1
|
Boulgakoff L, D'Amato G, Miquerol L. Molecular Regulation of Cardiac Conduction System Development. Curr Cardiol Rep 2024; 26:943-952. [PMID: 38990492 DOI: 10.1007/s11886-024-02094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE OF REVIEW The cardiac conduction system, composed of pacemaker cells and conducting cardiomyocytes, orchestrates the propagation of electrical activity to synchronize heartbeats. The conduction system plays a crucial role in the development of cardiac arrhythmias. In the embryo, the cells of the conduction system derive from the same cardiac progenitors as the contractile cardiomyocytes and and the key question is how this choice is made during development. RECENT FINDINGS This review focuses on recent advances in developmental biology using the mouse as animal model to better understand the cellular origin and molecular regulations that control morphogenesis of the cardiac conduction system, including the latest findings in single-cell transcriptomics. The conducting cell fate is acquired during development starting with pacemaking activity and last with the formation of a complex fast-conducting network. Cardiac conduction system morphogenesis is controlled by complex transcriptional and gene regulatory networks that differ in the components of the cardiac conduction system.
Collapse
Affiliation(s)
| | - Gaetano D'Amato
- Aix-Marseille Université, CNRS IBDM UMR7288, Marseille, France
| | - Lucile Miquerol
- Aix-Marseille Université, CNRS IBDM UMR7288, Marseille, France.
| |
Collapse
|
2
|
Folkerts EJ, Snihur KN, Zhang Y, Martin JW, Alessi DS, Goss GG. Embryonic cardio-respiratory impairments in rainbow trout (Oncorhynchus mykiss) following exposure to hydraulic fracturing flowback and produced water. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 310:119886. [PMID: 35934150 DOI: 10.1016/j.envpol.2022.119886] [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: 01/04/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
During hydraulic fracturing, wastewaters - termed flowback and produced water (FPW) - are created as a by-product during hydrocarbon extraction. Given the large volumes of FPW that a single well can produce, and the history of FPW release to surface water bodies, it is imperative to understand the hazards that hydraulic fracturing and FPW pose to aquatic biota. Using rainbow trout embryos as model organisms, we investigated impacts to cardio-respiratory system development and function following acute (48 h) and sub-chronic (28-day) FPW exposure by examining occurrences of developmental deformities, rates of embryonic respiration (MO2), and changes in expression of critical cardiac-specific genes. FPW-exposed embryos had significantly increased rates of pericardial edema, yolk-sac edema, and tail/trunk curvatures at hatch. Furthermore, when exposed at three days post-fertilization (dpf), acute 5% FPW exposures significantly increased embryonic MO2 through development until 15 dpf, where a switch to significantly reduced MO2 rates was subsequently recorded. A similar trend was observed during sub-chronic 1% FPW exposures. Interestingly, at certain specific developmental timepoints, previous salinity exposure seemed to affect embryonic MO2; a result not previously observed. Following acute FPW exposures, embryonic genes for cardiac development and function were significantly altered, although at termination of sub-chronic exposures, significant changes to these same genes were not found. Together, our evidence of induced developmental deformities, modified embryonic MO2, and altered cardiac transcript expression suggest that cardio-respiratory tissues are toxicologically targeted following FPW exposure in developing rainbow trout. These results may be helpful to regulatory bodies when developing hazard identification and risk management protocols concerning hydraulic fracturing activities.
Collapse
Affiliation(s)
- Erik J Folkerts
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada.
| | - Katherine N Snihur
- Department of Earth and Atmospheric Sciences, University of Alberta, Edmonton, AB, T6G 2E3, Canada
| | - Yifeng Zhang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, T6G 2G3, Alberta, Canada
| | - Jonathan W Martin
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, T6G 2G3, Alberta, Canada; Department of Environmental Science, Stockholm University, Stockholm, Sweden
| | - Daniel S Alessi
- Department of Earth and Atmospheric Sciences, University of Alberta, Edmonton, AB, T6G 2E3, Canada
| | - Greg G Goss
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada; NRC- University of Alberta Nanotechnology Initiative, Edmonton, AB, T6G 2M9, Canada
| |
Collapse
|
3
|
Nkx2-5 defines distinct scaffold and recruitment phases during formation of the murine cardiac Purkinje fiber network. Nat Commun 2020; 11:5300. [PMID: 33082351 PMCID: PMC7575572 DOI: 10.1038/s41467-020-19150-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 09/29/2020] [Indexed: 01/24/2023] Open
Abstract
The ventricular conduction system coordinates heartbeats by rapid propagation of electrical activity through the Purkinje fiber (PF) network. PFs share common progenitors with contractile cardiomyocytes, yet the mechanisms of segregation and network morphogenesis are poorly understood. Here, we apply genetic fate mapping and temporal clonal analysis to identify murine cardiomyocytes committed to the PF lineage as early as E7.5. We find that a polyclonal PF network emerges by progressive recruitment of conductive precursors to this scaffold from a pool of bipotent progenitors. At late fetal stages, the segregation of conductive cells increases during a phase of rapid recruitment to build the definitive PF network through a non-cell autonomous mechanism. We also show that PF differentiation is impaired in Nkx2-5 haploinsufficient embryos leading to failure to extend the scaffold. In particular, late fetal recruitment fails, resulting in PF hypoplasia and persistence of bipotent progenitors. Our results identify how transcription factor dosage regulates cell fate divergence during distinct phases of PF network morphogenesis. Here, the authors apply genetic fate mapping and temporal clonal analysis to study progenitor recruitment and network morphogenesis of murine cardiac Purkinje fibers. Additionally, they characterize how transcription factor dosage regulates cell fate divergence during distinct phases of this process.
Collapse
|
4
|
Genetic evolution and codon usage analysis of NKX-2.5 gene governing heart development in some mammals. Genomics 2019; 112:1319-1329. [PMID: 31377427 DOI: 10.1016/j.ygeno.2019.07.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 11/21/2022]
Abstract
NKX-2.5 gene is responsible for cardiac development and its targeted disruption apprehends cardiac development at the linear heart tube stage. Bioinformatic analysis was employed to investigate the codon usage pattern and dN/dS of mammalian NKX-2.5 gene. The relative synonymous codon usage analysis revealed variation in codon usage and two synonymous codons namely ATA (Ile) and GTA (Val) were absent in NKX-2.5 gene across selected mammalian species suggesting that these two codons were possibly selected against during evolution. Parity rule 2 analysis of two and four fold amino acids showed CT bias whereas six-fold amino acids revealed GA bias. Neutrality analysis suggests that selection played a prominent role while mutation had a minor role. The dN/dS analysis suggests synonymous substitution played a significant role and it negatively correlated with p-distance of the gene. Purifying natural selection played a dominant role in the genetic evolution of NKX-2.5 gene in mammals.
Collapse
|
5
|
Goodyer W, Wu SM. Fates Aligned: Origins and Mechanisms of Ventricular Conduction System and Ventricular Wall Development. Pediatr Cardiol 2018; 39:1090-1098. [PMID: 29594502 PMCID: PMC6093793 DOI: 10.1007/s00246-018-1869-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/14/2018] [Indexed: 12/19/2022]
Abstract
The cardiac conduction system is a network of distinct cell types necessary for the coordinated contraction of the cardiac chambers. The distal portion, known as the ventricular conduction system, allows for the rapid transmission of impulses from the atrio-ventricular node to the ventricular myocardium and plays a central role in cardiac function as well as disease when perturbed. Notably, its patterning during embryogenesis is intimately linked to that of ventricular wall formation, including trabeculation and compaction. Here, we review our current understanding of the underlying mechanisms responsible for the development and maturation of these interdependent processes.
Collapse
Affiliation(s)
- William Goodyer
- Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA,Division of Pediatric Cardiology, Department of Pediatrics, Lucille Packard Children’s Hospital, Stanford, CA 94305, USA
| | - Sean M. Wu
- Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA,Correspondence: Sean M. Wu, M.D. PhD., Lokey Stem Cell Building, Room G1120A, 265 Campus Drive, Stanford, CA 94305, Phone No. 650-724-4498, Fax No. 650-726-4689,
| |
Collapse
|
6
|
Abstract
TBX5 is a member of the T-box transcription factor family and is primarily known for its role in cardiac and forelimb development. Human patients with dominant mutations in TBX5 are characterized by Holt-Oram syndrome, and show defects of the cardiac septa, cardiac conduction system, and the anterior forelimb. The range of cardiac defects associated with TBX5 mutations in humans suggests multiple roles for the transcription factor in cardiac development and function. Animal models demonstrate similar defects and have provided a useful platform for investigating the roles of TBX5 during embryonic development. During early cardiac development, TBX5 appears to act primarily as a transcriptional activator of genes associated with cardiomyocyte maturation and upstream of morphological signals for septation. During later cardiac development, TBX5 is required for patterning of the cardiac conduction system and maintenance of mature cardiomyocyte function. A comprehensive understanding of the integral roles of TBX5 throughout cardiac development and adult life will be critical for understanding human cardiac morphology and function.
Collapse
Affiliation(s)
- J D Steimle
- University of Chicago, Chicago, IL, United States
| | | |
Collapse
|
7
|
Du Z, Wang G, Gao S, Wang Z. Aryl organophosphate flame retardants induced cardiotoxicity during zebrafish embryogenesis: by disturbing expression of the transcriptional regulators. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2015; 161:25-32. [PMID: 25661707 DOI: 10.1016/j.aquatox.2015.01.027] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 01/22/2015] [Accepted: 01/28/2015] [Indexed: 05/03/2023]
Abstract
As a result of the ban on some brominated flame retardants (BFRs), the use of organophosphate flame retardants (OPFRs) increases, and they are detected in multi-environment media at higher frequency and concentrations. However, the toxicity data of OPFRs, especially those on developmental toxicology are quite limited, which prevents an accurate evaluation of their environmental and health risk. Because a previous study reported that two aryl-OPFRs induced cardiotoxicity during zebrafish embryogenesis, we designed experiments to compare the heart developmental toxicity of a series of aryl-OPFRs with alkyl-OPFRs and explored possible internal mechanism. First, acute toxicity of 9 frequently used OPFRs were studied with zebrafish embryos (2-96 hpf). By comparing the LC50 and EC50 (pericardium edema) data, two aryl-OPFRs, triphenyl phosphate (TPhP) and cresyl diphenyl phosphate (CDP) showed greater heart developmental toxicity than the others. It was also found that the acute toxicity of OPFRs varied mainly depending on their hydrophobicity. Further study on the cardiotoxicity of TPhP and CDP showed that the cardiac looping progress can be impeded by 0.10mg/L TPhP or CDP exposure. Bradycardia and reduction of myocardium were also observed in 0.50 and 1.0mg/L TPhP groups and 0.10, 0.50, and 1.0mg/L CDP groups. 0-48 hpf is the vulnerable window of zebrafish cardiogenesis that can be easily affected by TPhP and CDP. RT-qPCR measurement on the expressions of key transcriptional regulators in cardiogenesis showed that BMP4, NKX2-5, and TBX5 were significantly inhibited at the exposure points of 12 hpf and 24 hpf which may be the internal factors related to the heart developmental toxicity. As zebrafish is a good model organism for human health study, the present results call for a greater attention to the health risk of fetus in pregnant women exposed to such OPFRs.
Collapse
Affiliation(s)
- Zhongkun Du
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, PR China.
| | - Guowei Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, PR China.
| | - Shixiang Gao
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, PR China.
| | - Zunyao Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
8
|
NKX2-5 mutations in an inbred consanguineous population: genetic and phenotypic diversity. Sci Rep 2015; 5:8848. [PMID: 25742962 DOI: 10.1038/srep08848] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/14/2015] [Indexed: 11/08/2022] Open
Abstract
NKX2-5 mutations are associated with different forms of congenital heart disease. Despite the knowledge gained from molecular and animal studies, genotype-phenotype correlations in humans are limited by the lack of large cohorts and the incomplete assessment of family members. We hypothesized that studying the role of NKX2-5 in inbred populations with homogeneous genetic backgrounds and high consanguinity rates such as Lebanon could help closing this gap. We sequenced NKX2-5 in 188 index CHD cases (25 with ASD). Five variants (three segregated in families) were detected in eleven families including the previously documented p.R25C variant, which was found in seven patients from different families, and in one healthy individual. In 3/5 familial dominant ASD cases, we identified an NKX2-5 mutation. In addition to the heterogeneity of NKX2-5 mutations, a diversity of phenotypes occurred within the families with predominant ASD and AV block. We did in fact identify a large prevalence of Sudden Cardiac Death (SCD) in families with truncating mutations, and two patients with coronary sinus disease. NKX2-5 is thus responsible for dominant familial ASD even in consanguineous populations, and a wide genetic and phenotypic diversity is characteristic of NKX2-5 mutations in the Lebanese population.
Collapse
|
9
|
Ma X, Chen Y, Zhao X, Chen J, Shen C, Yang S. Association study of TGFBR2 and miR-518 gene polymorphisms with age at natural menopause, premature ovarian failure, and early menopause among Chinese Han women. Medicine (Baltimore) 2014; 93:e93. [PMID: 25365407 PMCID: PMC4616299 DOI: 10.1097/md.0000000000000093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Age at natural menopause (ANM), a highly heritable phenotype, has been identified to be closely associated with major hormone-related diseases, including breast cancer and gynecological cancers. We previously identified an important role for the transforming growth factor, β receptor II (TGFBR2) gene polymorphisms in breast cancer susceptibility among Asian women. Considering the important role of ANM in breast carcinogenesis, we hypothesized that TGFBR2 signals were involved in the formation of natural menopause.In a population-based study of 1844 Chinese women, we evaluated the effect of the genetic polymorphisms of TGFBR2 and miR-518 to determine if they are associated with ANM, premature ovarian failure (POF), and early menopause (EM) risk.No significant differences in the distribution of body mass index, education levels, smoking, drinking, and hypertension were detected between POF and EM cases and controls except for POF cases that were older (P = 0.015) than controls and more likely to have dyslipidemia (P = 0.002). The results showed that miR-518 rs7256241 was significantly associated with ANM. The carriers of minor allele G of rs7256241 have significantly higher ANM than those of the major allele homozygotes TT (β = 0.385, P = 0.035). TGFBR2 rs3773661 was significantly associated with POF, with odds ratio (OR) (95% confidence intervals [CIs]) of 0.66 (0.47-0.94) associated with per minor allele C (P = 0.023). The quartiles of genetic risk score were significantly associated with POF (OR, 1.27; 95% CI, 1.02-1.58; Ptrend = 0.034). Sensitivity analyses confirmed the robustness of these findings and no significant interactions were detected.This study provides evidence to implicate TGFBR2 and miR-518 gene polymorphisms as novel susceptibility factors for ANM, POF, and EM in Asians. Further research on these genetic regions will enhance our understanding of the genetic basis of natural menopause.
Collapse
Affiliation(s)
- Xiangyu Ma
- Department of Epidemiology (XM), College of Preventive Medicine, Third Military Medical University, Chongqing; Department of Cardiology (YC, XZ, JC, SY), Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing; and Department of Epidemiology and Biostatistics (CS), School of Public Health, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | |
Collapse
|
10
|
Wu M, Zuo Z, Li B, Huang L, Chen M, Wang C. Effects of low-level hexabromocyclododecane (HBCD) exposure on cardiac development in zebrafish embryos. ECOTOXICOLOGY (LONDON, ENGLAND) 2013; 22:1200-1207. [PMID: 23903933 DOI: 10.1007/s10646-013-1107-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/18/2013] [Indexed: 06/02/2023]
Abstract
Hexabromocyclododecane (HBCD) is one of the most widely used brominated flame retardants. In the present study, zebrafish embryos were exposed to HBCD at the low concentrations of 0, 2, 20 and 200 nM. The results showed HBCD exposure resulted in an increase in heart rate and cardiac arrhythmia after exposure for 72 h, though the survival rate and the whole malformation rate were not significantly affected. These results demonstrated that the heart might be a target of HBCD. Low-level HBCD exposure may not share the same mechanisms as exposure to high concentrations, since no obvious increase of apoptotic cells around the heart was observed in the HBCD-treated groups. It was observed that the expression of Tbx5 and Nkx2.5 was significantly elevated by HBCD treatment in a dose-dependent manner using real-time quantitative PCR, which may be mainly responsible for the alteration of heart rate, given that Tbx5 and Nkx2.5 are two factors regulating ventricle conduction. The mRNA expression of RyR2 and Atp2a2b (SERCA2a) was up-regulated in the exposure group, which may be one of reasons to affect the normal heart rate, since SERCA2a and RyR2 play an important role in calcium ion transport of cadiomyocytes. However, HBCD exposure did not significantly change the expression of Actc1l, Tnnt2, and Myh6, which are mainly muscle contractile genes that play key roles in the formation of cardiac structure. These results were consistent with the lack of effect seen on the other measurements of cardiac function, end diastolic volume, end-systolic volume, stroke volume, and cardiac output.
Collapse
Affiliation(s)
- Meifang Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, People's Republic of China
| | | | | | | | | | | |
Collapse
|
11
|
Risebro CA, Petchey LK, Smart N, Gomes J, Clark J, Vieira JM, Yanni J, Dobrzynski H, Davidson S, Zuberi Z, Tinker A, Shui B, Tallini YI, Kotlikoff MI, Miquerol L, Schwartz RJ, Riley PR. Epistatic rescue of Nkx2.5 adult cardiac conduction disease phenotypes by prospero-related homeobox protein 1 and HDAC3. Circ Res 2012; 111:e19-31. [PMID: 22647876 DOI: 10.1161/circresaha.111.260695] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Nkx2.5 is one of the most widely studied cardiac-specific transcription factors, conserved from flies to man, with multiple essential roles in both the developing and adult heart. Specific dominant mutations in NKX2.5 have been identified in adult congenital heart disease patients presenting with conduction system anomalies and recent genome-wide association studies implicate the NKX2.5 locus, as causative for lethal arrhythmias ("sudden cardiac death") that occur at a frequency in the population of 1 in 1000 per annum worldwide. Haploinsufficiency for Nkx2.5 in the mouse phenocopies human conduction disease pathology yet the phenotypes, described in both mouse and man, are highly pleiotropic, implicit of unknown modifiers and/or factors acting in epistasis with Nkx2.5/NKX2.5. OBJECTIVE To identify bone fide upstream genetic modifier(s) of Nkx2.5/NKX2.5 function and to determine epistatic effects relevant to the manifestation of NKX2.5-dependent adult congenital heart disease. METHODS AND RESULTS A study of cardiac function in prospero-related homeobox protein 1 (Prox1) heterozygous mice, using pressure-volume loop and micromannometry, revealed rescue of hemodynamic parameters in Nkx2.5(Cre/+); Prox1(loxP/+) animals versus Nkx2.5(Cre/+) controls. Anatomic studies, on a Cx40(EGFP) background, revealed Cre-mediated knock-down of Prox1 restored the anatomy of the atrioventricular node and His-Purkinje network both of which were severely hypoplastic in Nkx2.5(Cre/+) littermates. Steady state surface electrocardiography recordings and high-speed multiphoton imaging, to assess Ca(2+) handling, revealed atrioventricular conduction and excitation-contraction were also normalized by Prox1 haploinsufficiency, as was expression of conduction genes thought to act downstream of Nkx2.5. Chromatin immunoprecipitation on adult hearts, in combination with both gain and loss-of-function reporter assays in vitro, revealed that Prox1 recruits the corepressor HDAC3 to directly repress Nkx2.5 via a proximal upstream enhancer as a mechanism for regulating Nkx2.5 function in adult cardiac conduction. CONCLUSIONS Here we identify Prox1 as a direct upstream modifier of Nkx2.5 in the maintenance of the adult conduction system and rescue of Nkx2.5 conduction disease phenotypes. This study is the first example of rescue of Nkx2.5 function and establishes a model for ensuring electrophysiological function within the adult heart alongside insight into a novel Prox1-HDAC3-Nkx2.5 signaling pathway for therapeutic targeting in conduction disease.
Collapse
|
12
|
Zhu K, Wang H, Gul Y, Zhao Y, Wang W, Liu S, Wang M. Expression characterization and the promoter activity analysis of zebrafish hdac4. FISH PHYSIOLOGY AND BIOCHEMISTRY 2012; 38:585-593. [PMID: 21773810 DOI: 10.1007/s10695-011-9540-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 07/04/2011] [Indexed: 05/31/2023]
Abstract
Histone deacetylase 4 (HDAC4) is an important modifier enzyme for chromatin remodeling and plays an essential role in regulating gene expression. Spatio-temporal expression spectrum revealed that zebrafish hdac4 mRNA, ubiquitously distributed in various tissues, were significantly higher at 36 hpf (hours post-fertilization) and 6 dpf (days post-fertilization) than other periods. Trichostatin A (TSA) inhibited the development of zebrafish embryos and transcription of hdac4 and mef2a (myocyte enhancer factor-2A). Moreover, five vectors containing different promoter regions of hdac4 were constructed in order to analyze promoter activity. The vector containing the region from -125 to +160 exhibited maximum luciferase activity that was approximately 30.3-fold and 58.9-fold higher than the control in two kinds of cells, respectively. By comparing the luciferase activities between the region from -302 to +30 and -698 to +30, it was suggested that the region between -698 and -302 might contain mild negative regulatory elements.
Collapse
Affiliation(s)
- Kecheng Zhu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
13
|
Jongbloed MRM, Vicente-Steijn R, Douglas YL, Wisse LJ, Mori K, Yokota Y, Bartelings MM, Schalij MJ, Mahtab EA, Poelmann RE, Gittenberger-De Groot AC. Expression of Id2 in the second heart field and cardiac defects in Id2 knock-out mice. Dev Dyn 2012; 240:2561-77. [PMID: 22012595 DOI: 10.1002/dvdy.22762] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The inhibitor of differentiation Id2 is expressed in mesoderm of the second heart field, which contributes myocardial and mesenchymal cells to the primary heart tube. The role of Id2 in cardiac development is insufficiently known. Heart development was studied in sequential developmental stages in Id2 wildtype and knockout mouse embryos. Expression patterns of Id2, MLC-2a, Nkx2.5, HCN4, and WT-1 were analyzed. Id2 is expressed in myocardial progenitor cells at the inflow and outflow tract, in the endocardial and epicardial lineage, and in neural crest cells. Id2 knockout embryos show severe cardiac defects including abnormal orientation of systemic and pulmonary drainage, abnormal myocardialization of systemic and pulmonary veins, hypoplasia of the sinoatrial node, large interatrial communications, ventricular septal defects, double outlet right ventricle, and myocardial hypoplasia. Our results indicate a role for Id2 in the second heart field contribution at both the arterial and the venous poles of the heart.
Collapse
Affiliation(s)
- M R M Jongbloed
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Gonzales C, Ullrich ND, Gerber S, Berthonneche C, Niggli E, Pedrazzini T. Isolation of cardiovascular precursor cells from the human fetal heart. Tissue Eng Part A 2011; 18:198-207. [PMID: 21902604 DOI: 10.1089/ten.tea.2011.0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Weakening of cardiac function in patients with heart failure results from a loss of cardiomyocytes in the damaged heart. Cell replacement therapies as a way to induce myocardial regeneration in humans could represent attractive alternatives to classical drug-based approaches. However, a suitable source of precursor cells, which could produce a functional myocardium after transplantation, remains to be identified. In the present study, we isolated cardiovascular precursor cells from ventricles of human fetal hearts at 12 weeks of gestation. These cells expressed Nkx2.5 but not late cardiac markers such as α-actinin and troponin I. In addition, proliferating cells expressed the mesenchymal stem cell markers CD73, CD90, and CD105. Evidence for functional cardiogenic differentiation in vitro was demonstrated by the upregulation of cardiac gene expression as well as the appearance of cells with organized sarcomeric structures. Importantly, differentiated cells presented spontaneous and triggered calcium signals. Differentiation into smooth muscle cells was also detected. In contrast, precursor cells did not produce endothelial cells. The engraftment and differentiation capacity of green fluorescent protein (GFP)-labeled cardiac precursor cells were then tested in vivo after transfer into the heart of immunodeficient severe combined immunodeficient mice. Engrafted human cells were readily detected in the mouse myocardium. These cells retained their cardiac commitment and differentiated into α-actinin-positive cardiomyocytes. Expression of connexin-43 at the interface between GFP-labeled and endogenous cardiomyocytes indicated that precursor-derived cells connected to the mouse myocardium. Together, these results suggest that human ventricular nonmyocyte cells isolated from fetal hearts represent a suitable source of precursors for cell replacement therapies.
Collapse
Affiliation(s)
- Christine Gonzales
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
15
|
Sizarov A, Devalla HD, Anderson RH, Passier R, Christoffels VM, Moorman AFM. Molecular analysis of patterning of conduction tissues in the developing human heart. Circ Arrhythm Electrophysiol 2011; 4:532-42. [PMID: 21576278 DOI: 10.1161/circep.111.963421] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Recent studies in experimental animals have revealed some molecular mechanisms underlying the differentiation of the myocardium making up the conduction system. To date, lack of gene expression data for the developing human conduction system has precluded valid extrapolations from experimental studies to the human situation. METHODS AND RESULTS We performed immunohistochemical analyses of the expression of key transcription factors, such as ISL1, TBX3, TBX18, and NKX2-5, ion channel HCN4, and connexins in the human embryonic heart. We supplemented our molecular analyses with 3-dimensional reconstructions of myocardial TBX3 expression. TBX3 is expressed in the developing conduction system and in the right venous valve, atrioventricular ring bundles, and retro-aortic nodal region. TBX3-positive myocardium, with exception of the top of the ventricular septum, is devoid of fast-conducting connexin40 and connexin43 and hence identifies slowly conducting pathways. In the early embryonic heart, we found wide expression of the pacemaker channel HCN4 at the venous pole, including the atrial chambers. HCN4 expression becomes confined during later developmental stages to the components of the conduction system. Patterns of expression of transcription factors, known from experimental studies to regulate the development of the sinus node and atrioventricular conduction system, are similar in the human and mouse developing hearts. CONCLUSIONS Our findings point to the comparability of mechanisms governing the development of the cardiac conduction patterning in human and mouse, which provide a molecular basis for understanding the functioning of the human developing heart before formation of a discrete conduction system.
Collapse
Affiliation(s)
- Aleksander Sizarov
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
16
|
Kawashima T, Sasaki H. Gross anatomy of the human cardiac conduction system with comparative morphological and developmental implications for human application. Ann Anat 2011; 193:1-12. [DOI: 10.1016/j.aanat.2010.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 10/03/2010] [Accepted: 11/02/2010] [Indexed: 11/26/2022]
|
17
|
Boogerd CJJ, Moorman AFM, Barnett P. Expression of muscle segment homeobox genes in the developing myocardium. Anat Rec (Hoboken) 2010; 293:998-1001. [PMID: 20225205 DOI: 10.1002/ar.21112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Msx1 and Msx2 are essential for the development of many organs. In the heart, they act redundantly in development of the cardiac cushions. Additionally, Msx2 is expressed in the developing conduction system. However, the exact expression of Msx1 has not been established. We show that Msx1 is expressed in the cardiac cushions, but not in the myocardium. In Msx2-null mice, Msx1 is not ectopically expressed in the myocardium. The absence of myocardial defects in the Msx2 knock-out can therefore not be attributed to a redundant action of Msx1 in the myocardium.
Collapse
Affiliation(s)
- Cornelis J J Boogerd
- Heart Failure Research Center, Academic Medical Center of Amsterdam, Amsterdam, The Netherlands
| | | | | |
Collapse
|
18
|
Antonella Cecchetto, Alessandra Rampazzo, Annalisa Angelini,. From molecular mechanisms of cardiac development to genetic substrate of congenital heart diseases. Future Cardiol 2010; 6:373-93. [DOI: 10.2217/fca.10.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Congenital heart disease is one of the most important chapters in medicine because its incidence is increasing and nowadays it is close to 1.2%. Most congenital heart disorders are the result of defects during embryogenesis, which implies that they are due to alterations in genes involved in cardiac development. This review summarizes current knowledge regarding the molecular mechanisms involved in cardiac development in order to clarify the genetic basis of congenital heart disease.
Collapse
|
19
|
Abstract
Diseases of the cardiovascular system that cause sudden cardiac deaths are often caused by lethal arrhythmias that originate from defects in the cardiac conduction system. Development of the cardiac conduction system is a complex biological process that can be wrought with problems. Although several genes involved in mature conduction system function have been identified, their association with development of specific subcomponents of the cardiac conduction system remains challenging. Several transcription factors, including homeodomain proteins and T-box proteins, are essential for cardiac conduction system morphogenesis and activation or repression of key regulatory genes. In addition, several transcription factors modify expression of genes encoding the ion channel proteins that contribute to the electrophysiological properties of the conduction system and govern contraction of the surrounding myocardium. Loss of transcriptional regulation during cardiac development has detrimental effects on cardiogenesis that may lead to arrhythmias. Human genetic mutations in some of these transcription factors have been identified and are known to cause congenital heart diseases that include cardiac conduction system malformations. In this review, we summarize the contributions of several key transcription factors to specification, patterning, maturation, and function of the cardiac conduction system. Further analysis of the molecular programs involved in this process should lead to improved diagnosis and therapy of conduction system disease.
Collapse
Affiliation(s)
- Cathy J Hatcher
- Center for Molecular Cardiology, Greenberg Division of Cardiology, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | | |
Collapse
|
20
|
Belvís R, Tizzano EF, Martí-Fàbregas J, Leta RG, Baena M, Carreras F, Pons-Lladó G, Baiget M, Martí-Vilalta JL. Mutations in the NKX2-5 gene in patients with stroke and patent foramen ovale. Clin Neurol Neurosurg 2009; 111:574-8. [DOI: 10.1016/j.clineuro.2009.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2008] [Revised: 04/20/2009] [Accepted: 04/21/2009] [Indexed: 11/25/2022]
|
21
|
Bakker ML, Boukens BJ, Mommersteeg MTM, Brons JF, Wakker V, Moorman AFM, Christoffels VM. Transcription factor Tbx3 is required for the specification of the atrioventricular conduction system. Circ Res 2008; 102:1340-9. [PMID: 18467625 DOI: 10.1161/circresaha.107.169565] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cardiac conduction system consists of distinctive heart muscle cells that initiate and propagate the electric impulse required for coordinated contraction. The conduction system expresses the transcriptional repressor Tbx3, which is required for vertebrate development and controls the formation of the sinus node. In humans, mutations in Tbx3 cause ulnar-mammary syndrome. Here, we investigated the role of Tbx3 in the molecular specification of the atrioventricular conduction system. Expression analysis revealed early delineation of the atrioventricular bundle and proximal bundle branches by Tbx3 expression in human, mouse, and chicken. Tbx3-deficient mice, which die between embryonic day 12.5 and 15.5, ectopically expressed genes for connexin (Cx)43, atrial natriuretic factor (Nppa), Tbx18, and Tbx20 in the atrioventricular bundle and proximal bundle branches. Cx40 was precociously upregulated in the atrioventricular bundle of Tbx3 mutants. Moreover, the atrioventricular bundle and branches failed to exit the cell cycle in Tbx3 mutant embryos. Finally, Tbx3-deficient embryos developed outflow tract malformations and ventricular septal defects. These data reveal that Tbx3 is required for the molecular specification of the atrioventricular bundle and bundle branches and for the development of the ventricular septum and outflow tract. Our data suggest a mechanism in which Tbx3 represses differentiation into ventricular working myocardium, thereby imposing the conduction system phenotype on cells within its expression domain.
Collapse
Affiliation(s)
- Martijn L Bakker
- Heart Failure Research Center, Academic Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
Moskowitz IPG, Kim JB, Moore ML, Wolf CM, Peterson MA, Shendure J, Nobrega MA, Yokota Y, Berul C, Izumo S, Seidman JG, Seidman CE. A molecular pathway including Id2, Tbx5, and Nkx2-5 required for cardiac conduction system development. Cell 2007; 129:1365-76. [PMID: 17604724 DOI: 10.1016/j.cell.2007.04.036] [Citation(s) in RCA: 211] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2006] [Revised: 12/15/2006] [Accepted: 04/10/2007] [Indexed: 10/23/2022]
Abstract
The cardiac conduction system is an anatomically discrete segment of specialized myocardium that initiates and propagates electrical impulses to coordinate myocardial contraction. To define the molecular composition of the mouse ventricular conduction system we used microdissection and transcriptional profiling by serial analysis of gene expression (SAGE). Conduction-system-specific expression for Id2, a member of the Id gene family of transcriptional repressors, was identified. Analyses of Id2-deficient mice demonstrated structural and functional conduction system abnormalities, including left bundle branch block. A 1.2 kb fragment of the Id2 promoter proved sufficient for cooperative regulation by Nkx2-5 and Tbx5 in vitro and for conduction-system-specific gene expression in vivo. Furthermore, compound haploinsufficiency of Tbx5 and Nkx2-5 or Tbx5 and Id2 prevented embryonic specification of the ventricular conduction system. We conclude that a molecular pathway including Tbx5, Nkx2-5, and Id2 coordinates specification of ventricular myocytes into the ventricular conduction system lineage.
Collapse
|
24
|
Meysen S, Marger L, Hewett KW, Jarry-Guichard T, Agarkova I, Chauvin JP, Perriard JC, Izumo S, Gourdie RG, Mangoni ME, Nargeot J, Gros D, Miquerol L. Nkx2.5 cell-autonomous gene function is required for the postnatal formation of the peripheral ventricular conduction system. Dev Biol 2007; 303:740-53. [PMID: 17250822 DOI: 10.1016/j.ydbio.2006.12.044] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Revised: 11/16/2006] [Accepted: 12/07/2006] [Indexed: 11/26/2022]
Abstract
The ventricular conduction system is responsible for rapid propagation of electrical activity to coordinate ventricular contraction. To investigate the role of the transcription factor Nkx2.5 in the morphogenesis of the ventricular conduction system, we crossed Nkx2.5(+/-) mice with Cx40(eGFP/+) mice in which eGFP expression permits visualization of the His-Purkinje conduction system. Major anatomical and functional disturbances were detected in the His-Purkinje system of adult Nkx2.5(+/-)/Cx40(eGFP/+) mice, including hypoplasia of eGFP-positive Purkinje fibers and the disorganization of the Purkinje fiber network in the ventricular apex. Although the action potential properties of the individual eGFP-positive cells were normal, the deficiency of Purkinje fibers in Nkx2.5 haploinsufficient mice was associated with abnormalities of ventricular electrical activation, including slowed and decremented conduction along the left bundle branch. During embryonic development, eGFP expression in the ventricular trabeculae of Nkx2.5(+/-) hearts was qualitatively normal, with a measurable deficiency in eGFP-positive cells being observed only after birth. Chimeric analyses showed that maximal Nkx2.5 levels are required cell-autonomously. Reduced Nkx2.5 levels are associated with a delay in cell cycle withdrawal in surrounding GFP-negative myocytes. Our results suggest that the formation of the peripheral conduction system is time- and dose-dependent on the transcription factor Nkx2.5 that is cell-autonomously required for the postnatal differentiation of Purkinje fibers.
Collapse
Affiliation(s)
- Sonia Meysen
- Institut de Biologie du Développement de Marseille-Luminy, IBDML, Université de la Méditerranée, CNRS UMR6216, Campus de Luminy, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The cardiac conduction system (CCS) is a specialized tissue network that initiates and maintains a rhythmic heartbeat. The CCS consists of several functional subcomponents responsible for producing a pacemaking impulse and distributing action potentials across the heart in a coordinated manner. The formation of the distinct subcomponents of the CCS occurs within a precise temporal and spatial framework; thereby assuring that as the system matures from a tubular to a complex chambered organ, a rhythmic heartbeat is always maintained. Therefore, a defect in differentiation of any CCS component would lead to severe rhythm disturbances. Recent molecular, cell biological and physiological approaches have provided fresh and unexpected perspectives of the relationships between cell fate, gene expression and differentiation of specialized function within the developing myocardium. In particular, biomechanical forces created by the heartbeat itself have important roles in the inductive patterning and functional integration of the developing conduction system. This new understanding of the cellular origin and molecular induction of CCS tissues during embryogenesis may provide the foundation for tissue engineering, replacement and repair of these essential cardiac tissues in the future.
Collapse
Affiliation(s)
- Takashi Mikawa
- University of California San Francisco, Cardiovascular Research Institute, Box 2711, Rock Hall Room 384D, 1550 4th Street, San Francisco, CA 94158-2324, United States.
| | | |
Collapse
|
26
|
Dunwoodie SL. Combinatorial signaling in the heart orchestrates cardiac induction, lineage specification and chamber formation. Semin Cell Dev Biol 2007; 18:54-66. [PMID: 17236794 DOI: 10.1016/j.semcdb.2006.12.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The complexity of mammalian cardiogenesis is compounded, as the heart must function in the embryo whilst it is still being formed. Great advances have been made recently as additional cardiac progenitor cell populations have been identified. The induction and maintenance of these progenitors, and their deployment to the developing heart relies on combinatorial molecular signalling, a feature also of cardiac chamber formation. Many forms of congenital heart disease in humans are likely to arise from defects in the early stages of heart development; therefore it is important to understand the molecular pathways that underlie some of the key events that shape the heart during the early stages of it development.
Collapse
Affiliation(s)
- Sally L Dunwoodie
- Developmental Biology Program, Victor Chang Cardiac Research Institute, 384 Victoria Street, Darlinghurst, NSW, Australia.
| |
Collapse
|
27
|
Gittenberger-de Groot AC, Mahtab EAF, Hahurij ND, Wisse LJ, Deruiter MC, Wijffels MCEF, Poelmann RE. Nkx2.5-negative myocardium of the posterior heart field and its correlation with podoplanin expression in cells from the developing cardiac pacemaking and conduction system. Anat Rec (Hoboken) 2007; 290:115-22. [PMID: 17441204 DOI: 10.1002/ar.20406] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Recent advances in the study of cardiac development have shown the relevance of addition of myocardium to the primary myocardial heart tube. In wild-type mouse embryos (E9.5-15.5), we have studied the myocardium at the venous pole of the heart using immunohistochemistry and 3D reconstructions of expression patterns of MLC-2a, Nkx2.5, and podoplanin, a novel coelomic and myocardial marker. Podoplanin-positive coelomic epithelium was continuous with adjacent podoplanin- and MLC-2a-positive myocardium that formed a conspicuous band along the left cardinal vein extending through the base of the atrial septum to the posterior myocardium of the atrioventricular canal, the atrioventricular nodal region, and the His-Purkinje system. Later on, podoplanin expression was also found in the myocardium surrounding the pulmonary vein. On the right side, podoplanin-positive cells were seen along the right cardinal vein, which during development persisted in the sinoatrial node and part of the venous valves. In the MLC-2a- and podoplanin-positive myocardium, Nkx2.5 expression was absent in the sinoatrial node and the wall of the cardinal veins. There was a mosaic positivity in the wall of the common pulmonary vein and the atrioventricular conduction system as opposed to the overall Nkx2.5 expression seen in the chamber myocardium. We conclude that we have found podoplanin as a marker that links a novel Nkx2.5-negative sinus venosus myocardial area, which we refer to as the posterior heart field, with the cardiac conduction system.
Collapse
|
28
|
Chau MDL, Tuft R, Fogarty K, Bao ZZ. Notch signaling plays a key role in cardiac cell differentiation. Mech Dev 2006; 123:626-40. [PMID: 16843648 PMCID: PMC1567976 DOI: 10.1016/j.mod.2006.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2006] [Revised: 05/22/2006] [Accepted: 06/07/2006] [Indexed: 11/17/2022]
Abstract
Results from lineage tracing studies indicate that precursor cells in the ventricles give rise to both cardiac muscle and conduction cells. Cardiac conduction cells are specialized cells responsible for orchestrating the rhythmic contractions of the heart. Here, we show that Notch signaling plays an important role in the differentiation of cardiac muscle and conduction cell lineages in the ventricles. Notch1 expression coincides with a conduction marker, HNK-1, at early stages. Misexpression of constitutively active Notch1 (NIC) in early heart tubes in chick exhibited multiple effects on cardiac cell differentiation. Cells expressing NIC had a significant decrease in expression of cardiac muscle markers, but an increase in expression of conduction cell markers, HNK-1, and SNAP-25. However, the expression of the conduction marker connexin 40 was inhibited. Loss-of-function study, using a dominant-negative form of Suppressor-of-Hairless, further supports that Notch1 signaling is important for the differentiation of these cardiac cell types. Functional studies show that the expression of constitutively active Notch1 resulted in abnormalities in ventricular conduction pathway patterns.
Collapse
Affiliation(s)
- Mary D L Chau
- Department of Medicine and Cell Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
29
|
Abstract
Although there have been important advances in diagnostic modalities and therapeutic strategies for congenital heart defects (CHD), these malformations still lead to significant morbidity and mortality in the human population. Over the past 10 years, characterization of the genetic causes of CHD has begun to elucidate some of the molecular causes of these defects. Linkage analysis and candidate-gene approaches have been used to identify gene mutations that are associated with both familial and sporadic cases of CHD. Complementation of the human studies with developmental studies in mouse models provides information for the roles of these genes in normal development as well as indications for disease pathogenesis. Biochemical analysis of these gene mutations has provided further insight into the molecular effects of these genetic mutations. Here we review genetic, developmental, and biochemical studies of six cardiac transcription factors that have been identified as genetic causes for CHD in humans.
Collapse
Affiliation(s)
- Krista L Clark
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA.
| | | | | |
Collapse
|
30
|
Graham V, Zhang H, Willis S, Creazzo TL. Expression of a two-pore domain K+ channel (TASK-1) in developing avian and mouse ventricular conduction systems. Dev Dyn 2006; 235:143-51. [PMID: 16145663 DOI: 10.1002/dvdy.20558] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this study, we report the identification and amino acid sequence of a novel two-pore domain potassium channel (TASK-1) in chicken. This protein, cTASK-1, is highly similar to mouse and human TASK-1 particularly within the pore regions. We describe the expression profile of both chicken and mouse TASK-1 in the embryonic heart as the ventricular conduction system develops. The developmental distribution of TASK-1 is similar in chicken and mouse. Initially, TASK-1 is expressed throughout the myocardium of the early heart tube. However, as cardiogenesis proceeds, ventricular expression becomes restricted to the trabeculated myocardium and eventually the bundle of His, bundle branches, and Purkinje fibers of the mature conduction system. This finding suggests that components of the ventricular conduction system differentiate from TASK-1-positive myocytes of the early heart tube that retain TASK-1 expression as they mature. Our results are consistent with a common mechanism for ventricular conduction system development in avians and mammals, despite differences in the anatomy of the mature conduction systems of these organisms.
Collapse
Affiliation(s)
- Victoria Graham
- Department of Pediatrics/Neonatology, Neonatal/Perinatal Research Institute, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | |
Collapse
|
31
|
Harris BS, Spruill L, Edmonson AM, Rackley MS, Benson DW, O’Brien TX, Gourdie RG. Differentiation of cardiac Purkinje fibers requires precise spatiotemporal regulation of Nkx2-5 expression. Dev Dyn 2006; 235:38-49. [PMID: 16245335 PMCID: PMC2610391 DOI: 10.1002/dvdy.20580] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Nkx2-5 gene mutations cause cardiac abnormalities, including deficits of function in the atrioventricular conduction system (AVCS). In the chick, Nkx2-5 is elevated in Purkinje fiber AVCS cells relative to working cardiomyocytes. Here, we show that Nkx2-5 expression rises to a peak as Purkinje fibers progressively differentiate. To disrupt this pattern, we overexpressed Nkx2-5 from embryonic day 10, as Purkinje fibers are recruited within developing chick hearts. Overexpression of Nkx2-5 caused inhibition of slow tonic myosin heavy chain protein (sMHC), a late Purkinje fiber marker but did not affect Cx40 levels. Working cardiomyocytes overexpressing Nkx2-5 in these hearts ectopically up-regulated Cx40 but not sMHC. Isolated embryonic cardiomyocytes overexpressing Nkx2-5 also displayed increased Cx40 and suppressed sMHC. By contrast, overexpression of a human NKX2-5 mutant did not effect these markers in vivo or in vitro, suggesting one possible mechanism for clinical phenotypes. We conclude that a prerequisite for normal Purkinje fiber maturation is precise regulation of Nkx2-5 levels.
Collapse
Affiliation(s)
- Brett S. Harris
- Departments of Cell Biology and Anatomy, Medical University of South Carolina, Charleston SC
- Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC
| | - Laura Spruill
- Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC
| | - Angela M. Edmonson
- Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC
- Medical Research Service, Ralph H. Johnson Department of Veteran Affairs Medical Center, Medical University of South Carolina, Charleston SC
| | - Mary S. Rackley
- Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC
- Medical Research Service, Ralph H. Johnson Department of Veteran Affairs Medical Center, Medical University of South Carolina, Charleston SC
| | - D. Woodrow Benson
- Department of Molecular and Cardiovascular Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Terrence X. O’Brien
- Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston SC
- Medical Research Service, Ralph H. Johnson Department of Veteran Affairs Medical Center, Medical University of South Carolina, Charleston SC
| | - Robert G. Gourdie
- Departments of Cell Biology and Anatomy, Medical University of South Carolina, Charleston SC
| |
Collapse
|
32
|
Akazawa H, Komuro I. Cardiac transcription factor Csx/Nkx2-5: Its role in cardiac development and diseases. Pharmacol Ther 2005; 107:252-68. [PMID: 15925411 DOI: 10.1016/j.pharmthera.2005.03.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2005] [Indexed: 11/20/2022]
Abstract
During the past decade, an emerging body of evidence has accumulated that cardiac transcription factors control a cardiac gene program and play a critical role in transcriptional regulation during cardiogenesis and during the adaptive process in adult hearts. Especially, an evolutionally conserved homeobox transcription factor Csx/Nkx2-5 has been in the forefront in the field of cardiac biology, providing molecular insights into the mechanisms of cardiac development and diseases. Csx/Nkx2-5 is indispensable for normal cardiac development, and mutations of the gene are associated with human congenital heart diseases (CHD). In the present review, the regulation of a cardiac gene program by Csx/Nkx2-5 is summarized, with an emphasis on its role in the cardiac development and diseases.
Collapse
Affiliation(s)
- Hiroshi Akazawa
- Division of Cardiovascular Pathophysiology and Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | | |
Collapse
|
33
|
|
34
|
Yu Z, Li Y, Meng Q, Yuan J, Zhao Z, Li W, Hu X, Yan B, Fan B, Yu S, Li N. Comparative analysis of the pig BAC sequence involved in the regulation of myostatin gene. ACTA ACUST UNITED AC 2005; 48:168-80. [PMID: 15986890 DOI: 10.1007/bf02879670] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Myostatin (GDF8, MSTN) is a member of the transforming growth factor beta superfamily that is essential for proper regulation of skeletal muscle mass. In order to study its expression and regulatory mechanism deeply, we have presented a comparative analysis of about 170-kb pig BAC sequence containing the myostatin gene among pig, human and mouse. The genomic region is characterized by high interspersed repeats and low G+C content. As for the myostatin gene, a higher sequence similarity is found between human and pig than between these species and the mouse. One striking feature is that the structure of two TATA-boxes in the nearby downstream of CCAAT-box is identified in the promoter. Further analysis reveals that the TATA-box1 is responsible for the transcription in pig and human, but the TATA-box2 acts on the transcription in mouse. The other interesting feature is that two polyadenylation signal sequences (AATAAA) exist in 3'UTR of the pig myostatin gene. Moreover, a large number of potential transcription factor-binding sites are also identified in evolutionary conserved regions (ECRs), which may be associated with the regulation of myostatin. Many putative transcription factors play an important role in the muscle development, and the complex interaction between myostatin and these factors may be required for proper muscle development.
Collapse
Affiliation(s)
- Zhengquan Yu
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing 100094, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Blom NA, Ottenkamp J, Deruiter MC, Wenink ACG, Gittenberger-de Groot AC. Development of the cardiac conduction system in atrioventricular septal defect in human trisomy 21. Pediatr Res 2005; 58:516-20. [PMID: 16148066 DOI: 10.1203/01.pdr.0000179388.10921.44] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In patients with atrioventricular septal defect (AVSD), the occurrence of nonsurgical AV block has been reported. We have looked for an explanation in the development of the AV conduction system. Human embryos with AVSD and trisomy 21 and normal embryos were examined (age 5-16 wk gestation). Antibodies to human natural killer cell-1 (HNK-1), muscle actin (HHF-35), and collagen VI were used to delineate the conduction system. As in normal hearts, HNK-1 transiently stains the AV conduction system, the sinoatrial node, and parts of the sinus venosus in AVSD. A large distance is present between the superior and inferior node-like part of the right AV ring bundle, comparable to 6-wk-old normal hearts. The definitive inferior AV node remains in dorsal position from 7 wk onward and does not appose to the superior node-like part as seen in normal hearts. Furthermore, in AVSD, a transient third HNK-1-positive "middle bundle" branch that is continuous with the retroaortic root branch and the superior node-like part can be identified, and thus the AV conduction system forms a figure-of-eight loop. At later stages, the AV node remains in dorsal position close to the coronary sinus ostium with a long nonbranching bundle that runs through thin fibrous tissue toward the ventricular septum. The formation of the AV node and the ventricular conduction system in AVSD and Down syndrome differs from normal development, which can be a causative factor in the development of AV conduction disturbances.
Collapse
Affiliation(s)
- Nico A Blom
- Department of Pediatric Cardiology, Leiden University Medical Center, The Netherlands
| | | | | | | | | |
Collapse
|
36
|
Patel R, Kos L. Endothelin-1 and Neuregulin-1 convert embryonic cardiomyocytes into cells of the conduction system in the mouse. Dev Dyn 2005; 233:20-8. [PMID: 15759273 DOI: 10.1002/dvdy.20284] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The cells that form the cardiac conduction system (CCS) are recruited from embryonic cardiomyocytes. Endothelin-1 (ET-1) and Neuregulin-1 (NRG-1) have been associated with this transition in the avian and murine systems, respectively. We established murine embryonic cardiomyocyte cultures induced or not with ET-1 and/or NRG-1 to compare the expression of cardiomyocyte and CCS-specific genes. Semiquantitative reverse transcription-polymerase chain reaction analysis showed that cardiomyogenesis and CCS-specific markers, such as Nkx2.5, GATA4, Irx4, Connexin 40, Connexin 45, HF-1b, and MinK, were up-regulated in the presence of either growth factor. Additionally, immunofluorescence analysis demonstrated that ET-1 or NRG-1 increased the number of cells expressing the Purkinje fiber-specific marker Connexin 40 in induced cultures but did not selectively increase their proliferation rate. Interestingly, additive effects were not observed in ET-1 and NRG-1 combination treatments. Among other possibilities, this observation suggests that these factors may interact to promote the differentiation of the murine CCS.
Collapse
Affiliation(s)
- Rita Patel
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | | |
Collapse
|
37
|
Ismat FA, Zhang M, Kook H, Huang B, Zhou R, Ferrari VA, Epstein JA, Patel VV. Homeobox protein Hop functions in the adult cardiac conduction system. Circ Res 2005; 96:898-903. [PMID: 15790958 PMCID: PMC1615918 DOI: 10.1161/01.res.0000163108.47258.f3] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hop is an unusual homeobox gene expressed in the embryonic and adult heart. Hop acts downstream of Nkx2-5 during development, and Nkx2-5 mutations are associated with cardiac conduction system (CCS) defects. Inactivation of Hop in the mouse is lethal in half of the expected null embryos. Here, we show that Hop is expressed strongly in the adult CCS. Hop-/- adult mice display conduction defects below the atrioventricular node (AVN) as determined by invasive electrophysiological testing. These defects are associated with decreased expression of connexin40. Our results suggest that Hop functions in the adult CCS and demonstrate conservation of molecular hierarchies between embryonic myocardium and the specialized conduction tissue of the mature heart.
Collapse
Affiliation(s)
- Fraz A. Ismat
- From the Division of CardiologyDepartment of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pa
| | | | - Hyun Kook
- the Department of Pharmacology and Medical Research Center for Gene Regulation Chonnam National University Medical School, Gwangju, South Korea
| | | | - Rong Zhou
- Department of RadiologyUniversity of Pennsylvania, Philadelphia, Pa
| | | | | | | |
Collapse
|
38
|
Jay PY, Maguire CT, Wakimoto H, Izumo S, Berul CI. Absence of Msx2 Does Not Affect Cardiac Conduction or Rescue Conduction Defects Associated with Nkx2-5 Mutation. J Cardiovasc Electrophysiol 2005; 16:82-5. [PMID: 15673394 DOI: 10.1046/j.1540-8167.2005.04365.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The transcription factors governing embryonic development of the AV conduction system are largely unknown. Heterozygous mutations of the cardiac transcription factor Nkx2-5 cause AV conduction defects, which are associated with anatomic hypoplasia of the conduction system. In situ expression patterns of Msx2 in the mouse and chick embryonic heart have suggested a developmental function for this transcription factor. Homozygous Nkx2-5 knockout mouse embryos express Msx2 ectopically throughout the myocardium, suggesting Msx2 affects conduction system development through a transcriptional cascade starting with Nkx2-5. Several observations support a model in which Msx2 negatively regulates formation of the conduction system and inappropriate Msx2 up-regulation causes the conduction defects associated with Nkx2-5 mutation. METHODS AND RESULTS We obtained surface ECGs and performed intracardiac electrophysiologic studies in Msx2 knockout mice and in Nkx2-5 wild-type and heterozygous null mutant mice in an Msx2 null mutant background. Msx2 null mutant mice had normal cardiac conduction and no increased vulnerability to inducible arrhythmia. Absence of Msx2 did not alter the conduction defects observed in heterozygous Nkx2-5 knockout mice. CONCLUSION Msx2 likely does not contribute to development of the conduction system. Abnormal Msx2 expression likely does not cause the AV conduction defects present in Nkx2-5 knockout mice.
Collapse
Affiliation(s)
- Patrick Y Jay
- Department of Cardiology, Children's Hospital, Boston, Massachusetts 02139, USA.
| | | | | | | | | |
Collapse
|
39
|
Harris BS, Gourdie RG, O'Brien TX. Atrioventricular Conduction System and Transcription Factors Nkx2.5 and Msx2. J Cardiovasc Electrophysiol 2005; 16:86-7. [PMID: 15673395 DOI: 10.1046/j.1540-8167.2005.04667.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Linhares VL, Almeida NA, Menezes DC, Elliott DA, Lai D, Beyer EC, Campos de Carvalho AC, Costa MW. Transcriptional regulation of the murine Connexin40 promoter by cardiac factors Nkx2-5, GATA4 and Tbx5. Cardiovasc Res 2004; 64:402-411. [PMID: 15537493 PMCID: PMC3252638 DOI: 10.1016/j.cardiores.2004.09.021] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2004] [Accepted: 09/28/2004] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE Connexin40 (Cx40) is a gap junction protein expressed specifically in developing and mature atrial myocytes and cells of the conduction system. In this report, we identify cis-acting elements within the mouse Cx40 promoter and unravel part of the complex pathways involved in the cardiac expression of this gene. METHODS To identify the factors involved in the cardiac expression of Cx40, we used transient transfections in mammalian cells coupled with electrophoretic mobility shift assays (EMSA) and RT-PCR. RESULTS Within the promoter region, we identified the minimal elements required for transcriptional activity within 150 base pairs (bp) upstream of the transcriptional start site. Several putative regulatory sites for transcription factors were predicted within this region by computer analysis, and we demonstrated that the nuclear factors Sp1, Nkx2-5, GATA4 and Tbx5 could interact specifically with elements present in the minimal promoter region of the Cx40. Furthermore, co-transfection experiments showed the ability of Nkx2-5 and GATA4 to transactivate the minimal Cx40 promoter while Tbx5 repressed Nkx2-5/GATA4-mediated activation. Mutagenesis of the Nkx2-5 core site in the Cx40 promoter led to significantly decreased activity in rat smooth muscle cell line A7r5. Consistent with this, mouse embryos lacking Nkx2-5 showed a marked decrease in Cx40 expression. CONCLUSION In this work, we cloned the promoter region of the Cx40 and demonstrated that the core promoter was modulated by cardiac transcriptional factors Nkx2-5, Tbx5 and GATA4 acting together with ubiquitous Sp1.
Collapse
Affiliation(s)
- Vania L.F. Linhares
- Laboratório de Cardiologia Celular e Molecular-Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 20941-900, Brazil
| | - Norma A.S. Almeida
- Laboratório de Cardiologia Celular e Molecular-Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 20941-900, Brazil
| | - Diego C. Menezes
- Laboratório de Cardiologia Celular e Molecular-Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 20941-900, Brazil
| | - David A. Elliott
- Developmental Biology Unit, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Donna Lai
- Developmental Biology Unit, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Eric C. Beyer
- Section of Pediatric Hematology/Oncology, Department of Pediatrics, University of Chicago, Chicago, IL 60637-1470, USA
| | - Antonio C. Campos de Carvalho
- Laboratório de Cardiologia Celular e Molecular-Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 20941-900, Brazil
| | - Mauro W. Costa
- Laboratório de Cardiologia Celular e Molecular-Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 20941-900, Brazil
- Developmental Biology Unit, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
41
|
Jay PY, Harris BS, Buerger A, Rozhitskaya O, Maguire CT, Barbosky LA, McCusty E, Berul CI, O'brien TX, Gourdie RG, Izumo S. Function follows form: cardiac conduction system defects in Nkx2-5 mutation. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2004; 280:966-72. [PMID: 15368343 DOI: 10.1002/ar.a.20102] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Mutations of Nkx2-5 cause congenital heart disease and atrioventricular block in man. The altered expression of an electrophysiologic protein regulated by Nkx2-5 was originally presumed to cause the conduction defect, but when no such protein was found, an alternative hypothesis was considered. In pediatric patients, the association of certain cardiac malformations with congenital atrioventricular block suggests that errors in specific developmental pathways could cause both an anatomic and a physiologic defect. We therefore hypothesized that Nkx2-5 insufficiency perturbs the conduction system during development, which in turn manifests as a postnatal conduction defect. Experimental results from Nkx2-5 knockout mouse models support the developmental hypothesis. Hypoplasia of the atrioventricular node, His bundle, and Purkinje system can explain in whole or in part specific conduction and electrophysiologic defects present in Nkx2-5 haploinsufficiency.
Collapse
Affiliation(s)
- Patrick Y Jay
- Department of Cardiology, Children's Hospital, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mjaatvedt CH, Klewer S, Wessels A, Gourdie RG. Een geslaagd jubileum! The 10th Weinstein Developmental Cardiovascular Conference in Leiden. Dev Dyn 2004; 231:655-9. [PMID: 15376316 DOI: 10.1002/dvdy.20169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Corey H Mjaatvedt
- Department of Cell Biology and Anatomy, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA.
| | | | | | | |
Collapse
|
43
|
Pashmforoush M, Lu JT, Chen H, Amand TS, Kondo R, Pradervand S, Evans SM, Clark B, Feramisco JR, Giles W, Ho SY, Benson DW, Silberbach M, Shou W, Chien KR. Nkx2-5 pathways and congenital heart disease; loss of ventricular myocyte lineage specification leads to progressive cardiomyopathy and complete heart block. Cell 2004; 117:373-86. [PMID: 15109497 DOI: 10.1016/s0092-8674(04)00405-2] [Citation(s) in RCA: 317] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2003] [Revised: 02/03/2004] [Accepted: 03/09/2004] [Indexed: 01/09/2023]
Abstract
Human mutations in Nkx2-5 lead to progressive cardiomyopathy and conduction defects via unknown mechanisms. To define these pathways, we generated mice with a ventricular-restricted knockout of Nkx2-5, which display no structural defects but have progressive complete heart block, and massive trabecular muscle overgrowth found in some patients with Nkx2-5 mutations. At birth, mutant mice display a hypoplastic atrioventricular (AV) node and then develop selective dropout of these conduction cells. Transcriptional profiling uncovered the aberrant expression of a unique panel of atrial and conduction system-restricted target genes, as well as the ectopic, high level BMP-10 expression in the adult ventricular myocardium. Further, BMP-10 is shown to be necessary and sufficient for a major component of the ventricular muscle defects. Accordingly, loss of ventricular muscle cell lineage specification into trabecular and conduction system myocytes is a new mechanistic pathway for progressive cardiomyopathy and conduction defects in congenital heart disease.
Collapse
Affiliation(s)
- Mohammad Pashmforoush
- UCSD Institute of Molecular Medicine, University of California San Diego School of Medicine, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jay PY, Harris BS, Maguire CT, Buerger A, Wakimoto H, Tanaka M, Kupershmidt S, Roden DM, Schultheiss TM, O'Brien TX, Gourdie RG, Berul CI, Izumo S. Nkx2-5 mutation causes anatomic hypoplasia of the cardiac conduction system. J Clin Invest 2004; 113:1130-7. [PMID: 15085192 PMCID: PMC385399 DOI: 10.1172/jci19846] [Citation(s) in RCA: 178] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2003] [Accepted: 02/17/2004] [Indexed: 11/17/2022] Open
Abstract
Heterozygous mutations of the cardiac transcription factor Nkx2-5 cause atrioventricular conduction defects in humans by unknown mechanisms. We show in KO mice that the number of cells in the cardiac conduction system is directly related to Nkx2-5 gene dosage. Null mutant embryos appear to lack the primordium of the atrioventricular node. In Nkx2-5 haploinsufficiency, the conduction system has half the normal number of cells. In addition, an entire population of connexin40(-)/connexin45(+) cells is missing in the atrioventricular node of Nkx2-5 heterozygous KO mice. Specific functional defects associated with Nkx2-5 loss of function can be attributed to hypoplastic development of the relevant structures in the conduction system. Surprisingly, the cellular expression of connexin40, the major gap junction isoform of Purkinje fibers and a putative Nkx2-5 target, is unaffected, consistent with normal conduction times through the His-Purkinje system measured in vivo. Postnatal conduction defects in Nkx2-5 mutation may result at least in part from a defect in the genetic program that governs the recruitment or retention of embryonic cardiac myocytes in the conduction system.
Collapse
Affiliation(s)
- Patrick Y Jay
- Department of Cardiology, Children's Hospital, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Atrioventricular (AV) conduction disease (block) describes impairment of the electrical continuity between the atria and ventricles. Classification of AV block has utilized biophysical characteristics, usually the extent (first, second, or third degree) and site of block (above or below His bundle recording site). The genetic significance of this classification is unknown. In young patients, AV block may result from injury or be the major cardiac manifestation of neuromuscular disease. However, in some cases, AV block has unknown or idiopathic cause. In such cases, familial clustering has been noted and published pedigrees show autosomal dominant inheritance; associated heart disease is common (e.g., congenital heart malformation, cardiomyopathy). The latter finding is not surprising given the common origin of working myocytes and specialized conduction system elements. Using genetic models incorporating reduced penetrance (disease absence in some individuals with disease gene), variable expressivity (individuals with disease gene have different phenotypes), and genetic heterogeneity (similar phenotypes, different genetic cause), molecular genetic causes of AV block are being identified. Mutations identified in genes with diverse functions (transcription, excitability, and energy homeostasis) for the first time provide the means to assess risk and offer insight into the molecular basis of this important clinical condition previously defined only by biophysical characteristics.
Collapse
Affiliation(s)
- D Woodrow Benson
- Division of Cardiology, ML7042, Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
46
|
Chuck ET, Meyers K, France D, Creazzo TL, Morley GE. Transitions in ventricular activation revealed by two-dimensional optical mapping. ACTA ACUST UNITED AC 2004; 280:990-1000. [PMID: 15372433 DOI: 10.1002/ar.a.20083] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
While cardiac function in the mature heart is dependent on a properly functioning His-Purkinje system, the early embryonic tubular heart efficiently pumps blood without a distinct specialized conduction system. Although His-Purkinje system precursors have been identified using immunohistological techniques in the looped heart, little is known whether these precursors function electrically. To address this question, we used high-resolution optical mapping and fluorescent dyes with two CCD cameras to describe the motion-corrected activation patterns of 76 embryonic chick hearts from tubular stages (stage 10) to mature septated hearts (stage 35). Ventricular activation in the tubular looped heart (stages 10-17) using both calcium-sensitive fluo-4 and voltage-sensitive di-4-ANEPPS shows sequentially uniform propagation. In late looped hearts (stages 18-22), domains of the dorsal and lateral ventricle are preferentially activated before spreading to the remaining myocardium and show alternating regions of fast and slow propagation. During stages 22-26, action potentials arise from the dorsal ventricle. By stages 27-29, action potential breakthrough is also observed at the right ventricle apex. By stage 31, activation of the heart proceeds from foci at the apex and dorsal surface of the heart. The breakthrough foci correspond to regions where putative conduction system precursors have been identified immunohistologically. To date, our study represents the most detailed electrophysiological characterization of the embryonic heart between the looped and preseptated stages and suggests that ventricular activation undergoes a gradual transformation from sequential to a mature pattern with right and left epicardial breakthroughs. Our investigation suggests that cardiac conduction system precursors may be electrophysiologically distinct and mature gradually throughout cardiac morphogenesis in the chick.
Collapse
Affiliation(s)
- Emil Thomas Chuck
- Neonatal-Perinatal Research Institute, Division of Neonatology, Department of Pediatrics and Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | |
Collapse
|
47
|
Harris BS, Jay PY, Rackley MS, Izumo S, O'brien TX, Gourdie RG. Transcriptional regulation of cardiac conduction system development: 2004 FASEB cardiac conduction system minimeeting, Washington, DC. ACTA ACUST UNITED AC 2004; 280:1036-45. [PMID: 15368344 DOI: 10.1002/ar.a.20101] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The development of the complex network of specialized cells that form the atrioventricular conduction system (AVCS) during cardiac morphogenesis occurs by progressive recruitment within a multipotent cardiomyogenic lineage. Understanding the molecular control of this developmental process has been the focus of recent research. Transcription factors representative of multiple subfamilies have been identified and include members of zinc-finger subfamilies (GATA4, GATA6 HF-1b), skeletal muscle transcription factors (MyoD), T-box genes (Tbx5), and also homeodomain transcription factors (Msx2 and Nkx2.5). Mutations in some of these transcription factors cause congenital heart disease and are associated with cardiac abnormalities, including deficits within the AVCS. Mouse models that closely phenocopy known human heart disease provide powerful tools for the study of molecular effectors of AVCS development. Indeed, investigations of the Nkx2.5 haploinsufficient mouse have shown that peripheral Purkinje fibers are significantly underrepresented. This piece of data corroborates our previous work showing in chick, mouse, and humans that Nkx2.5 is elevated in the differentiating AVCS relative to adjacent working ventricular myocardial tissues. Using the chick embryo as a model, we show that this elevation of Nkx2.5 is transient in the network of conduction cells comprising the peripheral Purkinje fiber system. Functional studies using defective adenoviral constructs, which disrupt the normal variation in level of this gene, result in perturbations of Purkinje fiber phenotype. Thus, the precise spatiotemporal regulation of Nkx2.5 levels during development may be required for the progressive emergence of gene expression patterns specific to differentiated Purkinje fiber cells.
Collapse
Affiliation(s)
- Brett S Harris
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Moorman AFM, Christoffels VM. Cardiac chamber formation: development, genes, and evolution. Physiol Rev 2003; 83:1223-67. [PMID: 14506305 DOI: 10.1152/physrev.00006.2003] [Citation(s) in RCA: 472] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Concepts of cardiac development have greatly influenced the description of the formation of the four-chambered vertebrate heart. Traditionally, the embryonic tubular heart is considered to be a composite of serially arranged segments representing adult cardiac compartments. Conversion of such a serial arrangement into the parallel arrangement of the mammalian heart is difficult to understand. Logical integration of the development of the cardiac conduction system into the serial concept has remained puzzling as well. Therefore, the current description needed reconsideration, and we decided to evaluate the essentialities of cardiac design, its evolutionary and embryonic development, and the molecular pathways recruited to make the four-chambered mammalian heart. The three principal notions taken into consideration are as follows. 1) Both the ancestor chordate heart and the embryonic tubular heart of higher vertebrates consist of poorly developed and poorly coupled "pacemaker-like" cardiac muscle cells with the highest pacemaker activity at the venous pole, causing unidirectional peristaltic contraction waves. 2) From this heart tube, ventricular chambers differentiate ventrally and atrial chambers dorsally. The developing chambers display high proliferative activity and consist of structurally well-developed and well-coupled muscle cells with low pacemaker activity, which permits fast conduction of the impulse and efficacious contraction. The forming chambers remain flanked by slowly proliferating pacemaker-like myocardium that is temporally prevented from differentiating into chamber myocardium. 3) The trabecular myocardium proliferates slowly, consists of structurally poorly developed, but well-coupled, cells and contributes to the ventricular conduction system. The atrial and ventricular chambers of the formed heart are activated and interconnected by derivatives of embryonic myocardium. The topographical arrangement of the distinct cardiac muscle cells in the forming heart explains the embryonic electrocardiogram (ECG), does not require the invention of nodes, and allows a logical transition from a peristaltic tubular heart to a synchronously contracting four-chambered heart. This view on the development of cardiac design unfolds fascinating possibilities for future research.
Collapse
Affiliation(s)
- Antoon F M Moorman
- Department of Anatomy & Embryology, Academic Medical Center, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| | | |
Collapse
|
49
|
Sedmera D, Reckova M, DeAlmeida A, Coppen SR, Kubalak SW, Gourdie RG, Thompson RP. Spatiotemporal pattern of commitment to slowed proliferation in the embryonic mouse heart indicates progressive differentiation of the cardiac conduction system. THE ANATOMICAL RECORD. PART A, DISCOVERIES IN MOLECULAR, CELLULAR, AND EVOLUTIONARY BIOLOGY 2003; 274:773-7. [PMID: 12923887 DOI: 10.1002/ar.a.10085] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Patterns of DNA synthesis in the developing mouse heart between ED7.5-18.5 were studied by a combination of thymidine and bromodeoxyuridine labeling techniques. From earliest stages, we found zones of slow myocyte proliferation at both the venous and arterial poles of the heart, as well as in the atrioventricular region. The labeling index was distinctly higher in nonmyocardial populations (endocardium, epicardium, and cardiac cushions). Ventricular trabeculae showed lower proliferative activity than the ventricular compact layer after their appearance at ED9.5. Low labeling was observed in the pectinate muscles of the atria from ED11.5. The His bundle, bundle branches, and Purkinje fiber network likewise were distinguished by their lack of labeling. Thymidine birthdating (label dilution) showed that the cells in these emerging components of the cardiac conduction system terminally differentiated between ED8.5-13.5. These patterns of slowed proliferation correlate well with those in other species, and can serve as a useful marker for the forming conduction system.
Collapse
Affiliation(s)
- David Sedmera
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Bond J, Sedmera D, Jourdan J, Zhang Y, Eisenberg CA, Eisenberg LM, Gourdie RG. Wnt11 and Wnt7a are up-regulated in association with differentiation of cardiac conduction cells in vitro and in vivo. Dev Dyn 2003; 227:536-43. [PMID: 12889062 DOI: 10.1002/dvdy.10333] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The heart beat is coordinated by a precisely timed sequence of action potentials propagated through cells of the conduction system. Previously, we have shown that conduction cells in the chick embryo are derived from multipotent, cardiomyogenic progenitors present in the looped, tubular heart. Moreover, analyses of heterogeneity within myocyte clones and cell birth dating have indicated that elaboration of the conduction system occurs by ongoing, localized recruitment from within this multipotent pool. In this study, we have focused on a potential role for Wnt signaling in development of the cardiac conduction system. Treatment of embryonic myocytes from chick with endothelin-1 (ET-1) has been shown to promote expression of markers of Purkinje fiber cells. By using this in vitro model, we find that Wnt11 are Wnt7a are up-regulated in association with ET-1 treatment. Moreover, in situ hybridization reveals expression, although not temporal coincidence of, Wnt11 and Wnt7a in specialized tissues in the developing heart in vivo. Specifically, whereas Wnt11 shows transient and prominent expression in central elements of the developing conduction system (e.g., the His bundle), relative increases in Wnt7a expression emerge at sites consistent with the location of peripheral conduction cells (e.g., subendocardial Purkinje fibers). The patterns of Wnt11 and Wnt7a expression observed in vitro and in the embryonic chick heart appear to be consistent with roles for these two Wnts in differentiation of cardiac conduction tissues.
Collapse
Affiliation(s)
- Jacqueline Bond
- Department of Cell Biology and Anatomy, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | | | | | |
Collapse
|