Li
- Key Laboratory of Arrhythmias, Ministry of Education of China, School of Medicine, Tongji University, Shanghai 200092, China;
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China; (Y.-J.L.); (X.-B.Q.)
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China; (S.Z.); (Y.-Z.B.); (C.-X.Y.); (Y.-J.X.)
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China; (S.Z.); (Y.-Z.B.); (C.-X.Y.); (Y.-J.X.)
- Cardiovascular Research Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
- Central Laboratory, Shanghai Fifth People’s Hospital, Fudan University, Shanghai 200240, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
3
|
Javanshir E, Ebrahimi ZJ, Mirzohreh ST, Ghaffari S, Banisefid E, Alamdari NM, Roshanravan N. Disparity of gene expression in coronary artery disease: insights from MEIS1, HIRA, and Myocardin. Mol Biol Rep 2024; 51:712. [PMID: 38824221 DOI: 10.1007/s11033-024-09657-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.
Collapse
Affiliation(s)
- Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Banisefid
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Roshanravan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Kimyon AS, Çetinkaya A, Hallıoğlu Kılınç O, Aras N. The evaluation of the SMAD1 rs1016792 polymorphism and gene expression on pulmonary hypertension due to congenital heart disease in children: a preliminary study. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2024; 43:1302-1315. [PMID: 38410024 DOI: 10.1080/15257770.2024.2322109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 02/28/2024]
Abstract
Smad Family Member (SMAD), a protein family responsible for transducing the signal induced by TGF-β into the nucleus, is thought to play a role in the pathology of many heart diseases. Therefore, we aimed to evaluate the influence of the SMAD1 rs1016792 polymorphism and gene expression on pulmonary arterial hypertension (PAH) due to congenital heart disease (CHD) in children. A total of 90 children, 45 of whom were PAH-CHD children and 45 healthy children, were included in the study. Patients were selected from those who were diagnosed and followed in the Department of Pediatric Cardiology.The SMAD1 rs1016792 genotyping and expression analysis was performed using a real-time polymerase chain reaction (RT-PCR)-based system. It was determined that the left ventricular end-diastolic diameter (LVEDD) value was lower in the patient group than in the control group, while the pulmonary artery pressure (PAP) value was higher in the patient group than in the control group. When the SMAD1 gene expression level was examined, a statistically significant difference was found between the patient and control groups. Patients had decreased SMAD1 expression compared to controls (p˂0.001). We found no significant difference between the patient and control groups in terms of SMAD1 rs1016792 genotype distribution or allele frequency (p > 0.05). There was no difference between genotype distribution and SMAD1 expression levels in the groups. In this study, we showed for the first time that SMAD1 expression is decreased in children with PAH-CHD. These results will be a preliminary step toward understanding the role of SMAD1 in the etiopathogenesis of CHD.
Collapse
Affiliation(s)
- Adnan Selim Kimyon
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Ayşegül Çetinkaya
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | | | - Nurcan Aras
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
5
|
Gao X, Pang S, Ding L, Yan H, Cui Y, Yan B. Genetic and functional variants of the TBX20 gene promoter in dilated cardiomyopathy. Mol Genet Genomic Med 2024; 12:e2355. [PMID: 38284443 PMCID: PMC10795084 DOI: 10.1002/mgg3.2355] [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: 01/24/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a major cause of heart failure and sudden cardiac death. As DCM is a genetically heterogeneous disease, genetic variants of cardiac transcription factor genes may play an important role. Transcription factor TBX20, an indispensable factor in normal heart development, is involved in the regulation of cardiac structure and function. Although the TBX20 gene is associated with the occurrence and development of DCM, the influence of genetic variants of the TBX20 gene promoter region on DCM has not been reported. METHODS We conducted a case-control study consisting of 107 DCM patients and 210 healthy controls. Genetic variants within TBX20 gene promoter region were identified using sequencing techniques and were functionally analyzed by dual-luciferase reporting assay. Electrophoretic mobility shift assay (EMSA) was used to investigate DNA-protein interactions. RESULTS In this study cohort (n = 317), we identified eight variants within TBX20 gene promoter. One novel DNA sequence variants (DSV) (g.4275G>T) and four single-nucleotide polymorphisms (SNPs) [g.4169G>A (rs1263874255), g.4949C>T (rs1191745927), g.5114G>A (rs112076877), g.5252C>T (rs1356932911)] were identified in DCM patients, but in none of controls. Among them, the DSV (g.4275G>T) and three SNPs [g.4949C>T (rs1191745927), g.5114G>A (rs112076877) and g.5252C>T (rs1356932911)] significantly altered the transcription activity of TBX20 gene promoter by dual-luciferase reporting assay (p < 0.05). Further, EMSA assay indicated that the DSV (g.4275G>T) and three SNPs [g.4949C>T (rs1191745927), g.5114G>A (rs112076877) and g.5252C>T (rs1356932911)] affected the binding of transcription factors. CONCLUSIONS These data indicate that the DSV (g.4275G>T) and three SNPs [g.4949C>T (rs1191745927), g.5114G>A (rs112076877) and g.5252C>T (rs1356932911)] increase transcription activity of TBX20 gene promoter in both HEK-293 and neonatal rat cardiomyocytes (NRCMs) cell lines by affecting the binding of transcription factors. But the mechanism remains to be verified in vivo.
Collapse
Affiliation(s)
- Xue Gao
- Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Shuchao Pang
- The Center for Molecular Genetics of Cardiovascular DiseasesAffiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
- Shandong Provincial Sino‐US Cooperation Research Center for Translational MedicineAffiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
| | - Liangcai Ding
- Center for Molecular MedicineYanzhou People's Hospital, Jining Medical UniversityJiningShandongChina
| | - Han Yan
- Center for Molecular MedicineYanzhou People's Hospital, Jining Medical UniversityJiningShandongChina
| | - Yinghua Cui
- Division of CardiologyAffiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
| | - Bo Yan
- The Center for Molecular Genetics of Cardiovascular DiseasesAffiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
- Shandong Provincial Sino‐US Cooperation Research Center for Translational MedicineAffiliated Hospital of Jining Medical University, Jining Medical UniversityJiningShandongChina
- Center for Molecular MedicineYanzhou People's Hospital, Jining Medical UniversityJiningShandongChina
- Institute of Precision MedicineJining Medical UniversityJiningShandongChina
| |
Collapse
|
6
|
Wang Z, Qiao XH, Xu YJ, Liu XY, Huang RT, Xue S, Qiu HY, Yang YQ. SMAD1 Loss-of-Function Variant Responsible for Congenital Heart Disease. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9916325. [PMID: 35281600 PMCID: PMC8913148 DOI: 10.1155/2022/9916325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/16/2022] [Indexed: 12/22/2022]
Abstract
As the most common form of developmental malformation affecting the heart and endothoracic great vessels, congenital heart disease (CHD) confers substantial morbidity and mortality as well as socioeconomic burden on humans globally. Aggregating convincing evidence highlights the genetic origin of CHD, and damaging variations in over 100 genes have been implicated with CHD. Nevertheless, the genetic basis underpinning CHD remains largely elusive. In this study, via whole-exosome sequencing analysis of a four-generation family inflicted with autosomal-dominant CHD, a heterozygous SMAD1 variation, NM_005900.3: c.264C > A; p.(Tyr88∗), was detected and validated by Sanger sequencing analysis to be in cosegregation with CHD in the whole family. The truncating variation was not observed in 362 unrelated healthy volunteers employed as control persons. Dual-luciferase reporter gene assay in cultured COS7 cells demonstrated that Tyr88∗-mutant SMAD1 failed to transactivate the genes TBX20 and NKX2.5, two already well-established CHD-causative genes. Additionally, the variation nullified the synergistic transcriptional activation between SMAD1 and MYOCD, another recognized CHD-causative gene. These data indicate SMAD1 as a new gene responsible for CHD, which provides new insight into the genetic mechanism underlying CHD, suggesting certain significance for genetic risk assessment and precise antenatal prevention of the family members inflicted with CHD.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Xiao-Hui Qiao
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Ying-Jia Xu
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Ri-Tai Huang
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Song Xue
- Department of Cardiovascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hai-Yan Qiu
- Department of Pediatric Internal Medicine, Ningbo Women & Children's Hospital, Ningbo 315031, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Cardiovascular Research Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
- Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, Shanghai 200240, China
| |
Collapse
|
7
|
Yang L, Yang Y, Liu X, Chen Y, Chen Y, Lin Y, Sun Y, Shen B. CHDGKB: a knowledgebase for systematic understanding of genetic variations associated with non-syndromic congenital heart disease. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2021; 2020:5865522. [PMID: 32608479 PMCID: PMC7327432 DOI: 10.1093/database/baaa048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 02/07/2023]
Abstract
Congenital heart disease (CHD) is one of the most common birth defects, with complex genetic and environmental etiologies. The reports of genetic variation associated with CHD have increased dramatically in recent years due to the revolutionary development of molecular technology. However, CHD is a heterogeneous disease, and its genetic origins remain inconclusive in most patients. Here we present a database of genetic variations for non-syndromic CHD (NS-CHD). By manually literature extraction and analyses, 5345 NS-CHD-associated genetic variations were collected, curated and stored in the public online database. The objective of our database is to provide the most comprehensive updates on NS-CHD genetic research and to aid systematic analyses of pathogenesis of NS-CHD in molecular level and the correlation between NS-CHD genotypes and phenotypes. Database URL: http://www.sysbio.org.cn/CHDGKB/.
Collapse
Affiliation(s)
- Lan Yang
- Center for Systems Biology, Soochow University, Suzhou 215006, China.,Center of Prenatal Diagnosis, Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Yang Yang
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Xingyun Liu
- Center for Systems Biology, Soochow University, Suzhou 215006, China.,Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongquan Chen
- School of Computer Science and Technology, Soochow University, Suzhou 215006, China
| | - Yalan Chen
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Yuxin Lin
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| | - Yan Sun
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bairong Shen
- Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Chen Y, Xiao D, Zhang L, Cai CL, Li BY, Liu Y. The Role of Tbx20 in Cardiovascular Development and Function. Front Cell Dev Biol 2021; 9:638542. [PMID: 33585493 PMCID: PMC7876368 DOI: 10.3389/fcell.2021.638542] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/08/2021] [Indexed: 01/05/2023] Open
Abstract
Tbx20 is a member of the Tbx1 subfamily of T-box-containing genes and is known to play a variety of fundamental roles in cardiovascular development and homeostasis as well as cardiac remodeling in response to pathophysiological stresses. Mutations in TBX20 are widely associated with the complex spectrum of congenital heart defects (CHDs) in humans, which includes defects in chamber septation, chamber growth, and valvulogenesis. In addition, genetic variants of TBX20 have been found to be associated with dilated cardiomyopathy and heart arrhythmia. This broad spectrum of cardiac morphogenetic and functional defects is likely due to its broad expression pattern in multiple cardiogenic cell lineages and its critical regulation of transcriptional networks during cardiac development. In this review, we summarize recent findings in our general understanding of the role of Tbx20 in regulating several important aspects of cardiac development and homeostasis and heart function.
Collapse
Affiliation(s)
- Yuwen Chen
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.,Cardiovascular Developmental Biology Program, Herman B Wells Center for Pediatric Research, Indianapolis, IN, United States
| | - Deyong Xiao
- Cardiovascular Developmental Biology Program, Herman B Wells Center for Pediatric Research, Indianapolis, IN, United States
| | - Lu Zhang
- Cardiovascular Developmental Biology Program, Herman B Wells Center for Pediatric Research, Indianapolis, IN, United States
| | - Chen-Leng Cai
- Cardiovascular Developmental Biology Program, Herman B Wells Center for Pediatric Research, Indianapolis, IN, United States
| | - Bai-Yan Li
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ying Liu
- Cardiovascular Developmental Biology Program, Herman B Wells Center for Pediatric Research, Indianapolis, IN, United States
| |
Collapse
|
9
|
Hoppler S, Conlon FL. Xenopus: Experimental Access to Cardiovascular Development, Regeneration Discovery, and Cardiovascular Heart-Defect Modeling. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037200. [PMID: 31767648 DOI: 10.1101/cshperspect.a037200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Xenopus has been used to study a wide array of developmental processes, benefiting from vast quantities of relatively large, externally developing eggs. Xenopus is particularly amenable to examining the cardiac system because many of the developmental processes and genes involved in cardiac specification, differentiation, and growth are conserved between Xenopus and human and have been characterized in detail. Furthermore, compared with other higher vertebrate models, Xenopus embryos can survive longer without a properly functioning heart or circulatory system, enabling investigation of later consequences of early embryological manipulations. This biology is complemented by experimental technology, such as embryonic explants to study the heart, microinjection of overexpression constructs, and, most recently, the generation of genetic mutations through gene-editing technologies. Recent investigations highlight Xenopus as a powerful experimental system for studying injury/repair and regeneration and for congenital heart disease (CHD) modeling, which reinforces why this model system remains ideal for studying heart development.
Collapse
Affiliation(s)
- Stefan Hoppler
- Aberdeen Cardiovascular & Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, Scotland, United Kingdom
| | - Frank L Conlon
- Department of Biology and Genetics, University of North Carolina McAllister Heart Institute, Chapel Hill, North Carolina 27599-3280, USA
| |
Collapse
|
10
|
Mollazadeh S, Fazly Bazzaz BS, Neshati V, de Vries AAF, Naderi-Meshkin H, Mojarad M, Neshati Z, Kerachian MA. T- Box20 inhibits osteogenic differentiation in adipose-derived human mesenchymal stem cells: the role of T- Box20 on osteogenesis. ACTA ACUST UNITED AC 2019; 26:8. [PMID: 31548928 PMCID: PMC6751895 DOI: 10.1186/s40709-019-0099-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
Abstract
Background Skeletal development and its cellular function are regulated by various transcription factors. The T-box (Tbx) family of transcription factors have critical roles in cellular differentiation as well as heart and limbs organogenesis. These factors possess activator and/or repressor domains to modify the expression of target genes. Despite the obvious effects of Tbx20 on heart development, its impact on bone development is still unknown. Methods To investigate the consequence by forced Tbx20 expression in the osteogenic differentiation of human mesenchymal stem cells derived from adipose tissue (Ad-MSCs), these cells were transduced with a bicistronic lentiviral vector encoding Tbx20 and an enhanced green fluorescent protein. Results Tbx20 gene delivery system suppressed the osteogenic differentiation of Ad-MSCs, as indicated by reduction in alkaline phosphatase activity and Alizarin Red S staining. Consistently, reverse transcription-polymerase chain reaction analyses showed that Tbx20 gain-of-function reduced the expression levels of osteoblast marker genes in osteo-inductive Ad-MSCs cultures. Accordingly, Tbx20 negatively affected osteogenesis through modulating expression of key factors involved in this process. Conclusion The present study suggests that Tbx20 could inhibit osteogenic differentiation in adipose-derived human mesenchymal stem cells.
Collapse
Affiliation(s)
- Samaneh Mollazadeh
- 1Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.,2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Sedigheh Fazly Bazzaz
- 2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,3Department of Food and Drug Control, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,4School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vajiheh Neshati
- 2Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Antoine A F de Vries
- 5Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hojjat Naderi-Meshkin
- 6Stem Cell and Regenerative Medicine Research Group, Academic Center for Education, Culture Research (ACECR), Khorasan Razavi Branch, Mashhad, Iran
| | - Majid Mojarad
- 7Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,8Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zeinab Neshati
- 9Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Mohammad Amin Kerachian
- 7Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,8Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Nenni MJ, Fisher ME, James-Zorn C, Pells TJ, Ponferrada V, Chu S, Fortriede JD, Burns KA, Wang Y, Lotay VS, Wang DZ, Segerdell E, Chaturvedi P, Karimi K, Vize PD, Zorn AM. Xenbase: Facilitating the Use of Xenopus to Model Human Disease. Front Physiol 2019; 10:154. [PMID: 30863320 PMCID: PMC6399412 DOI: 10.3389/fphys.2019.00154] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/08/2019] [Indexed: 01/02/2023] Open
Abstract
At a fundamental level most genes, signaling pathways, biological functions and organ systems are highly conserved between man and all vertebrate species. Leveraging this conservation, researchers are increasingly using the experimental advantages of the amphibian Xenopus to model human disease. The online Xenopus resource, Xenbase, enables human disease modeling by curating the Xenopus literature published in PubMed and integrating these Xenopus data with orthologous human genes, anatomy, and more recently with links to the Online Mendelian Inheritance in Man resource (OMIM) and the Human Disease Ontology (DO). Here we review how Xenbase supports disease modeling and report on a meta-analysis of the published Xenopus research providing an overview of the different types of diseases being modeled in Xenopus and the variety of experimental approaches being used. Text mining of over 50,000 Xenopus research articles imported into Xenbase from PubMed identified approximately 1,000 putative disease- modeling articles. These articles were manually assessed and annotated with disease ontologies, which were then used to classify papers based on disease type. We found that Xenopus is being used to study a diverse array of disease with three main experimental approaches: cell-free egg extracts to study fundamental aspects of cellular and molecular biology, oocytes to study ion transport and channel physiology and embryo experiments focused on congenital diseases. We integrated these data into Xenbase Disease Pages to allow easy navigation to disease information on external databases. Results of this analysis will equip Xenopus researchers with a suite of experimental approaches available to model or dissect a pathological process. Ideally clinicians and basic researchers will use this information to foster collaborations necessary to interrogate the development and treatment of human diseases.
Collapse
Affiliation(s)
- Mardi J Nenni
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Malcolm E Fisher
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Christina James-Zorn
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Troy J Pells
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Virgilio Ponferrada
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Stanley Chu
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Joshua D Fortriede
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Kevin A Burns
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Ying Wang
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Vaneet S Lotay
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Dong Zhou Wang
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Erik Segerdell
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, United States
| | - Praneet Chaturvedi
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| | - Kamran Karimi
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Peter D Vize
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - Aaron M Zorn
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH, United States
| |
Collapse
|
12
|
Duncan AR, Khokha MK. Xenopus as a model organism for birth defects-Congenital heart disease and heterotaxy. Semin Cell Dev Biol 2016; 51:73-9. [PMID: 26910255 PMCID: PMC4809202 DOI: 10.1016/j.semcdb.2016.02.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 02/15/2016] [Indexed: 02/06/2023]
Abstract
Congenital heart disease is the leading cause of birth defects, affecting 9 out of 1000 newborns each year. A particularly severe form of congenital heart disease is heterotaxy, a disorder of left-right development. Despite aggressive surgical management, patients with heterotaxy have poor survival rates and severe morbidity due to their complex congenital heart disease. Recent genetic analysis of affected patients has found novel candidate genes for heterotaxy although their underlying mechanisms remain unknown. In this review, we discuss the importance and challenges of birth defects research including high locus heterogeneity and few second alleles that make defining disease causality difficult. A powerful strategy moving forward is to analyze these candidate genes in a high-throughput human disease model. Xenopus is ideal for these studies. We present multiple examples demonstrating the power of Xenopus in discovering new biology from the analysis of candidate heterotaxy genes such as GALNT11, NEK2 and BCOR. These genes have diverse roles in embryos and have led to a greater understanding of complex signaling pathways and basic developmental biology. It is our hope that the mechanistic analysis of these candidate genes in Xenopus enabled by next generation sequencing of patients will provide clinicians with a greater understanding of patient pathophysiology allowing more precise and personalized medicine, to help patients more effectively in the future.
Collapse
Affiliation(s)
- Anna R Duncan
- Department of Pediatrics, Children's Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, United States
| | - Mustafa K Khokha
- Department of Pediatrics and Genetics, Yale University School of Medicine, 333 Cedar St, New Haven, CT 06520, United States.
| |
Collapse
|
13
|
Wu CC, Kruse F, Vasudevarao MD, Junker JP, Zebrowski DC, Fischer K, Noël ES, Grün D, Berezikov E, Engel FB, van Oudenaarden A, Weidinger G, Bakkers J. Spatially Resolved Genome-wide Transcriptional Profiling Identifies BMP Signaling as Essential Regulator of Zebrafish Cardiomyocyte Regeneration. Dev Cell 2015; 36:36-49. [PMID: 26748692 DOI: 10.1016/j.devcel.2015.12.010] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 10/14/2015] [Accepted: 11/20/2015] [Indexed: 12/21/2022]
Abstract
In contrast to mammals, zebrafish regenerate heart injuries via proliferation of cardiomyocytes located near the wound border. To identify regulators of cardiomyocyte proliferation, we used spatially resolved RNA sequencing (tomo-seq) and generated a high-resolution genome-wide atlas of gene expression in the regenerating zebrafish heart. Interestingly, we identified two wound border zones with distinct expression profiles, including the re-expression of embryonic cardiac genes and targets of bone morphogenetic protein (BMP) signaling. Endogenous BMP signaling has been reported to be detrimental to mammalian cardiac repair. In contrast, we find that genetic or chemical inhibition of BMP signaling in zebrafish reduces cardiomyocyte dedifferentiation and proliferation, ultimately compromising myocardial regeneration, while bmp2b overexpression is sufficient to enhance it. Our results provide a resource for further studies on the molecular regulation of cardiac regeneration and reveal intriguing differential cellular responses of cardiomyocytes to a conserved signaling pathway in regenerative versus non-regenerative hearts.
Collapse
Affiliation(s)
- Chi-Chung Wu
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Fabian Kruse
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | | | - Jan Philipp Junker
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - David C Zebrowski
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstr 8-10, 91054 Erlangen, Germany
| | - Kristin Fischer
- Institute of Clinical Genetics, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Emily S Noël
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Dominic Grün
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands; Skolkovo Institute of Science and Technology (Skoltech), Novaya Street 100, Skolkovo, Moscow Region 143025, Russia
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Krankenhausstr 8-10, 91054 Erlangen, Germany
| | - Alexander van Oudenaarden
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | - Jeroen Bakkers
- Hubrecht Institute, University Medical Centre Utrecht, Uppsalaan 8, 3584 CT Utrecht, the Netherlands; Medical Physiology, University Medical Centre Utrecht, Yalelaan 50, 3584 CM Utrecht, the Netherlands.
| |
Collapse
|
14
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
15
|
Parikh A, Wu J, Blanton RM, Tzanakakis ES. Signaling Pathways and Gene Regulatory Networks in Cardiomyocyte Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:377-92. [PMID: 25813860 DOI: 10.1089/ten.teb.2014.0662] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Strategies for harnessing stem cells as a source to treat cell loss in heart disease are the subject of intense research. Human pluripotent stem cells (hPSCs) can be expanded extensively in vitro and therefore can potentially provide sufficient quantities of patient-specific differentiated cardiomyocytes. Although multiple stimuli direct heart development, the differentiation process is driven in large part by signaling activity. The engineering of hPSCs to heart cell progeny has extensively relied on establishing proper combinations of soluble signals, which target genetic programs thereby inducing cardiomyocyte specification. Pertinent differentiation strategies have relied as a template on the development of embryonic heart in multiple model organisms. Here, information on the regulation of cardiomyocyte development from in vivo genetic and embryological studies is critically reviewed. A fresh interpretation is provided of in vivo and in vitro data on signaling pathways and gene regulatory networks (GRNs) underlying cardiopoiesis. The state-of-the-art understanding of signaling pathways and GRNs presented here can inform the design and optimization of methods for the engineering of tissues for heart therapies.
Collapse
Affiliation(s)
- Abhirath Parikh
- 1 Lonza Walkersville, Inc. , Lonza Group, Walkersville, Maryland
| | - Jincheng Wu
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts
| | - Robert M Blanton
- 3 Division of Cardiology, Molecular Cardiology Research Institute , Tufts Medical Center, Tufts School of Medicine, Boston, Massachusetts
| | - Emmanuel S Tzanakakis
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts.,4 Tufts Clinical and Translational Science Institute (CTSI) , Boston, Massachusetts
| |
Collapse
|
16
|
Charpentier MS, Tandon P, Trincot CE, Koutleva EK, Conlon FL. A distinct mechanism of vascular lumen formation in Xenopus requires EGFL7. PLoS One 2015; 10:e0116086. [PMID: 25705891 PMCID: PMC4338030 DOI: 10.1371/journal.pone.0116086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 12/04/2014] [Indexed: 01/03/2023] Open
Abstract
During vertebrate blood vessel development, lumen formation is the critical process by which cords of endothelial cells transition into functional tubular vessels. Here, we use Xenopus embryos to explore the cellular and molecular mechanisms underlying lumen formation of the dorsal aorta and the posterior cardinal veins, the primary major vessels that arise via vasculogenesis within the first 48 hours of life. We demonstrate that endothelial cells are initially found in close association with one another through the formation of tight junctions expressing ZO-1. The emergence of vascular lumens is characterized by elongation of endothelial cell shape, reorganization of junctions away from the cord center to the periphery of the vessel, and onset of Claudin-5 expression within tight junctions. Furthermore, unlike most vertebrate vessels that exhibit specialized apical and basal domains, we show that early Xenopus vessels are not polarized. Moreover, we demonstrate that in embryos depleted of the extracellular matrix factor Epidermal Growth Factor-Like Domain 7 (EGFL7), an evolutionarily conserved factor associated with vertebrate vessel development, vascular lumens fail to form. While Claudin-5 localizes to endothelial tight junctions of EGFL7-depleted embryos in a timely manner, endothelial cells of the aorta and veins fail to undergo appropriate cell shape changes or clear junctions from the cell-cell contact. Taken together, we demonstrate for the first time the mechanisms by which lumens are generated within the major vessels in Xenopus and implicate EGFL7 in modulating cell shape and cell-cell junctions to drive proper lumen morphogenesis.
Collapse
Affiliation(s)
- Marta S. Charpentier
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Panna Tandon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Claire E. Trincot
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Elitza K. Koutleva
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
| | - Frank L. Conlon
- University of North Carolina McAllister Heart Institute, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Genetics and Molecular Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Department of Biology, UNC-CH, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, UNC-CH, Chapel Hill, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
17
|
Abstract
BACKGROUND Defects in cardiac septation are the most common form of congenital heart disease, but the mechanisms underlying these defects are still poorly understood. The small GTPase Rac1 is implicated in planar cell polarity of epithelial cells in Drosophila; however, its role in mammalian cardiomyocyte polarity is not clear. We tested the hypothesis that Rac1 signaling in the second heart field regulates cardiomyocyte polarity, chamber septation, and right ventricle development during embryonic heart development. METHODS AND RESULTS Mice with second heart field-specific deficiency of Rac1 (Rac1(SHF)) exhibited ventricular and atrial septal defects, a thinner right ventricle myocardium, and a bifid cardiac apex. Fate-mapping analysis showed that second heart field contribution to the interventricular septum and right ventricle was deficient in Rac1(SHF) hearts. Notably, cardiomyocytes had a spherical shape with disrupted F-actin filaments in Rac1(SHF) compared with elongated and well-aligned cardiomyocytes in littermate controls. Expression of Scrib, a core protein in planar cell polarity, was lost in Rac1(SHF) hearts with decreased expression of WAVE and Arp2/3, leading to decreased migratory ability. In addition, Rac1-deficient neonatal cardiomyocytes displayed defects in cell projections, lamellipodia formation, and cell elongation. Furthermore, apoptosis was increased and the expression of Gata4, Tbx5, Nkx2.5, and Hand2 transcription factors was decreased in the Rac1(SHF) right ventricle myocardium. CONCLUSIONS Deficiency of Rac1 in the second heart field impairs elongation and cytoskeleton organization of cardiomyocytes and results in congenital septal defects, thin right ventricle myocardium, and a bifid cardiac apex. Our study suggests that Rac1 signaling is critical to cardiomyocyte polarity and embryonic heart development.
Collapse
Affiliation(s)
- Carmen Leung
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada (C.L., X.L., Q.F.) Collaborative Program in Developmental Biology, The University of Western Ontario, London, Ontario, Canada (C.L.)
| | - Xiangru Lu
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada (C.L., X.L., Q.F.)
| | - Murong Liu
- Lawson Health Research Institute, The University of Western Ontario, London, Ontario, Canada (M.L., Q.F.)
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada (C.L., X.L., Q.F.) Department of Medicine, Schulich School of Medicine and Dentistry The University of Western Ontario, London, Ontario, Canada (Q.F.) Lawson Health Research Institute, The University of Western Ontario, London, Ontario, Canada (M.L., Q.F.)
| |
Collapse
|
18
|
Amin NM, Gibbs D, Conlon FL. Differential regulation of CASZ1 protein expression during cardiac and skeletal muscle development. Dev Dyn 2014; 243:948-56. [PMID: 24633745 DOI: 10.1002/dvdy.24126] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 02/19/2014] [Accepted: 02/27/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The zinc-finger transcription factor CASZ1 is required for differentiation of a distinct population of cardiomyocytes during development. However, expression of Casz1 mRNA is detected throughout the developing heart, suggesting the spatial regulation of CASZ1 occurs at the protein level. Relatively little is known about posttranscriptional regulation of Casz1 in the heart. RESULTS We generated antibodies that specifically recognize CASZ1 in developing Xenopus embryos, and performed immunofluorescence analysis of CASZ1 during cardiac development. CASZ1 was detected throughout the developing myocardium. CASZ1 was restricted to terminally differentiated cardiomyocytes, and was down-regulated in cells that re-enter the cell cycle. We determined that CASZ1 expression correlated with terminal differentiation in cardiac muscle cells, skeletal muscle cells, and lymph-heart musculature. CONCLUSIONS This study indicates that spatially distinct expression of CASZ1 protein may be due to posttranscriptional control of Casz1 mRNA during cardiac development. The results of this study provide insights into the role of Casz1 in cardiac function and in the differentiation of other cell types, including skeletal muscle and lymph heart.
Collapse
Affiliation(s)
- Nirav M Amin
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, North Carolina; Department of Genetics, UNC-Chapel Hill, Chapel Hill, North Carolina
| | | | | |
Collapse
|
19
|
Amin NM, Greco TM, Kuchenbrod LM, Rigney MM, Chung MI, Wallingford JB, Cristea IM, Conlon FL. Proteomic profiling of cardiac tissue by isolation of nuclei tagged in specific cell types (INTACT). Development 2014; 141:962-73. [PMID: 24496632 DOI: 10.1242/dev.098327] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The proper dissection of the molecular mechanisms governing the specification and differentiation of specific cell types requires isolation of pure cell populations from heterogeneous tissues and whole organisms. Here, we describe a method for purification of nuclei from defined cell or tissue types in vertebrate embryos using INTACT (isolation of nuclei tagged in specific cell types). This method, previously developed in plants, flies and worms, utilizes in vivo tagging of the nuclear envelope with biotin and the subsequent affinity purification of the labeled nuclei. In this study we successfully purified nuclei of cardiac and skeletal muscle from Xenopus using this strategy. We went on to demonstrate the utility of this approach by coupling the INTACT approach with liquid chromatography-tandem mass spectrometry (LC-MS/MS) proteomic methodologies to profile proteins expressed in the nuclei of developing hearts. From these studies we have identified the Xenopus orthologs of 12 human proteins encoded by genes, which when mutated in human lead to congenital heart disease. Thus, by combining these technologies we are able to identify tissue-specific proteins that are expressed and required for normal vertebrate organ development.
Collapse
Affiliation(s)
- Nirav M Amin
- University of North Carolina McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Samson SC, Ferrer T, Jou CJ, Sachse FB, Shankaran SS, Shaw RM, Chi NC, Tristani-Firouzi M, Yost HJ. 3-OST-7 regulates BMP-dependent cardiac contraction. PLoS Biol 2013; 11:e1001727. [PMID: 24311987 PMCID: PMC3849020 DOI: 10.1371/journal.pbio.1001727] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 10/23/2013] [Indexed: 11/29/2022] Open
Abstract
During zebrafish cardiac development, 3-OST-7 constrains BMP signaling to the atrioventricular junction and precludes it from contractile myocardium, allowing tropomyosin-dependent sarcomere assembly and contraction. The 3-O-sulfotransferase (3-OST) family catalyzes rare modifications of glycosaminoglycan chains on heparan sulfate proteoglycans, yet their biological functions are largely unknown. Knockdown of 3-OST-7 in zebrafish uncouples cardiac ventricular contraction from normal calcium cycling and electrophysiology by reducing tropomyosin4 (tpm4) expression. Normal 3-OST-7 activity prevents the expansion of BMP signaling into ventricular myocytes, and ectopic activation of BMP mimics the ventricular noncontraction phenotype seen in 3-OST-7 depleted embryos. In 3-OST-7 morphants, ventricular contraction can be rescued by overexpression of tropomyosin tpm4 but not by troponin tnnt2, indicating that tpm4 serves as a lynchpin for ventricular sarcomere organization downstream of 3-OST-7. Contraction can be rescued by expression of 3-OST-7 in endocardium, or by genetic loss of bmp4. Strikingly, BMP misregulation seen in 3-OST-7 morphants also occurs in multiple cardiac noncontraction models, including potassium voltage-gated channel gene, kcnh2, affected in Romano-Ward syndrome and long-QT syndrome, and cardiac troponin T gene, tnnt2, affected in human cardiomyopathies. Together these results reveal 3-OST-7 as a key component of a novel pathway that constrains BMP signaling from ventricular myocytes, coordinates sarcomere assembly, and promotes cardiac contractile function. A highly complex environment at the cell surface and in the space between cells is thought to modulate cell behavior. Heparan sulfate proteoglycans are cell surface and extracellular matrix molecules that are covalently linked to long chains of repeating sugar units called glycosaminoglycan chains. These chains can be subjected to rare modifications and they are believed to influence specific cell signaling events in a lineage specific fashion in what is called the “glycocode.” Here we explore the functions of one member of a family of enzymes, 3-O-sulfotransferases (3-OSTs) that catalyzes a rare modification (3-O-sulfation) of glycosaminoglycans in zebrafish. We show that knockdown of 3-OST-7 results in a very specific phenotype, including loss of cardiac ventricle contraction. Knockdown of other 3-OST family members did not result in the same phenotype, suggesting that distinct 3-OST family members have distinct functions in vertebrates and lending in vivo evidence for the glycocode hypothesis. Mechanistically, we found that cardiac contraction can be rescued by reducing the amount of endogenous BMP4, and can be blocked by increasing BMP signaling, suggesting that the glycocode generated by 3-OST-7 is necessary to constrain BMP signaling in the heart for normal cardiac contraction. Furthermore, we show that tropomyosin4 (tpm4) is downstream of 3-OST-7 function, indicating that Tpm4 is key in this pathway to building the sarcomere, the functional contraction unit of the cardiomyocyte.
Collapse
Affiliation(s)
- Shiela C. Samson
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
| | - Tania Ferrer
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Chuanchau J. Jou
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Frank B. Sachse
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Sunita S. Shankaran
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Robin M. Shaw
- Department of Medicine, Cedars-Sinai Heart Institute, University of California, Los Angeles, California, United States of America
| | - Neil C. Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, California, United States of America
| | - Martin Tristani-Firouzi
- Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - H. Joseph Yost
- Department of Neurobiology & Anatomy, University of Utah Molecular Medicine Program, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
21
|
Kaltenbrun E, Greco TM, Slagle CE, Kennedy LM, Li T, Cristea IM, Conlon FL. A Gro/TLE-NuRD corepressor complex facilitates Tbx20-dependent transcriptional repression. J Proteome Res 2013; 12:5395-409. [PMID: 24024827 DOI: 10.1021/pr400818c] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cardiac transcription factor Tbx20 has a critical role in the proper morphogenetic development of the vertebrate heart, and its misregulation has been implicated in human congenital heart disease. Although it is established that Tbx20 exerts its function in the embryonic heart through positive and negative regulation of distinct gene programs, it is unclear how Tbx20 mediates proper transcriptional regulation of its target genes. Here, using a combinatorial proteomic and bioinformatic approach, we present the first characterization of Tbx20 transcriptional protein complexes. We have systematically investigated Tbx20 protein-protein interactions by immunoaffinity purification of tagged Tbx20 followed by proteomic analysis using GeLC-MS/MS, gene ontology classification, and functional network analysis. We demonstrate that Tbx20 is associated with a chromatin remodeling network composed of TLE/Groucho corepressors, members of the Nucleosome Remodeling and Deacetylase (NuRD) complex, the chromatin remodeling ATPases RUVBL1/RUVBL2, and the T-box repressor Tbx18. We determined that the interaction with TLE corepressors is mediated via an eh1 binding motif in Tbx20. Moreover, we demonstrated that ablation of this motif results in a failure to properly assemble the repression network and disrupts Tbx20 function in vivo. Importantly, we validated Tbx20-TLE interactions in the mouse embryonic heart, and identified developmental genes regulated by Tbx20-TLE binding, thereby confirming a primary role for a Tbx20-TLE repressor complex in embryonic heart development. Together, these studies suggest a model in which Tbx20 associates with a Gro/TLE-NuRD repressor complex to prevent inappropriate gene activation within the forming heart.
Collapse
Affiliation(s)
- Erin Kaltenbrun
- Departments of Biology and ‡Genetics, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | | | | | | | | | | | | |
Collapse
|
22
|
Charpentier MS, Christine KS, Amin NM, Dorr KM, Kushner EJ, Bautch VL, Taylor JM, Conlon FL. CASZ1 promotes vascular assembly and morphogenesis through the direct regulation of an EGFL7/RhoA-mediated pathway. Dev Cell 2013; 25:132-43. [PMID: 23639441 DOI: 10.1016/j.devcel.2013.03.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 01/22/2013] [Accepted: 03/01/2013] [Indexed: 01/11/2023]
Abstract
The formation of the vascular system is essential for embryonic development and homeostasis. However, transcriptional control of this process is not fully understood. Here we report an evolutionarily conserved role for the transcription factor CASZ1 (CASTOR) in blood vessel assembly and morphogenesis. In the absence of CASZ1, Xenopus embryos fail to develop a branched and lumenized vascular system, and CASZ1-depleted human endothelial cells display dramatic alterations in adhesion, morphology, and sprouting. Mechanistically, we show that CASZ1 directly regulates Epidermal Growth Factor-Like Domain 7 (Egfl7). We further demonstrate that defects of CASZ1- or EGFL7-depleted cells are in part due to diminished RhoA expression and impaired focal adhesion localization. Moreover, these abnormal endothelial cell behaviors in CASZ1-depleted cells can be rescued by restoration of Egfl7. Collectively, these studies show that CASZ1 is required to directly regulate an EGFL7/RhoA-mediated pathway to promote vertebrate vascular development.
Collapse
Affiliation(s)
- Marta S Charpentier
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Tandon P, Miteva YV, Kuchenbrod LM, Cristea IM, Conlon FL. Tcf21 regulates the specification and maturation of proepicardial cells. Development 2013; 140:2409-21. [PMID: 23637334 DOI: 10.1242/dev.093385] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The epicardium is a mesothelial cell layer essential for vertebrate heart development and pertinent for cardiac repair post-injury in the adult. The epicardium initially forms from a dynamic precursor structure, the proepicardial organ, from which cells migrate onto the heart surface. During the initial stage of epicardial development crucial epicardial-derived cell lineages are thought to be determined. Here, we define an essential requirement for transcription factor Tcf21 during early stages of epicardial development in Xenopus, and show that depletion of Tcf21 results in a disruption in proepicardial cell specification and failure to form a mature epithelial epicardium. Using a mass spectrometry-based approach we defined Tcf21 interactions and established its association with proteins that function as transcriptional co-repressors. Furthermore, using an in vivo systems-based approach, we identified a panel of previously unreported proepicardial precursor genes that are persistently expressed in the epicardial layer upon Tcf21 depletion, thereby confirming a primary role for Tcf21 in the correct determination of the proepicardial lineage. Collectively, these studies lead us to propose that Tcf21 functions as a transcriptional repressor to regulate proepicardial cell specification and the correct formation of a mature epithelial epicardium.
Collapse
Affiliation(s)
- Panna Tandon
- University of North Carolina McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | |
Collapse
|
24
|
Joshi P, Quach OL, Giguere SSB, Cristea IM. A Functional Proteomics Perspective of DBC1 as a Regulator of Transcription. JOURNAL OF PROTEOMICS & BIOINFORMATICS 2013; Suppl 2:002. [PMID: 24273392 PMCID: PMC3837576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The past few years have seen significant advances in the use of modern proteomics approaches for biological discoveries. Among the fields impacted by proteomics is that of epigenetics, as mass spectrometry-based approaches have allowed the identification and characterization of transcriptional regulators, epigenetic marks, and the constantly evolving epigenetic landscape of a cell in health and disease states. These studies have substantially expanded our understanding of critical genes that mediate cell processes, such as differentiation, cell cycle regulation, and apoptosis. Not surprisingly, a great emphasis has been placed on defining factors that are de-regulated in cancers, in an attempt to define new and specific targets for therapeutic design. Differential gene expression observed during carcinogenesis can be induced by aberrant activities of transcription factors and chromatin remodeling enzymes. Through a series of recent mass spectrometry studies of histone deacetylases and nuclear receptors, Deleted in Breast Cancer 1 (DBC1) has emerged as a master regulator of transcriptional processes. DBC1 acts as a modulator of cellular epigenetic mechanisms and is frequently associated with human metastasis. Through its negative regulation of SIRT1 and HDAC3 deacetylation activities, DBC1 has a broad impact on gene expression, downstream cellular pathways, and associated human diseases. Here, we review the identified roles of DBC1, highlighting the critical contribution of mass spectrometry to these findings. Additionally, we provide a perspective of integrative proteomics approaches that can continue to shed light on the interplay between DBC1 and its protein targets, helping to further define its role in epigenetic modifications and to identify novel targets for cancer therapy.
Collapse
Affiliation(s)
- P Joshi
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - O L Quach
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - S S B Giguere
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - I M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ 08544, USA
| |
Collapse
|
25
|
Kerstjens-Frederikse WS, Bongers EMHF, Roofthooft MTR, Leter EM, Douwes JM, Van Dijk A, Vonk-Noordegraaf A, Dijk-Bos KK, Hoefsloot LH, Hoendermis ES, Gille JJP, Sikkema-Raddatz B, Hofstra RMW, Berger RMF. TBX4 mutations (small patella syndrome) are associated with childhood-onset pulmonary arterial hypertension. J Med Genet 2013; 50:500-6. [PMID: 23592887 PMCID: PMC3717587 DOI: 10.1136/jmedgenet-2012-101152] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background Childhood-onset pulmonary arterial hypertension (PAH) is rare and differs from adult-onset disease in clinical presentation, with often unexplained mental retardation and dysmorphic features (MR/DF). Mutations in the major PAH gene, BMPR2, were reported to cause PAH in only 10–16% of childhood-onset patients. We aimed to identify more genes associated with childhood-onset PAH. Methods We studied 20 consecutive cases with idiopathic or heritable PAH. In patients with accompanying MR/DF (n=6) array-comparative genomic hybridisation analysis was performed, with the aim of finding common deletion regions containing candidate genes for PAH. Three patients had overlapping deletions of 17q23.2. TBX2 and TBX4 were selected from this area as candidate genes and sequenced in all 20 children. After identifying TBX4 mutations in these children, we subsequently sequenced TBX4 in a cohort of 49 adults with PAH. Because TBX4 mutations are known to cause small patella syndrome (SPS), all patients with newly detected TBX4 mutations were screened for features of SPS. We also screened a third cohort of 23 patients with SPS for PAH. Results TBX4 mutations (n=3) or TBX4-containing deletions (n=3) were detected in 6 out of 20 children with PAH (30%). All living patients and two parents with TBX4 mutations appeared to have previously unrecognised SPS. In the adult PAH-cohort, one TBX4 mutation (2%) was detected. Screening in the cohort of (predominantly adult) SPS patients revealed no PAH. Conclusions These data indicate that TBX4 mutations are associated with childhood-onset PAH, but that the prevalence of PAH in adult TBX4 mutation carriers is low.
Collapse
|
26
|
Poirier O, Ciumas M, Eyries M, Montagne K, Nadaud S, Soubrier F. Inhibition of apelin expression by BMP signaling in endothelial cells. Am J Physiol Cell Physiol 2012; 303:C1139-45. [DOI: 10.1152/ajpcell.00168.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transforming growth factor-β/bone morphogenic protein (BMP) system is a major pathway for angiogenesis and is involved in hereditary vascular diseases. Here we report that the gene encoding the vasoactive and vascular cell growth-regulating peptide apelin is a target of the BMP pathway. We demonstrate that apelin expression is strongly downregulated by BMP in an endothelial cell line as well as in lung endothelial microvascular cells. We show that BMP signals through the BMPR2-SMAD pathway to downregulate apelin expression and that a transcriptional direct and indirect mechanism is required. The BMP-induced downregulation of apelin expression was found to be critical for hypoxia-induced growth of endothelial cells, because the growth inhibitory effect of BMP in this condition is suppressed by enforced expression of apelin. Thus, we describe an important link between a signaling pathway involved in angiogenesis and vascular diseases and a peptide regulating vascular homeostasis.
Collapse
Affiliation(s)
- Odette Poirier
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Mariana Ciumas
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Mélanie Eyries
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Kevin Montagne
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Sophie Nadaud
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| | - Florent Soubrier
- UMR_S 956 INSERM, Université Pierre et Marie Curie, Paris, France
| |
Collapse
|
27
|
Evolution and development of the building plan of the vertebrate heart. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:783-94. [PMID: 23063530 DOI: 10.1016/j.bbamcr.2012.10.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 10/01/2012] [Accepted: 10/03/2012] [Indexed: 12/11/2022]
Abstract
Early cardiac development involves the formation of a heart tube, looping of the tube and formation of chambers. These processes are highly similar among all vertebrates, which suggest the existence of evolutionary conservation of the building plan of the heart. From the jawless lampreys to man, T-box transcription factors like Tbx5 and Tbx20 are fundamental for heart formation, whereas Tbx2 and Tbx3 repress chamber formation on the sinu-atrial and atrioventricular borders. Also, electrocardiograms from different vertebrates are alike, even though the fish heart only has two chambers whereas the mammalian heart has four chambers divided by septa and in addition has much higher heart rates. We conclude that most features of the high-performance hearts of mammals and birds can be traced back to less developed traits in the hearts of ectothermic vertebrates. This article is part of a Special Issue entitled: Cardiomyocyte biology: Cardiac pathways of differentiation, metabolism and contraction.
Collapse
|
28
|
Gaborit N, Sakuma R, Wylie JN, Kim KH, Zhang SS, Hui CC, Bruneau BG. Cooperative and antagonistic roles for Irx3 and Irx5 in cardiac morphogenesis and postnatal physiology. Development 2012; 139:4007-19. [PMID: 22992950 DOI: 10.1242/dev.081703] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Iroquois homeobox (Irx) homeodomain transcription factors are important for several aspects of embryonic development. In the developing heart, individual Irx genes are important for certain postnatal cardiac functions, including cardiac repolarization (Irx5) and rapid ventricular conduction (Irx3). Irx genes are expressed in dynamic and partially overlapping patterns in the developing heart. Here we show in mice that Irx3 and Irx5 have redundant function in the endocardium to regulate atrioventricular canal morphogenesis and outflow tract formation. Our data suggest that direct transcriptional repression of Bmp10 by Irx3 and Irx5 in the endocardium is required for ventricular septation. A postnatal deletion of Irx3 and Irx5 in the myocardium leads to prolongation of atrioventricular conduction, due in part to activation of expression of the Na(+) channel protein Nav1.5. Surprisingly, combined postnatal loss of Irx3 and Irx5 results in a restoration of the repolarization gradient that is altered in Irx5 mutant hearts, suggesting that postnatal Irx3 activity can be repressed by Irx5. Our results have uncovered complex genetic interactions between Irx3 and Irx5 in embryonic cardiac development and postnatal physiology.
Collapse
Affiliation(s)
- Nathalie Gaborit
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Tu S, Chi NC. Zebrafish models in cardiac development and congenital heart birth defects. Differentiation 2012; 84:4-16. [PMID: 22704690 DOI: 10.1016/j.diff.2012.05.005] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/02/2012] [Accepted: 05/21/2012] [Indexed: 12/31/2022]
Abstract
The zebrafish has become an ideal vertebrate animal system for investigating cardiac development due to its genetic tractability, external fertilization, early optical clarity and ability to survive without a functional cardiovascular system during development. In particular, recent advances in imaging techniques and the creation of zebrafish transgenics now permit the in vivo analysis of the dynamic cellular events that transpire during cardiac morphogenesis. As a result, the combination of these salient features provides detailed insight as to how specific genes may influence cardiac development at the cellular level. In this review, we will highlight how the zebrafish has been utilized to elucidate not only the underlying mechanisms of cardiac development and human congenital heart diseases (CHDs), but also potential pathways that may modulate cardiac regeneration. Thus, we have organized this review based on the major categories of CHDs-structural heart, functional heart, and vascular/great vessel defects, and will conclude with how the zebrafish may be further used to contribute to our understanding of specific human CHDs in the future.
Collapse
Affiliation(s)
- Shu Tu
- Department of Medicine, Division of Cardiology, University of California, San Diego, CA 92093-0613J, USA
| | | |
Collapse
|
30
|
Qiao Y, Wanyan H, Xing Q, Xie W, Pang S, Shan J, Yan B. Genetic analysis of the TBX20 gene promoter region in patients with ventricular septal defects. Gene 2012; 500:28-31. [DOI: 10.1016/j.gene.2012.03.055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 03/05/2012] [Accepted: 03/13/2012] [Indexed: 01/08/2023]
|
31
|
de Pater E, Ciampricotti M, Priller F, Veerkamp J, Strate I, Smith K, Lagendijk AK, Schilling TF, Herzog W, Abdelilah-Seyfried S, Hammerschmidt M, Bakkers J. Bmp signaling exerts opposite effects on cardiac differentiation. Circ Res 2012; 110:578-87. [PMID: 22247485 DOI: 10.1161/circresaha.111.261172] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE The importance for Bmp signaling during embryonic stem cell differentiation into myocardial cells has been recognized. The question when and where Bmp signaling in vivo regulates myocardial differentiation has remained largely unanswered. OBJECTIVE To identify when and where Bmp signaling regulates cardiogenic differentiation. METHODS AND RESULTS Here we have observed that in zebrafish embryos, Bmp signaling is active in cardiac progenitor cells prior to their differentiation into cardiomyocytes. Bmp signaling is continuously required during somitogenesis within the anterior lateral plate mesoderm to induce myocardial differentiation. Surprisingly, Bmp signaling is actively repressed in differentiating myocardial cells. We identified the inhibitory Smad6a, which is expressed in the cardiac tissue, to be required to inhibit Bmp signaling and thereby promote expansion of the ventricular myocardium. CONCLUSION Bmp signaling exerts opposing effects on myocardial differentiation in the embryo by promoting as well as inhibiting cardiac growth.
Collapse
Affiliation(s)
- Emma de Pater
- Cardiac development and genetics group, Hubrecht Institute for Developmental Biology and Stem Cell Research, Uppsalalaan 8, Utrecht, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lee MP, Yutzey KE. Twist1 directly regulates genes that promote cell proliferation and migration in developing heart valves. PLoS One 2011; 6:e29758. [PMID: 22242143 PMCID: PMC3248441 DOI: 10.1371/journal.pone.0029758] [Citation(s) in RCA: 35] [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: 09/01/2011] [Accepted: 12/05/2011] [Indexed: 11/19/2022] Open
Abstract
Twist1, a basic helix-loop-helix transcription factor, is expressed in mesenchymal precursor populations during embryogenesis and in metastatic cancer cells. In the developing heart, Twist1 is highly expressed in endocardial cushion (ECC) valve mesenchymal cells and is down regulated during valve differentiation and remodeling. Previous studies demonstrated that Twist1 promotes cell proliferation, migration, and expression of primitive extracellular matrix (ECM) molecules in ECC mesenchymal cells. Furthermore, Twist1 expression is induced in human pediatric and adult diseased heart valves. However, the Twist1 downstream target genes that mediate increased cell proliferation and migration during early heart valve development remain largely unknown. Candidate gene and global gene profiling approaches were used to identify transcriptional targets of Twist1 during heart valve development. Candidate target genes were analyzed for evolutionarily conserved regions (ECRs) containing E-box consensus sequences that are potential Twist1 binding sites. ECRs containing conserved E-box sequences were identified for Twist1 responsive genes Tbx20, Cdh11, Sema3C, Rab39b, and Gadd45a. Twist1 binding to these sequences in vivo was determined by chromatin immunoprecipitation (ChIP) assays, and binding was detected in ECCs but not late stage remodeling valves. In addition identified Twist1 target genes are highly expressed in ECCs and have reduced expression during heart valve remodeling in vivo, which is consistent with the expression pattern of Twist1. Together these analyses identify multiple new genes involved in cell proliferation and migration that are differentially expressed in the developing heart valves, are responsive to Twist1 transcriptional function, and contain Twist1-responsive regulatory sequences.
Collapse
Affiliation(s)
- Mary P. Lee
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Katherine E. Yutzey
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
33
|
Tbx20 regulation of cardiac cell proliferation and lineage specialization during embryonic and fetal development in vivo. Dev Biol 2011; 363:234-46. [PMID: 22226977 DOI: 10.1016/j.ydbio.2011.12.034] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 11/29/2011] [Accepted: 12/20/2011] [Indexed: 11/21/2022]
Abstract
TBX20 gain-of-function mutations in humans are associated with congenital heart malformations and myocardial defects. However the effects of increased Tbx20 function during cardiac chamber development and maturation have not been reported previously. CAG-CAT-Tbx20 transgenic mice were generated for Cre-dependent induction of Tbx20 in myocardial lineages in the developing heart. βMHCCre-mediated overexpression of Tbx20 in fetal ventricular cardiomyocytes results in increased thickness of compact myocardium, induction of cardiomyocyte proliferation, and increased expression of Bmp10 and pSmad1/5/8 at embryonic day (E) 14.5. βMHCCre-mediated Tbx20 overexpression also leads to increased expression of cardiac conduction system (CCS) genes Tbx5, Cx40, and Cx43 throughout the ventricular myocardium. In contrast, Nkx2.5Cre mediated overexpression of Tbx20 in the embryonic heart results in reduced cardiomyocyte proliferation, increased expression of a cell cycle inhibitor, p21(CIP1), and decreased expression of Tbx2, Tbx5, and N-myc1 at E9.5, concomitant with decreased phospho-ERK1/2 expression. Together, these analyses demonstrate that Tbx20 differentially regulates cell proliferation and cardiac lineage specification in embryonic versus fetal cardiomyocytes. Induction of pSmad1/5/8 at E14.5 and inhibition of dpERK expression at E9.5 are consistent with selective Tbx20 regulation of these pathways in association with stage-specific effects on cardiomyocyte proliferation. Together, these in vivo data support distinct functions for Tbx20 in regulation of cardiomyocyte lineage maturation and cell proliferation at embryonic and fetal stages of heart development.
Collapse
|
34
|
Cai X, Nomura-Kitabayashi A, Cai W, Yan J, Christoffels VM, Cai CL. Myocardial Tbx20 regulates early atrioventricular canal formation and endocardial epithelial-mesenchymal transition via Bmp2. Dev Biol 2011; 360:381-90. [PMID: 21983003 PMCID: PMC3217163 DOI: 10.1016/j.ydbio.2011.09.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 09/05/2011] [Accepted: 09/21/2011] [Indexed: 01/18/2023]
Abstract
During early embryogenesis, the formation of the cardiac atrioventricular canal (AVC) facilitates the transition of the heart from a linear tube into a chambered organ. However, the genetic pathways underlying this developmental process are poorly understood. The T-box transcription factor Tbx20 is expressed predominantly in the AVC of early heart tube. It was shown that Tbx20 activates Nmyc1 and suppresses Tbx2 expression to promote proliferation and specification of the atrial and ventricular chambers, yet it is not known if Tbx20 is involved in early AVC development. Here, we report that mice lacking Tbx20 in the AVC myocardium fail to form the AVC constriction, and the endocardial epithelial-mesenchymal transition (EMT) is severely perturbed. Tbx20 maintains expression of a variety of genes, including Bmp2, Tbx3 and Hand1 in the AVC myocardium. Intriguingly, we found Bmp2 downstream genes involved in the EMT initiation are also downregulated. In addition, re-expression of Bmp2 in the AVC myocardium substantially rescues the EMT defects resulting from the lack of Tbx20, suggesting Bmp2 is one of the key downstream targets of Tbx20 in AVC development. Our data support a complex signaling network with Tbx20 suppressing Tbx2 in the AVC myocardium but also indirectly promoting Tbx2 expression through Bmp2. The spatiotemporal expression of Tbx2 in the AVC appears to be balanced between these two opposing signals. Overall, our study provides genetic evidence that Tbx20 has essential roles in regulating AVC development that coordinate early cardiac chamber formation.
Collapse
Affiliation(s)
- Xiaoqiang Cai
- Department of Developmental and Regenerative Biology, Center for Molecular Cardiology, the Child Health and Development Institute and the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Aya Nomura-Kitabayashi
- Department of Developmental and Regenerative Biology, Center for Molecular Cardiology, the Child Health and Development Institute and the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Weibin Cai
- Department of Developmental and Regenerative Biology, Center for Molecular Cardiology, the Child Health and Development Institute and the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Jianyun Yan
- Department of Developmental and Regenerative Biology, Center for Molecular Cardiology, the Child Health and Development Institute and the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Vincent M. Christoffels
- Heart Failure Research Center, Academic Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Chen-Leng Cai
- Department of Developmental and Regenerative Biology, Center for Molecular Cardiology, the Child Health and Development Institute and the Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|
35
|
Shen T, Aneas I, Sakabe N, Dirschinger RJ, Wang G, Smemo S, Westlund JM, Cheng H, Dalton N, Gu Y, Boogerd CJ, Cai CL, Peterson K, Chen J, Nobrega MA, Evans SM. Tbx20 regulates a genetic program essential to adult mouse cardiomyocyte function. J Clin Invest 2011; 121:4640-54. [PMID: 22080862 DOI: 10.1172/jci59472] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 10/05/2011] [Indexed: 12/19/2022] Open
Abstract
Human mutations in or variants of TBX20 are associated with congenital heart disease, cardiomyopathy, and arrhythmias. To investigate whether cardiac disease in patients with these conditions results from an embryonic or ongoing requirement for Tbx20 in myocardium, we ablated Tbx20 specifically in adult cardiomyocytes in mice. This ablation resulted in the onset of severe cardiomyopathy accompanied by arrhythmias, with death ensuing within 1 to 2 weeks of Tbx20 ablation. Accounting for this dramatic phenotype, we identified molecular signatures that posit Tbx20 as a central integrator of a genetic program that maintains cardiomyocyte function in the adult heart. Expression of a number of genes encoding critical transcription factors, ion channels, and cytoskeletal/myofibrillar proteins was downregulated consequent to loss of Tbx20. Genome-wide ChIP analysis of Tbx20-binding regions in the adult heart revealed that many of these genes were direct downstream targets of Tbx20 and uncovered a previously undescribed DNA-binding site for Tbx20. Bioinformatics and in vivo functional analyses revealed a cohort of transcription factors that, working with Tbx20, integrated multiple environmental signals to maintain ion channel gene expression in the adult heart. Our data provide insight into the mechanisms by which mutations in TBX20 cause adult heart disease in humans.
Collapse
Affiliation(s)
- Tao Shen
- Skaggs School of Pharmacy, UCSD, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhang W, Chen H, Wang Y, Yong W, Zhu W, Liu Y, Wagner GR, Payne RM, Field LJ, Xin H, Cai CL, Shou W. Tbx20 transcription factor is a downstream mediator for bone morphogenetic protein-10 in regulating cardiac ventricular wall development and function. J Biol Chem 2011; 286:36820-9. [PMID: 21890625 DOI: 10.1074/jbc.m111.279679] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic protein 10 (BMP10) belongs to the TGFβ-superfamily. Previously, we had demonstrated that BMP10 is a key regulator for ventricular chamber formation, growth, and maturation. Ablation of BMP10 leads to hypoplastic ventricular wall formation, and elevated levels of BMP10 are associated with abnormal ventricular trabeculation/compaction and wall maturation. However, the molecular mechanism(s) by which BMP10 regulates ventricle wall growth and maturation is still largely unknown. In this study, we sought to identify the specific transcriptional network that is potentially mediated by BMP10. We analyzed and compared the gene expression profiles between α-myosin heavy chain (αMHC)-BMP10 transgenic hearts and nontransgenic littermate controls using Affymetrix mouse exon arrays. T-box 20 (Tbx20), a cardiac transcription factor, was significantly up-regulated in αMHC-BMP10 transgenic hearts, which was validated by quantitative RT-PCR and in situ hybridization. Ablation of BMP10 reduced Tbx20 expression specifically in the BMP10-expressing region of the developing ventricle. In vitro promoter analysis demonstrated that BMP10 was able to induce Tbx20 promoter activity through a conserved Smad binding site in the Tbx20 promoter proximal region. Furthermore, overexpression of Tbx20 in myocardium led to dilated cardiomyopathy that exhibited ventricular hypertrabeculation and an abnormal muscular septum, which phenocopied genetically modified mice with elevated BMP10 levels. Taken together, our findings demonstrate that the BMP10-Tbx20 signaling cascade is important for ventricular wall development and maturation.
Collapse
Affiliation(s)
- Wenjun Zhang
- Riley Heart Research Center, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Song L, Zhao M, Wu B, Zhou B, Wang Q, Jiao K. Cell autonomous requirement of endocardial Smad4 during atrioventricular cushion development in mouse embryos. Dev Dyn 2011; 240:211-20. [PMID: 21089072 DOI: 10.1002/dvdy.22493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrioventricular (AV) cushions are the precursors of AV septum and valves. In this study, we examined roles of Smad4 during AV cushion development using a conditional gene inactivation approach. We found that endothelial/endocardial inactivation of Smad4 led to the hypocellular AV cushion defect and that both reduced cell proliferation and increased apoptosis contributed to the defect. Expression of multiple genes critical for cushion development was down-regulated in mutant embryos. In collagen gel assays, the number of mesenchymal cells formed is significantly reduced in mutant AV explants compared to that in control explants, suggesting that the reduction of cushion mesenchyme formation in mutants is unlikely secondary to their gross vasculature abnormalities. Using a previously developed immortal endocardial cell line, we showed that Smad4 is required for BMP signaling- stimulated upregulation of Tbx20 and Gata4. Therefore, our data collectively support the cell-autonomous requirement of endocardial Smad4 in regulating AV cushion development.
Collapse
Affiliation(s)
- Langying Song
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
38
|
Kaltenbrun E, Tandon P, Amin NM, Waldron L, Showell C, Conlon FL. Xenopus: An emerging model for studying congenital heart disease. ACTA ACUST UNITED AC 2011; 91:495-510. [PMID: 21538812 DOI: 10.1002/bdra.20793] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Revised: 01/18/2011] [Accepted: 01/28/2011] [Indexed: 02/02/2023]
Abstract
Congenital heart defects affect nearly 1% of all newborns and are a significant cause of infant death. Clinical studies have identified a number of congenital heart syndromes associated with mutations in genes that are involved in the complex process of cardiogenesis. The African clawed frog, Xenopus, has been instrumental in studies of vertebrate heart development and provides a valuable tool to investigate the molecular mechanisms underlying human congenital heart diseases. In this review, we discuss the methodologies that make Xenopus an ideal model system to investigate heart development and disease. We also outline congenital heart conditions linked to cardiac genes that have been well studied in Xenopus and describe some emerging technologies that will further aid in the study of these complex syndromes.
Collapse
Affiliation(s)
- Erin Kaltenbrun
- University of North Carolina McAllister Heart Institute, Chapel Hill, NC 27599, USA
| | | | | | | | | | | |
Collapse
|
39
|
Abstract
The multi-chambered mammalian heart arises from a simple tube by polar elongation, myocardial differentiation and morphogenesis. Members of the large family of T-box (Tbx) transcription factors have been identified as crucial players that act in distinct subprogrammes during cardiac regionalization. Tbx1 and Tbx18 ensure elongation of the cardiac tube at the anterior and posterior pole, respectively. Tbx1 acts in the pharyngeal mesoderm to maintain proliferation of mesenchymal precursor cells for formation of a myocardialized and septated outflow tract. Tbx18 is expressed in the sinus venosus region and is required for myocardialization of the caval veins and the sinoatrial node. Tbx5 and Tbx20 function in the early heart tube and independently activate the chamber myocardial gene programme, whereas Tbx2 and Tbx3 locally repress this programme to favour valvuloseptal and conduction system development. Here, we summarize that these T-box factors act in different molecular circuits and control target gene expression using diverse molecular strategies including binding to distinct protein interaction partners.
Collapse
Affiliation(s)
- Franziska Greulich
- Institute for Molecular Biology, OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | | | | |
Collapse
|
40
|
Domyan ET, Ferretti E, Throckmorton K, Mishina Y, Nicolis SK, Sun X. Signaling through BMP receptors promotes respiratory identity in the foregut via repression of Sox2. Development 2011; 138:971-81. [PMID: 21303850 DOI: 10.1242/dev.053694] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The mammalian foregut gives rise to the dorsally located esophagus and stomach and the ventrally located trachea and lung. Proper patterning and morphogenesis of the common foregut tube and its derived organs is essential for viability of the organism at birth. Here, we show that conditional inactivation of BMP type I receptor genes Bmpr1a and Bmpr1b (Bmpr1a;b) in the ventral endoderm leads to tracheal agenesis and ectopic primary bronchi. Molecular analyses of these mutants reveal a reduction of ventral endoderm marker NKX2-1 and an expansion of dorsal markers SOX2 and P63 into the prospective trachea and primary bronchi. Subsequent genetic experiments show that activation of canonical WNT signaling, previously shown to induce ectopic respiratory fate in otherwise wild-type mice, is incapable of promoting respiratory fate in the absence of Bmpr1a;b. Furthermore, we find that inactivation of Sox2 in Bmpr1a;b mutants does not suppress ectopic lung budding but does rescue trachea formation and NKX2-1 expression. Together, our data suggest that signaling through BMPR1A;B performs at least two roles in early respiratory development: first, it promotes tracheal formation through repression of Sox2; and second, it restricts the site of lung bud initiation.
Collapse
Affiliation(s)
- Eric T Domyan
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | | | | | | | | |
Collapse
|
41
|
Wang J, Greene SB, Martin JF. BMP signaling in congenital heart disease: new developments and future directions. ACTA ACUST UNITED AC 2011; 91:441-8. [PMID: 21384533 DOI: 10.1002/bdra.20785] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 12/22/2010] [Accepted: 01/03/2011] [Indexed: 01/07/2023]
Abstract
Congenital heart malformations are the most common of all congenital human birth anomalies. During the past decade, research with zebrafish, chick, and mouse models have elucidated many fundamental genetic pathways that govern early cardiac patterning and differentiation. This review highlights the roles of the bone morphogenetic protein (BMP) signaling pathway in cardiogenesis and how defective BMP signals can disrupt the intricate steps of cardiac formation and cause congenital heart defects.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Biosciences and Technology, Texas A&M System Health Science Center, 2121 W. Holcombe Blvd., Houston, TX 77030, USA
| | | | | |
Collapse
|
42
|
Laux DW, Febbo JA, Roman BL. Dynamic analysis of BMP-responsive smad activity in live zebrafish embryos. Dev Dyn 2011; 240:682-94. [PMID: 21337466 DOI: 10.1002/dvdy.22558] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2010] [Indexed: 11/06/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are critical players in development and disease, regulating such diverse processes as dorsoventral patterning, palate formation, and ossification. These ligands are classically considered to signal via BMP receptor-specific Smad proteins 1, 5, and 8. To determine the spatiotemporal pattern of Smad1/5/8 activity and thus canonical BMP signaling in the developing zebrafish embryo, we generated a transgenic line expressing EGFP under the control of a BMP-responsive element. EGFP is expressed in many established BMP signaling domains and is responsive to alterations in BMP type I receptor activity and smad1 and smad5 expression. This transgenic Smad1/5/8 reporter line will be useful for determining ligand and receptor requirements for specific domains of BMP activity, as well as for genetic and pharmacological screens aimed at identifying enhancers or suppressors of canonical BMP signaling.
Collapse
|
43
|
Abstract
The myocardium of the heart is composed of multiple highly specialized myocardial lineages, including those of the ventricular and atrial myocardium, and the specialized conduction system. Specification and maturation of each of these lineages during heart development is a highly ordered, ongoing process involving multiple signaling pathways and their intersection with transcriptional regulatory networks. Here, we attempt to summarize and compare much of what we know about specification and maturation of myocardial lineages from studies in several different vertebrate model systems. To date, most research has focused on early specification, and although there is still more to learn about early specification, less is known about factors that promote subsequent maturation of myocardial lineages required to build the functioning adult heart.
Collapse
Affiliation(s)
- Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla CA 92093, USA.
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Michela Noseda
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Tessa Peterkin
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Filipa C. Simões
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Roger Patient
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| | - Michael D. Schneider
- From the British Heart Foundation Centre of Research Excellence (M.N., M.D.S.), National Heart and Lung Institute, Imperial College London; and the Weatherall Institute of Molecular Medicine (T.P., F.C.S., R.P.), University of Oxford, United Kingdom
| |
Collapse
|
45
|
Sengupta A, Molkentin JD, Paik JH, DePinho RA, Yutzey KE. FoxO transcription factors promote cardiomyocyte survival upon induction of oxidative stress. J Biol Chem 2010; 286:7468-78. [PMID: 21159781 DOI: 10.1074/jbc.m110.179242] [Citation(s) in RCA: 257] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Transcriptional regulatory mechanisms of cardiac oxidative stress resistance are not well defined. FoxO transcription factors are critical mediators of oxidative stress resistance in multiple cell types, but cardioprotective functions have not been reported previously. FoxO function in oxidative stress resistance was investigated in cultured cardiomyocytes and in mice with cardiomyocyte-specific combined deficiency of FoxO1 and FoxO3 subjected to myocardial infarction (MI) or acute ischemia/reperfusion (I/R) injury. Induction of oxidative stress in cardiomyocytes promotes FoxO1 and FoxO3 nuclear localization and target gene activation. Infection of cardiomyocytes with a dominant-negative FoxO1(Δ256) adenovirus results in a significant increase in reactive oxygen species and cell death, whereas increased FoxO1 or FoxO3 expression reduces reactive oxygen species and cell death. Mice generated with combined conditional deletion of FoxO1 and FoxO3 specifically in cardiomyocytes were subjected to I/R or MI. Loss of FoxO1 and FoxO3 in cardiomyocytes results in a significant increase in infarct area with decreased expression of the antiapoptotic molecules, PTEN-induced kinase1 (PINK1) and CBP/P300-interacting transactivator (CITED2). Expressions of the antioxidants catalase and manganese superoxide dismutase-2 (SOD2) and the autophagy-related proteins LC3II and Gabarapl1 also are decreased following I/R compared with controls. Mice with cardiomyocyte-specific FoxO deficiency subjected to MI have reduced cardiac function, increased scar formation, induction of stress-responsive signaling, and increased apoptotic cell death relative to controls. These data support a critical role for FoxOs in promoting cardiomyocyte survival during conditions of oxidative stress through induction of antioxidants and cell survival pathways.
Collapse
Affiliation(s)
- Arunima Sengupta
- Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Medical Center, Cincinnati, Ohio 45229, USA
| | | | | | | | | |
Collapse
|
46
|
|