1
|
Sukin Z, Moffett E, Wulfeck M, Lindfors D, Szilagyi S. Left Isomerism With Normal Bronchopulmonary Anatomy: Broadening the Heterotaxy Spectrum. Case Rep Radiol 2025; 2025:5512404. [PMID: 40276594 PMCID: PMC12021477 DOI: 10.1155/crra/5512404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
Situs ambiguous is a rare congenital condition characterized by the abnormal arrangement of thoracoabdominal organs along the left-right axis. This condition often presents as either left or right isomerism, leading to complex anatomical variations and associated clinical challenges. We present the case of a 44-year-old female who was incidentally discovered to have situs ambiguous with polysplenia and left atrial appendage isomerism during the evaluation of abdominal pain and urinary symptoms caused by a ureteral calculus. Notably, the patient exhibited normal bronchopulmonary anatomy. The patient underwent a ureteroscopy, laser lithotripsy, stone extraction, and right ureteral stent placement. The patient was discharged shortly thereafter. We believe our case underscores the critical importance of recognizing the potential dissociation between thoracic and abdominal isomerism. It also highlights the need for further investigation into the embryological processes that contribute to these unusual presentations.
Collapse
Affiliation(s)
- Zach Sukin
- Department of Radiology at HCA Healthcare/USF, Morsani College of Medicine, GME/HCA Florida Trinity Hospital, Florida, USA
| | - Erin Moffett
- Department of Radiology at HCA Healthcare/USF, Morsani College of Medicine, GME/HCA Florida Trinity Hospital, Florida, USA
| | - Madison Wulfeck
- Department of Radiology at HCA Healthcare/USF, Morsani College of Medicine, GME/HCA Florida Trinity Hospital, Florida, USA
| | - Dennis Lindfors
- Department of Radiology at HCA Healthcare/USF, Morsani College of Medicine, GME/HCA Florida Trinity Hospital, Florida, USA
| | - Sandor Szilagyi
- Department of Radiology at HCA Healthcare/USF, Morsani College of Medicine, GME/HCA Florida Trinity Hospital, Florida, USA
| |
Collapse
|
2
|
Arthur TD, Joshua IN, Nguyen JP, D'Antonio-Chronowska A, D'Antonio M, Frazer KA. IFNγ activates an immune-like regulatory network in the cardiac vascular endothelium. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100289. [PMID: 40104808 PMCID: PMC11919396 DOI: 10.1016/j.jmccpl.2025.100289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025]
Abstract
The regulatory mechanisms underlying the response to pro-inflammatory cytokines in cardiac diseases are poorly understood. Here, we use iPSC-derived cardiovascular progenitor cells (CVPCs) to model the response to interferon gamma (IFNγ) in human cardiac tissue. We generate RNA-seq and ATAC-seq for four CVPCs that were treated with IFNγ and compare them with paired untreated controls. Transcriptional differences after treatment show that IFNγ initiates an innate immune cell-like response, shifts the CVPC transcriptome toward coronary artery and aorta profiles, and stimulates expression of endothelial cell-specific genes. Analysis of the accessible chromatin shows that IFNγ is a potent chromatin remodeler and establishes an IRF-STAT immune-cell like regulatory network. Finally, we show that 11 GWAS risk variants for 8 common cardiac diseases overlap IFNγ-upregulated ATAC-seq peaks. Our findings reveal insights into IFNγ-induced activation of an immune-like regulatory network in human cardiac tissue and the potential role that regulatory elements in this pathway play in common cardiac diseases.
Collapse
Affiliation(s)
- Timothy D. Arthur
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla 92023, United States of America
- Department of Biomedical Informatics, University of California, San Diego, La Jolla 92023, United States of America
| | - Isaac N. Joshua
- Institute of Genomic Medicine, University of California, San Diego, La Jolla 92023, United States of America
| | - Jennifer P. Nguyen
- Department of Biomedical Informatics, University of California, San Diego, La Jolla 92023, United States of America
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla 92023, United States of America
| | | | - Matteo D'Antonio
- Department of Biomedical Informatics, University of California, San Diego, La Jolla 92023, United States of America
| | - Kelly A. Frazer
- Institute of Genomic Medicine, University of California, San Diego, La Jolla 92023, United States of America
- Department of Pediatrics, University of California, San Diego, La Jolla 92023, United States of America
| |
Collapse
|
3
|
Subati T, Kim K, Yang Z, Murphy MB, Van Amburg JC, Christopher IL, Dougherty OP, Woodall KK, Smart CD, Johnson JE, Fogo AB, Amarnath V, Agrawal V, Barnett JV, Saffitz JE, Murray KT. Oxidative Stress Causes Mitochondrial and Electrophysiologic Dysfunction to Promote Atrial Fibrillation in Pitx2+/- Mice. Circ Arrhythm Electrophysiol 2025; 18:e013199. [PMID: 39989351 PMCID: PMC11919554 DOI: 10.1161/circep.124.013199] [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] [Received: 06/28/2024] [Accepted: 01/30/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND The strongest genetic risk factors for atrial fibrillation (AF) are DNA variants on chromosome 4q25 near the transcription factor gene PITX2 (Pitx2:Paired-like homeodomain transcription factor 2). Mice deficient in Pitx2 (Pitx2+/-) have increased AF susceptibility, although the molecular mechanism(s) remains controversial. Pitx2 encodes a transcription factor that activates an antioxidant response to promote cardiac repair. Increased reactive oxygen species causing oxidation of polyunsaturated fatty acids generates reactive lipid dicarbonyl moieties that adduct to proteins and other macromolecules to promote cellular injury. We tested the hypothesis that oxidative stress, and specifically isolevuglandins, the most reactive lipid dicarbonyls identified, are increased in the setting of Pitx2 deficiency to promote proarrhythmic remodeling and AF. METHODS Pitx2+/- and Pitx2+/+ wild-type littermate control mice were treated orally with vehicle, the lipid dicarbonyl scavenger 2-hydroxybenzylamine, or an inactive control compound at weaning, until study at age 16 to 18 weeks. RESULTS Pitx2+/- mice demonstrated increased P wave duration indicative of slowed atrial conduction, as well as increased inducible AF burden and sustained AF, compared with wild type, and these abnormalities were prevented by 2-hydroxybenzylamine. Both reactive oxygen species and isolevuglandin protein adducts were elevated in Pitx2+/- atria with reduced expression of reactive oxygen species-protective genes. High-resolution respirometry demonstrated impaired mitochondrial function in Pitx2+/- atria, with disruption of mitochondrial integrity and cell-cell junctions with connexin lateralization, as well as decreased mitochondrial biogenesis gene expression. Proarrhythmic ionic current remodeling in Pitx2+/- atrial myocytes included elevated resting membrane potential, abbreviated action potential duration, and reduced maximum phase 0 upstroke velocity compared with wild type. Most of these abnormalities were ameliorated or prevented by 2-hydroxybenzylamine. CONCLUSIONS These results demonstrate a critical role for lipid dicarbonyl mediators of oxidative stress in the proarrhythmic remodeling and AF susceptibility that occurs with Pitx2 deficiency, implying the possibility of genotype-specific therapy to prevent AF.
Collapse
Affiliation(s)
- Tuerdi Subati
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Kyungsoo Kim
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Zhenjiang Yang
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Matthew B. Murphy
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Joseph C. Van Amburg
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Isis L. Christopher
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Owen P. Dougherty
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Kaylen K. Woodall
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Charles D. Smart
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Joyce E. Johnson
- Dept of Pathology, Microbiology, and Immunology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Agnes B. Fogo
- Dept of Pathology, Microbiology, and Immunology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Venkataraman Amarnath
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Vineet Agrawal
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
| | - Joey V. Barnett
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| | | | - Katherine T. Murray
- Dept of Medicine, Vanderbilt Univ School of Medicine, Nashville, TN
- Dept of Pharmacology, Vanderbilt Univ School of Medicine, Nashville, TN
| |
Collapse
|
4
|
Qiu X, Zhu DY, Lu Y, Yao J, Jing Z, Min KH, Cheng M, Pan H, Zuo L, King S, Fang Q, Zheng H, Wang M, Wang S, Zhang Q, Yu S, Liao S, Liu C, Wu X, Lai Y, Hao S, Zhang Z, Wu L, Zhang Y, Li M, Tu Z, Lin J, Yang Z, Li Y, Gu Y, Ellison D, Chen A, Liu L, Weissman JS, Ma J, Xu X, Liu S, Bai Y. Spatiotemporal modeling of molecular holograms. Cell 2024; 187:7351-7373.e61. [PMID: 39532097 DOI: 10.1016/j.cell.2024.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Quantifying spatiotemporal dynamics during embryogenesis is crucial for understanding congenital diseases. We developed Spateo (https://github.com/aristoteleo/spateo-release), a 3D spatiotemporal modeling framework, and applied it to a 3D mouse embryogenesis atlas at E9.5 and E11.5, capturing eight million cells. Spateo enables scalable, partial, non-rigid alignment, multi-slice refinement, and mesh correction to create molecular holograms of whole embryos. It introduces digitization methods to uncover multi-level biology from subcellular to whole organ, identifying expression gradients along orthogonal axes of emergent 3D structures, e.g., secondary organizers such as midbrain-hindbrain boundary (MHB). Spateo further jointly models intercellular and intracellular interaction to dissect signaling landscapes in 3D structures, including the zona limitans intrathalamica (ZLI). Lastly, Spateo introduces "morphometric vector fields" of cell migration and integrates spatial differential geometry to unveil molecular programs underlying asymmetrical murine heart organogenesis and others, bridging macroscopic changes with molecular dynamics. Thus, Spateo enables the study of organ ecology at a molecular level in 3D space over time.
Collapse
Affiliation(s)
- Xiaojie Qiu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| | - Daniel Y Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifan Lu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Electronic Information School, Wuhan University, Wuhan 430072, China
| | - Jiajun Yao
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kyung Hoi Min
- Ginkgo Bioworks, The Innovation and Design Building, Boston, MA 02210, USA
| | - Mengnan Cheng
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | | | - Lulu Zuo
- BGI Research, Shenzhen 518083, China
| | - Samuel King
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Fang
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuai Wang
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingquan Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Sichao Yu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Chao Liu
- BGI Research, Wuhan 430074, China
| | - Xinchao Wu
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiwei Lai
- BGI Research, Shenzhen 518083, China
| | | | - Zhewei Zhang
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liang Wu
- BGI Research, Chongqing 401329, China
| | | | - Mei Li
- STOmics Tech Co., Ltd, Shenzhen 518083, China
| | - Zhencheng Tu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinpei Lin
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China
| | - Zhuoxuan Yang
- BGI Research, Hangzhou 310030, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Ying Gu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ao Chen
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Jiayi Ma
- Electronic Information School, Wuhan University, Wuhan 430072, China.
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China.
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China; The Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, Guangdong, China.
| | - Yinqi Bai
- BGI Research, Sanya 572025, China; Hainan Technology Innovation Center for Marine Biological Resources Utilization (Preparatory Period), BGI Research, Sanya 572025, China.
| |
Collapse
|
5
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
6
|
Gabriel GC, Lo CW. Molecular Pathways and Animal Models of Defects in Situs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:719-738. [PMID: 38884745 DOI: 10.1007/978-3-031-44087-8_43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Left-right patterning is among the least well understood of the three axes defining the body plan, and yet it is no less important, with left-right patterning defects causing structural birth defects with high morbidity and mortality, such as complex congenital heart disease, biliary atresia, or intestinal malrotation. The cell signaling pathways governing left-right asymmetry are highly conserved and involve multiple components of the TGF-β superfamily of cell signaling molecules. Central to left-right patterning is the differential activation of Nodal on the left, and BMP signaling on the right. In addition, a plethora of other cell signaling pathways including Shh, FGF, and Notch also contribute to the regulation of left-right patterning. In vertebrate embryos such as the mouse, frog, or zebrafish, the specification of left-right identity requires the left-right organizer (LRO) containing cells with motile and primary cilia that mediate the left-sided propagation of Nodal signaling, followed by left-sided activation of Lefty and then Pitx2, a transcription factor that specifies visceral organ asymmetry. While this overall scheme is well conserved, there are striking species differences, including the finding that motile cilia do not play a role in left-right patterning in some vertebrates. Surprisingly, the direction of heart looping, one of the first signs of organ left-right asymmetry, was recently shown to be specified by intrinsic cell chirality, not Nodal signaling, possibly a reflection of the early origin of Nodal signaling in radially symmetric organisms. How this intrinsic chirality interacts with downstream molecular pathways regulating visceral organ asymmetry will need to be further investigated to elucidate how disturbance in left-right patterning may contribute to complex CHD.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Hikspoors JPJM, Kruepunga N, Mommen GMC, Köhler SE, Anderson RH, Lamers WH. Human Cardiac Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:3-55. [PMID: 38884703 DOI: 10.1007/978-3-031-44087-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Many aspects of heart development are topographically complex and require three-dimensional (3D) reconstruction to understand the pertinent morphology. We have recently completed a comprehensive primer of human cardiac development that is based on firsthand segmentation of structures of interest in histological sections. We visualized the hearts of 12 human embryos between their first appearance at 3.5 weeks and the end of the embryonic period at 8 weeks. The models were presented as calibrated, interactive, 3D portable document format (PDF) files. We used them to describe the appearance and the subsequent remodeling of around 70 different structures incrementally for each of the reconstructed stages. In this chapter, we begin our account by describing the formation of the single heart tube, which occurs at the end of the fourth week subsequent to conception. We describe its looping in the fifth week, the formation of the cardiac compartments in the sixth week, and, finally, the septation of these compartments into the physically separated left- and right-sided circulations in the seventh and eighth weeks. The phases are successive, albeit partially overlapping. Thus, the basic cardiac layout is established between 26 and 32 days after fertilization and is described as Carnegie stages (CSs) 9 through 14, with development in the outlet component trailing that in the inlet parts. Septation at the venous pole is completed at CS17, equivalent to almost 6 weeks of development. During Carnegie stages 17 and 18, in the seventh week, the outflow tract and arterial pole undergo major remodeling, including incorporation of the proximal portion of the outflow tract into the ventricles and transfer of the spiraling course of the subaortic and subpulmonary channels to the intrapericardial arterial trunks. Remodeling of the interventricular foramen, with its eventual closure, is complete at CS20, which occurs at the end of the seventh week. We provide quantitative correlations between the age of human and mouse embryos as well as the Carnegie stages of development. We have also set our descriptions in the context of variations in the timing of developmental features.
Collapse
Affiliation(s)
- Jill P J M Hikspoors
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands.
| | - Nutmethee Kruepunga
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
- Present address: Department of Anatomy, Mahidol University, Bangkok, Thailand
| | - Greet M C Mommen
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - S Eleonore Köhler
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
| | - Robert H Anderson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Wouter H Lamers
- Department of Anatomy & Embryology, Maastricht University, Maastricht, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Bileckyj C, Blotz B, Cripps RM. Drosophila as a Model to Understand Second Heart Field Development. J Cardiovasc Dev Dis 2023; 10:494. [PMID: 38132661 PMCID: PMC10744189 DOI: 10.3390/jcdd10120494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The genetic model system Drosophila has contributed fundamentally to our understanding of mammalian heart specification, development, and congenital heart disease. The relatively simple Drosophila heart is a linear muscular tube that is specified and develops in the embryo and persists throughout the life of the animal. It functions at all stages to circulate hemolymph within the open circulatory system of the body. During Drosophila metamorphosis, the cardiac tube is remodeled, and a new layer of muscle fibers spreads over the ventral surface of the heart to form the ventral longitudinal muscles. The formation of these fibers depends critically upon genes known to be necessary for mammalian second heart field (SHF) formation. Here, we review the prior contributions of the Drosophila system to the understanding of heart development and disease, discuss the importance of the SHF to mammalian heart development and disease, and then discuss how the ventral longitudinal adult cardiac muscles can serve as a novel model for understanding SHF development and disease.
Collapse
Affiliation(s)
| | | | - Richard M. Cripps
- Department of Biology, San Diego State University, San Diego, CA 92182, USA
| |
Collapse
|
9
|
Abstract
Axenfeld-Rieger syndrome is a rare multi-system disorder associated with cardiac anomalies. All patients with a diagnosis of Axenfeld-Rieger syndrome were identified from our electronic medical record. Chart review was performed to document the presence and types of CHD. Out of 58 patients, 14 (24.1%) had CHD and a wide variety of cardiac lesions were identified.
Collapse
Affiliation(s)
- Nishma Valikodath
- Department of Pediatrics, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, USA
| | - James A Johns
- Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, USA
| | - Justin Godown
- Division of Pediatric Cardiology, Monroe Carell Jr. Children's Hospital at Vanderbilt, Nashville, USA
| |
Collapse
|
10
|
Steimle JD, Grisanti Canozo FJ, Park M, Kadow ZA, Samee MAH, Martin JF. Decoding the PITX2-controlled genetic network in atrial fibrillation. JCI Insight 2022; 7:e158895. [PMID: 35471998 PMCID: PMC9221021 DOI: 10.1172/jci.insight.158895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Atrial fibrillation (AF), the most common sustained cardiac arrhythmia and a major risk factor for stroke, often arises through ectopic electrical impulses derived from the pulmonary veins (PVs). Sequence variants in enhancers controlling expression of the transcription factor PITX2, which is expressed in the cardiomyocytes (CMs) of the PV and left atrium (LA), have been implicated in AF predisposition. Single nuclei multiomic profiling of RNA and analysis of chromatin accessibility combined with spectral clustering uncovered distinct PV- and LA-enriched CM cell states. Pitx2-mutant PV and LA CMs exhibited gene expression changes consistent with cardiac dysfunction through cell type-distinct, PITX2-directed, cis-regulatory grammars controlling target gene expression. The perturbed network targets in each CM were enriched in distinct human AF predisposition genes, suggesting combinatorial risk for AF genesis. Our data further reveal that PV and LA Pitx2-mutant CMs signal to endothelial and endocardial cells through BMP10 signaling with pathogenic potential. This work provides a multiomic framework for interrogating the basis of AF predisposition in the PVs of humans.
Collapse
Affiliation(s)
| | | | | | - Zachary A. Kadow
- Program in Developmental Biology, and
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, USA
| | | | - James F. Martin
- Department of Integrative Physiology
- Texas Heart Institute, Houston, Texas, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
11
|
Derrick CJ, Santos-Ledo A, Eley L, Paramita IA, Henderson DJ, Chaudhry B. Sequential action of JNK genes establishes the embryonic left-right axis. Development 2022; 149:274898. [PMID: 35352808 PMCID: PMC9148569 DOI: 10.1242/dev.200136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 03/09/2022] [Indexed: 12/22/2022]
Abstract
The establishment of the left-right axis is crucial for the placement, morphogenesis and function of internal organs. Left-right specification is proposed to be dependent on cilia-driven fluid flow in the embryonic node. Planar cell polarity (PCP) signalling is crucial for patterning of nodal cilia, yet downstream effectors driving this process remain elusive. We have examined the role of the JNK gene family, a proposed downstream component of PCP signalling, in the development and function of the zebrafish node. We show jnk1 and jnk2 specify length of nodal cilia, generate flow in the node and restrict southpaw to the left lateral plate mesoderm. Moreover, loss of asymmetric southpaw expression does not result in disturbances to asymmetric organ placement, supporting a model in which nodal flow may be dispensable for organ laterality. Later, jnk3 is required to restrict pitx2c expression to the left side and permit correct endodermal organ placement. This work uncovers multiple roles for the JNK gene family acting at different points during left-right axis establishment. It highlights extensive redundancy and indicates JNK activity is distinct from the PCP signalling pathway.
Collapse
Affiliation(s)
- Christopher J Derrick
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Adrian Santos-Ledo
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Lorraine Eley
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Isabela Andhika Paramita
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Deborah J Henderson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
12
|
Shewale B, Dubois N. Of form and function: Early cardiac morphogenesis across classical and emerging model systems. Semin Cell Dev Biol 2021; 118:107-118. [PMID: 33994301 PMCID: PMC8434962 DOI: 10.1016/j.semcdb.2021.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022]
Abstract
The heart is the earliest organ to develop during embryogenesis and is remarkable in its ability to function efficiently as it is being sculpted. Cardiac heart defects account for a high burden of childhood developmental disorders with many remaining poorly understood mechanistically. Decades of work across a multitude of model organisms has informed our understanding of early cardiac differentiation and morphogenesis and has simultaneously opened new and unanswered questions. Here we have synthesized current knowledge in the field and reviewed recent developments in the realm of imaging, bioengineering and genetic technology and ex vivo cardiac modeling that may be deployed to generate more holistic models of early cardiac morphogenesis, and by extension, new platforms to study congenital heart defects.
Collapse
Affiliation(s)
- Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
13
|
French CR. Mechanistic Insights into Axenfeld-Rieger Syndrome from Zebrafish foxc1 and pitx2 Mutants. Int J Mol Sci 2021; 22:ijms221810001. [PMID: 34576164 PMCID: PMC8472202 DOI: 10.3390/ijms221810001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/03/2021] [Accepted: 09/05/2021] [Indexed: 12/11/2022] Open
Abstract
Axenfeld-Rieger syndrome (ARS) encompasses a group of developmental disorders that affect the anterior segment of the eye, as well as systemic developmental defects in some patients. Malformation of the ocular anterior segment often leads to secondary glaucoma, while some patients also present with cardiovascular malformations, craniofacial and dental abnormalities and additional periumbilical skin. Genes that encode two transcription factors, FOXC1 and PITX2, account for almost half of known cases, while the genetic lesions in the remaining cases remain unresolved. Given the genetic similarity between zebrafish and humans, as well as robust antisense inhibition and gene editing technologies available for use in these animals, loss of function zebrafish models for ARS have been created and shed light on the mechanism(s) whereby mutations in these two transcription factors cause such a wide array of developmental phenotypes. This review summarizes the published phenotypes in zebrafish foxc1 and pitx2 loss of function models and discusses possible mechanisms that may be used to target pharmaceutical development and therapeutic interventions.
Collapse
Affiliation(s)
- Curtis R French
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
14
|
Amoushahi M, Lykke-Hartmann K. Distinct Signaling Pathways Distinguish in vivo From in vitro Growth in Murine Ovarian Follicle Activation and Maturation. Front Cell Dev Biol 2021; 9:708076. [PMID: 34368158 PMCID: PMC8346253 DOI: 10.3389/fcell.2021.708076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022] Open
Abstract
Women with cancer and low ovarian reserves face serious challenges in infertility treatment. Ovarian tissue cryopreservation is currently used for such patients to preserve fertility. One major challenge is the activation of dormant ovarian follicles, which is hampered by our limited biological understanding of molecular determinants that activate dormant follicles and help maintain healthy follicles during growth. Here, we investigated the transcriptomes of oocytes isolated from dormant (primordial) and activated (primary) follicles under in vivo and in vitro conditions. We compared the biological relevance of the initial molecular markers of mature metaphase II (MII) oocytes developed in vivo or in vitro. The expression levels of genes involved in the cell cycle, signal transduction, and Wnt signaling were highly enriched in oocytes from primary follicles and MII oocytes. Interestingly, we detected strong downregulation of the expression of genes involved in mitochondrial and reactive oxygen species (ROS) production in oocytes from primordial follicles, in contrast to oocytes from primary follicles and MII oocytes. Our results showed a dynamic pattern in mitochondrial and ROS production-related genes, emphasizing their important role(s) in primordial follicle activation and oocyte maturation. The transcriptome of MII oocytes showed a major divergence from that of oocytes of primordial and primary follicles.
Collapse
Affiliation(s)
| | - Karin Lykke-Hartmann
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Clinical Genetics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Tran TQ, Kioussi C. Pitx genes in development and disease. Cell Mol Life Sci 2021; 78:4921-4938. [PMID: 33844046 PMCID: PMC11073205 DOI: 10.1007/s00018-021-03833-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/05/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Homeobox genes encode sequence-specific transcription factors (SSTFs) that recognize specific DNA sequences and regulate organogenesis in all eukaryotes. They are essential in specifying spatial and temporal cell identity and as a result, their mutations often cause severe developmental defects. Pitx genes belong to the PRD class of the highly evolutionary conserved homeobox genes in all animals. Vertebrates possess three Pitx paralogs, Pitx1, Pitx2, and Pitx3 while non-vertebrates have only one Pitx gene. The ancient role of regulating left-right (LR) asymmetry is conserved while new functions emerge to afford more complex body plan and functionalities. In mouse, Pitx1 regulates hindlimb tissue patterning and pituitary development. Pitx2 is essential for the development of the oral cavity and abdominal wall while regulates the formation and symmetry of other organs including pituitary, heart, gut, lung among others by controlling growth control genes upon activation of the Wnt/ß-catenin signaling pathway. Pitx3 is essential for lens development and migration and survival of the dopaminergic neurons of the substantia nigra. Pitx gene mutations are linked to various congenital defects and cancers in humans. Pitx gene family has the potential to offer a new approach in regenerative medicine and aid in identifying new drug targets.
Collapse
Affiliation(s)
- Thai Q Tran
- Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Chrissa Kioussi
- Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA.
| |
Collapse
|
16
|
Abstract
Cardiac development is a complex developmental process that is initiated soon after gastrulation, as two sets of precardiac mesodermal precursors are symmetrically located and subsequently fused at the embryonic midline forming the cardiac straight tube. Thereafter, the cardiac straight tube invariably bends to the right, configuring the first sign of morphological left–right asymmetry and soon thereafter the atrial and ventricular chambers are formed, expanded and progressively septated. As a consequence of all these morphogenetic processes, the fetal heart acquired a four-chambered structure having distinct inlet and outlet connections and a specialized conduction system capable of directing the electrical impulse within the fully formed heart. Over the last decades, our understanding of the morphogenetic, cellular, and molecular pathways involved in cardiac development has exponentially grown. Multiples aspects of the initial discoveries during heart formation has served as guiding tools to understand the etiology of cardiac congenital anomalies and adult cardiac pathology, as well as to enlighten novels approaches to heal the damaged heart. In this review we provide an overview of the complex cellular and molecular pathways driving heart morphogenesis and how those discoveries have provided new roads into the genetic, clinical and therapeutic management of the diseased hearts.
Collapse
|
17
|
The Axenfeld-Rieger Syndrome Gene FOXC1 Contributes to Left-Right Patterning. Genes (Basel) 2021; 12:genes12020170. [PMID: 33530637 PMCID: PMC7912076 DOI: 10.3390/genes12020170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Precise spatiotemporal expression of the Nodal-Lefty-Pitx2 cascade in the lateral plate mesoderm establishes the left–right axis, which provides vital cues for correct organ formation and function. Mutations of one cascade constituent PITX2 and, separately, the Forkhead transcription factor FOXC1 independently cause a multi-system disorder known as Axenfeld–Rieger syndrome (ARS). Since cardiac involvement is an established ARS phenotype and because disrupted left–right patterning can cause congenital heart defects, we investigated in zebrafish whether foxc1 contributes to organ laterality or situs. We demonstrate that CRISPR/Cas9-generated foxc1a and foxc1b mutants exhibit abnormal cardiac looping and that the prevalence of cardiac situs defects is increased in foxc1a−/−; foxc1b−/− homozygotes. Similarly, double homozygotes exhibit isomerism of the liver and pancreas, which are key features of abnormal gut situs. Placement of the asymmetric visceral organs relative to the midline was also perturbed by mRNA overexpression of foxc1a and foxc1b. In addition, an analysis of the left–right patterning components, identified in the lateral plate mesoderm of foxc1 mutants, reduced or abolished the expression of the NODAL antagonist lefty2. Together, these data reveal a novel contribution from foxc1 to left–right patterning, demonstrating that this role is sensitive to foxc1 gene dosage, and provide a plausible mechanism for the incidence of congenital heart defects in Axenfeld–Rieger syndrome patients.
Collapse
|
18
|
Reyat JS, Chua W, Cardoso VR, Witten A, Kastner PM, Kabir SN, Sinner MF, Wesselink R, Holmes AP, Pavlovic D, Stoll M, Kääb S, Gkoutos GV, de Groot JR, Kirchhof P, Fabritz L. Reduced left atrial cardiomyocyte PITX2 and elevated circulating BMP10 predict atrial fibrillation after ablation. JCI Insight 2020; 5:139179. [PMID: 32814717 PMCID: PMC7455124 DOI: 10.1172/jci.insight.139179] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/08/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUNDGenomic and experimental studies suggest a role for PITX2 in atrial fibrillation (AF). To assess if this association is relevant for recurrent AF in patients, we tested whether left atrial PITX2 affects recurrent AF after AF ablation.METHODSmRNA concentrations of PITX2 and its cardiac isoform, PITX2c, were quantified in left atrial appendages (LAAs) from patients undergoing thoracoscopic AF ablation, either in whole LAA tissue (n = 83) or in LAA cardiomyocytes (n = 52), and combined with clinical parameters to predict AF recurrence. Literature suggests that BMP10 is a PITX2-repressed, atrial-specific, secreted protein. BMP10 plasma concentrations were combined with 11 cardiovascular biomarkers and clinical parameters to predict recurrent AF after catheter ablation in 359 patients.RESULTSReduced concentrations of cardiomyocyte PITX2, but not whole LAA tissue PITX2, were associated with AF recurrence after thoracoscopic AF ablation (16% decreased recurrence per 2-(ΔΔCt) increase in PITX2). RNA sequencing, quantitative PCR, and Western blotting confirmed that BMP10 is one of the most PITX2-repressed atrial genes. Left atrial size (HR per mm increase [95% CI], 1.055 [1.028, 1.082]); nonparoxysmal AF (HR 1.672 [1.206, 2.318]), and elevated BMP10 (HR 1.339 [CI 1.159, 1.546] per quartile increase) were predictive of recurrent AF. BMP10 outperformed 11 other cardiovascular biomarkers in predicting recurrent AF.CONCLUSIONSReduced left atrial cardiomyocyte PITX2 and elevated plasma concentrations of the PITX2-repressed, secreted atrial protein BMP10 identify patients at risk of recurrent AF after ablation.TRIAL REGISTRATIONClinicalTrials.gov NCT01091389, NL50069.018.14, Dutch National Registry of Clinical Research Projects EK494-16.FUNDINGBritish Heart Foundation, European Union (H2020), Leducq Foundation.
Collapse
Affiliation(s)
| | | | - Victor R. Cardoso
- Institute of Cardiovascular Sciences and
- Institute of Cancer and Genomics Sciences, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Anika Witten
- Institute of Human Genetics, Genetic Epidemiology, WWU Münster, Münster, Germany
| | | | | | - Moritz F. Sinner
- Department of Medicine I, University Hospital Munich, Ludwig Maximilian University of Munich (LMU), Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Robin Wesselink
- Department of Cardiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Heart Center, Amsterdam, Netherlands
| | | | | | - Monika Stoll
- Institute of Human Genetics, Genetic Epidemiology, WWU Münster, Münster, Germany
- Cardiovascular Research Institute Maastricht, Genetic Epidemiology and Statistical Genetics, Maastricht University, Maastricht, Netherlands
| | - Stefan Kääb
- Department of Medicine I, University Hospital Munich, Ludwig Maximilian University of Munich (LMU), Munich, Germany
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
| | - Georgios V. Gkoutos
- Institute of Cardiovascular Sciences and
- Institute of Cancer and Genomics Sciences, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, United Kingdom
- Health Data Research Midlands, Birmingham, United Kingdom
| | - Joris R. de Groot
- Department of Cardiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Heart Center, Amsterdam, Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences and
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
- Department of Cardiology, University Hospitals Birmingham (UHB) and Sandwell and West Birmingham (SWBH) NHS Trusts, Birmingham, United Kingdom
- University Heart and Vascular Center, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg, Germany
- German Center for Cardiovascular Research, partner site Hamburg/Kiel/Lübeck, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences and
- Atrial Fibrillation NETwork (AFNET), Münster, Germany
- Department of Cardiology, University Hospitals Birmingham (UHB) and Sandwell and West Birmingham (SWBH) NHS Trusts, Birmingham, United Kingdom
| |
Collapse
|
19
|
Stothard CA, Mazzotta S, Vyas A, Schneider JE, Mohun TJ, Henderson DJ, Phillips HM, Bamforth SD. Pax9 and Gbx2 Interact in the Pharyngeal Endoderm to Control Cardiovascular Development. J Cardiovasc Dev Dis 2020; 7:jcdd7020020. [PMID: 32466118 PMCID: PMC7344924 DOI: 10.3390/jcdd7020020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
The correct formation of the aortic arch arteries depends on a coordinated and regulated gene expression profile within the tissues of the pharyngeal arches. Perturbation of the gene regulatory networks in these tissues results in congenital heart defects affecting the arch arteries and the outflow tract of the heart. Aberrant development of these structures leads to interruption of the aortic arch and double outlet right ventricle, abnormalities that are a leading cause of morbidity in 22q11 Deletion Syndrome (DS) patients. We have recently shown that Pax9 functionally interacts with the 22q11DS gene Tbx1 in the pharyngeal endoderm for 4th pharyngeal arch artery morphogenesis, with double heterozygous mice dying at birth with interrupted aortic arch. Mice lacking Pax9 die perinatally with complex cardiovascular defects and in this study we sought to validate further potential genetic interacting partners of Pax9, focussing on Gbx2 which is down-regulated in the pharyngeal endoderm of Pax9-null embryos. Here, we describe the Gbx2-null cardiovascular phenotype and demonstrate a genetic interaction between Gbx2 and Pax9 in the pharyngeal endoderm during cardiovascular development.
Collapse
Affiliation(s)
- Catherine A. Stothard
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
| | - Silvia Mazzotta
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
| | - Arjun Vyas
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
| | | | | | - Deborah J. Henderson
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
| | - Helen M. Phillips
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
| | - Simon D. Bamforth
- Newcastle University Biosciences Institute, Centre for Life, Newcastle-upon-Tyne NE1 3BZ, UK; (C.A.S.); (S.M.); (A.V.); (D.J.H.); (H.M.P.)
- Correspondence: ; Tel.: +44-191-241-8764
| |
Collapse
|
20
|
Yang F, Qi J. miR-430a regulates the development of left-right asymmetry by targeting sqt in the teleost. Gene 2020; 745:144628. [PMID: 32224271 DOI: 10.1016/j.gene.2020.144628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 10/24/2022]
Abstract
microRNAs (miRNAs) are short, endogenous non-coding RNAs that contain approximately 18-22 nucleotides. miRNAs are involved in gene regulation by recognizing and binding the 3'UTR of target gene. In our previous data, miR-430 family showed significant differential expression modes through metamorphosis in Japanese flounder. It was speculated that miR-430a plays a key role in left-right patterning. We predicted the targets of miR-430a and gene ontology (GO) was performed. We speculated miR-430a is involved in the basal molecular function and organ development. In Japanese flounder, sqt as a target of miR-430a was enriched into heart development term. Sqt has been reported to participate in mesendoderm formation and organ development. Cardiac morphogenesis is the first asymmetric development process, which breaks left-right symmetry in bilateria. It was used as a marker to detect L-R asymmetric effects of miR-430a. Overexpression and suppression of miR-430a resulted in abnormal KV (Kupffer's vesicles) development and disordered in nodal-related expression with consequent cardiac laterality. Squint mRNA of Japanese flounder (Posqt) as a target of miR-430a was overexpressed and caused similar phenotype with miR-430a suppression group, such as longer cilia in KV and high range of clmc2 and spaw ectopic expression. Moreover, rescue experiments were performed and suggested that cardiac and KV defections, induced by overexpressing miR-430a, could be rescued by injecting Posqt mRNA. These results suggested that miR-430a regulates the development of left-right asymmetry by targeting sqt in the teleost.
Collapse
Affiliation(s)
- Fan Yang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Qi
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| |
Collapse
|
21
|
Desgrange A, Lokmer J, Marchiol C, Houyel L, Meilhac SM. Standardised imaging pipeline for phenotyping mouse laterality defects and associated heart malformations, at multiple scales and multiple stages. Dis Model Mech 2019; 12:dmm.038356. [PMID: 31208960 PMCID: PMC6679386 DOI: 10.1242/dmm.038356] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
Laterality defects are developmental disorders resulting from aberrant left/right patterning. In the most severe cases, such as in heterotaxy, they are associated with complex malformations of the heart. Advances in understanding the underlying physiopathological mechanisms have been hindered by the lack of a standardised and exhaustive procedure in mouse models for phenotyping left/right asymmetries of all visceral organs. Here, we have developed a multimodality imaging pipeline, which combines non-invasive micro-ultrasound imaging, micro-computed tomography (micro-CT) and high-resolution episcopic microscopy (HREM) to acquire 3D images at multiple stages of development and at multiple scales. On the basis of the position in the uterine horns, we track in a single individual, the progression of organ asymmetry, the situs of all visceral organs in the thoracic or abdominal environment, and the fine anatomical left/right asymmetries of cardiac segments. We provide reference anatomical images and organ reconstructions in the mouse, and discuss differences with humans. This standardised pipeline, which we validated in a mouse model of heterotaxy, offers a fast and easy-to-implement framework. The extensive 3D phenotyping of organ asymmetry in the mouse uses the clinical nomenclature for direct comparison with patient phenotypes. It is compatible with automated and quantitative image analyses, which is essential to compare mutant phenotypes with incomplete penetrance and to gain mechanistic insight into laterality defects. Summary: Laterality defects, which combine anomalies in several visceral organs, are challenging to phenotype. We have developed here a standardised approach for multimodality 3D imaging in mice, generating quantifiable phenotypes.
Collapse
Affiliation(s)
- Audrey Desgrange
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France
| | - Johanna Lokmer
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France
| | - Carmen Marchiol
- Université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France.,INSERM U1016, Institut Cochin, 75014 Paris, France.,CNRS UMR8104, 75014 Paris, France
| | - Lucile Houyel
- Université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France.,Unité de Cardiologie Pédiatrique et Congénitale, Hôpital Necker Enfants Malades, Centre de référence des Malformations Cardiaques Congénitales Complexes-M3C, APHP, 75015 Paris, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France .,INSERM UMR1163, 75015 Paris, France.,Université Paris Descartes, Sorbonne Paris-Cité, 75006 Paris, France
| |
Collapse
|
22
|
Hill MC, Kadow ZA, Li L, Tran TT, Wythe JD, Martin JF. A cellular atlas of Pitx2-dependent cardiac development. Development 2019; 146:dev180398. [PMID: 31201182 PMCID: PMC6602352 DOI: 10.1242/dev.180398] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 05/13/2019] [Indexed: 12/12/2022]
Abstract
The Pitx2 gene encodes a homeobox transcription factor that is required for mammalian development. Disruption of PITX2 expression in humans causes congenital heart diseases and is associated with atrial fibrillation; however, the cellular and molecular processes dictated by Pitx2 during cardiac ontogeny remain unclear. To characterize the role of Pitx2 during murine heart development we sequenced over 75,000 single cardiac cell transcriptomes between two key developmental timepoints in control and Pitx2 null embryos. We found that cardiac cell composition was dramatically altered in mutants at both E10.5 and E13.5. Interestingly, the differentiation dynamics of both anterior and posterior second heart field-derived progenitor cells were disrupted in Pitx2 mutants. We also uncovered evidence for defects in left-right asymmetry within atrial cardiomyocyte populations. Furthermore, we were able to detail defects in cardiac outflow tract and valve development associated with Pitx2 Our findings offer insight into Pitx2 function and provide a compilation of gene expression signatures for further detailing the complexities of heart development that will serve as the foundation for future studies of cardiac morphogenesis, congenital heart disease and arrhythmogenesis.
Collapse
Affiliation(s)
- Matthew C Hill
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zachary A Kadow
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lele Li
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tien T Tran
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joshua D Wythe
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - James F Martin
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Texas Heart Institute, Houston, TX 77030, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
23
|
Mechakra A, Footz T, Walter M, Aránega A, Hernández-Torres F, Morel E, Millat G, Yang YQ, Chahine M, Chevalier P, Christé G. A Novel PITX2c Gain-of-Function Mutation, p.Met207Val, in Patients With Familial Atrial Fibrillation. Am J Cardiol 2019; 123:787-793. [PMID: 30558760 DOI: 10.1016/j.amjcard.2018.11.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 11/16/2018] [Accepted: 11/20/2018] [Indexed: 01/08/2023]
Abstract
Genome-wide studies have associated several genetic variants upstream of PITX2 on chromosome 4q25 with atrial fibrillation (AF), suggesting a potential role of PITX2 in AF. Our study aimed at identifying rare coding variants in PITX2 predisposing to AF. The Polymerase chain reaction sequencing of PITX2c was performed in 60 unrelated patients with idiopathic AF. The p.Met207Val variant was identified in 1 of 60 French patients with early onset AF and in none of 389 French referents. This variant, located in the inhibitory domain 1 distal to the homeodomain, was evaluated by the software MutationTaster as a disease-causing mutation with a probability of 0.999. Reporter gene assays demonstrated that p.Met207Val caused a 3.1-fold increase in transactivation activity of PITX2c in HeLa cells in comparison with its wild-type counterpart. When the variant was coexpressed with wild-type PITX2c in the HL-1 immortalized mouse atrial cell line, this gain-of-function caused an increase in the mRNA level of KCNH2 (2.6-fold), SCN1B (1.9-fold), GJA5 (3.1-fold), GJA1 (2.1-fold), and KCNQ1 in the homozygous form (1.8-fold). These genes encode for the IKr channel α subunit, the β-1 Na+ channel subunit, connexin 40, connexin 43 and the IKs channel α subunit, respectively. These conditions may contribute to the propensity to AF found in patients carrying the p.Met207Val variant. In conclusion, the present report is the first to associate a gain-of-function mutation of PITX2c with increased vulnerability to AF, therefore, restoration of normal PITX2c function may be a potential therapeutic target in AF patients.
Collapse
Affiliation(s)
- Asma Mechakra
- EA4612 Neurocardiologie, Université Lyon 1, Lyon, France
| | - Tim Footz
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Walter
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Amelia Aránega
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | | | - Elodie Morel
- EA4612 Neurocardiologie, Université Lyon 1, Lyon, France
| | - Gilles Millat
- EA4612 Neurocardiologie, Université Lyon 1, Lyon, France
| | - Yi-Qing Yang
- Department of Cardiology, La-boratory of Cardiovascular Research and Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Mohamed Chahine
- Institut Universitaire en Santé Mentale, Québec City, Québec, Canada
| | | | | |
Collapse
|
24
|
Chang CN, Singh AJ, Gross MK, Kioussi C. Requirement of Pitx2 for skeletal muscle homeostasis. Dev Biol 2019; 445:90-102. [PMID: 30414844 PMCID: PMC6289786 DOI: 10.1016/j.ydbio.2018.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/05/2018] [Accepted: 11/05/2018] [Indexed: 12/24/2022]
Abstract
Skeletal muscle is generated by the successive incorporation of primary (embryonic), secondary (fetal), and tertiary (adult) fibers into muscle. Conditional excision of Pitx2 function by an MCKCre driver resulted in animals with histological and ultrastructural defects in P30 muscles and fibers, respectively. Mutant muscle showed severe reduction in mitochondria and FoxO3-mediated mitophagy. Both oxidative and glycolytic energy metabolism were reduced. Conditional excision was limited to fetal muscle fibers after the G1-G0 transition and resulted in altered MHC, Rac1, MEF2a, and alpha-tubulin expression within these fibers. The onset of excision, monitored by a nuclear reporter gene, was observed as early as E16. Muscle at this stage was already severely malformed, but appeared to recover by P30 by the expansion of adjoining larger fibers. Our studies demonstrate that the homeodomain transcription factor Pitx2 has a postmitotic role in maintaining skeletal muscle integrity and energy homeostasis in fetal muscle fibers.
Collapse
Affiliation(s)
- Chih-Ning Chang
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA; Molecular Cell Biology Graduate Program, Oregon State University, Corvallis, OR 97331, USA
| | - Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Michael K Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
25
|
Desgrange A, Le Garrec JF, Meilhac SM. Left-right asymmetry in heart development and disease: forming the right loop. Development 2018; 145:145/22/dev162776. [PMID: 30467108 DOI: 10.1242/dev.162776] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Extensive studies have shown how bilateral symmetry of the vertebrate embryo is broken during early development, resulting in a molecular left-right bias in the mesoderm. However, how this early asymmetry drives the asymmetric morphogenesis of visceral organs remains poorly understood. The heart provides a striking model of left-right asymmetric morphogenesis, undergoing rightward looping to shape an initially linear heart tube and align cardiac chambers. Importantly, abnormal left-right patterning is associated with severe congenital heart defects, as exemplified in heterotaxy syndrome. Here, we compare the mechanisms underlying the rightward looping of the heart tube in fish, chick and mouse embryos. We propose that heart looping is not only a question of direction, but also one of fine-tuning shape. This is discussed in the context of evolutionary and clinical perspectives.
Collapse
Affiliation(s)
- Audrey Desgrange
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Jean-François Le Garrec
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France.,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| | - Sigolène M Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France .,INSERM UMR1163, Université Paris Descartes, 75015 Paris, France
| |
Collapse
|
26
|
Collins MM, Maischein HM, Dufourcq P, Charpentier M, Blader P, Stainier DY. Pitx2c orchestrates embryonic axis extension via mesendodermal cell migration. eLife 2018; 7:34880. [PMID: 29952749 PMCID: PMC6023614 DOI: 10.7554/elife.34880] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
Pitx2c, a homeodomain transcription factor, is classically known for its left-right patterning role. However, an early wave of pitx2 expression occurs at the onset of gastrulation in several species, indicating a possible earlier role that remains relatively unexplored. Here we show that in zebrafish, maternal-zygotic (MZ) pitx2c mutants exhibit a shortened body axis indicative of convergence and extension (CE) defects. Live imaging reveals that MZpitx2c mutants display less persistent mesendodermal migration during late stages of gastrulation. Transplant data indicate that Pitx2c functions cell non-autonomously to regulate this cell behavior by modulating cell shape and protrusive activity. Using transcriptomic analyses and candidate gene approaches, we identify transcriptional changes in components of the chemokine-ECM-integrin dependent mesendodermal migration network. Together, our results define pathways downstream of Pitx2c that are required during early embryogenesis and reveal novel functions for Pitx2c as a regulator of morphogenesis.
Collapse
Affiliation(s)
- Michelle M Collins
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hans-Martin Maischein
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Pascale Dufourcq
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université Toulouse III - Paul Sabatier, CNRS, Toulouse, France
| | | | - Patrick Blader
- Centre de Biologie du Développement, Centre de Biologie Intégrative, Université Toulouse III - Paul Sabatier, CNRS, Toulouse, France
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
27
|
Versacci P, Pugnaloni F, Digilio MC, Putotto C, Unolt M, Calcagni G, Baban A, Marino B. Some Isolated Cardiac Malformations Can Be Related to Laterality Defects. J Cardiovasc Dev Dis 2018; 5:jcdd5020024. [PMID: 29724030 PMCID: PMC6023464 DOI: 10.3390/jcdd5020024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/22/2022] Open
Abstract
Human beings are characterized by a left–right asymmetric arrangement of their internal organs, and the heart is the first organ to break symmetry in the developing embryo. Aberrations in normal left–right axis determination during embryogenesis lead to a wide spectrum of abnormal internal laterality phenotypes, including situs inversus and heterotaxy. In more than 90% of instances, the latter condition is accompanied by complex and severe cardiovascular malformations. Atrioventricular canal defect and transposition of the great arteries—which are particularly frequent in the setting of heterotaxy—are commonly found in situs solitus with or without genetic syndromes. Here, we review current data on morphogenesis of the heart in human beings and animal models, familial recurrence, and upstream genetic pathways of left–right determination in order to highlight how some isolated congenital heart diseases, very common in heterotaxy, even in the setting of situs solitus, may actually be considered in the pathogenetic field of laterality defects.
Collapse
Affiliation(s)
- Paolo Versacci
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Flaminia Pugnaloni
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Maria Cristina Digilio
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Carolina Putotto
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Marta Unolt
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| | - Giulio Calcagni
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Anwar Baban
- Department of Pediatric Cardiology and Cardiac Surgery, Bambino Gesù Children's Hospital and Research Institute, 00165 Rome, Italy.
| | - Bruno Marino
- Department of Pediatrics, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
28
|
Schubert FR, Singh AJ, Afoyalan O, Kioussi C, Dietrich S. To roll the eyes and snap a bite - function, development and evolution of craniofacial muscles. Semin Cell Dev Biol 2018; 91:31-44. [PMID: 29331210 DOI: 10.1016/j.semcdb.2017.12.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 02/06/2023]
Abstract
Craniofacial muscles, muscles that move the eyes, control facial expression and allow food uptake and speech, have long been regarded as a variation on the general body muscle scheme. However, evidence has accumulated that the function of head muscles, their developmental anatomy and the underlying regulatory cascades are distinct. This article reviews the key aspects of craniofacial muscle and muscle stem cell formation and discusses how this differs from the trunk programme of myogenesis; we show novel RNAseq data to support this notion. We also trace the origin of head muscle in the chordate ancestors of vertebrates and discuss links with smooth-type muscle in the primitive chordate pharynx. We look out as to how the special properties of head muscle precursor and stem cells, in particular their competence to contribute to the heart, could be exploited in regenerative medicine.
Collapse
Affiliation(s)
- Frank R Schubert
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - Arun J Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Oluwatomisin Afoyalan
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, 97331, USA
| | - Susanne Dietrich
- Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, UK.
| |
Collapse
|
29
|
Multiple Roles of Pitx2 in Cardiac Development and Disease. J Cardiovasc Dev Dis 2017; 4:jcdd4040016. [PMID: 29367545 PMCID: PMC5753117 DOI: 10.3390/jcdd4040016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 12/14/2022] Open
Abstract
Cardiac development is a complex morphogenetic process initiated as bilateral cardiogenic mesoderm is specified at both sides of the gastrulating embryo. Soon thereafter, these cardiogenic cells fuse at the embryonic midline configuring a symmetrical linear cardiac tube. Left/right bilateral asymmetry is first detected in the forming heart as the cardiac tube bends to the right, and subsequently, atrial and ventricular chambers develop. Molecular signals emanating from the node confer distinct left/right signalling pathways that ultimately lead to activation of the homeobox transcription factor Pitx2 in the left side of distinct embryonic organ anlagen, including the developing heart. Asymmetric expression of Pitx2 has therefore been reported during different cardiac developmental stages, and genetic deletion of Pitx2 provided evidence of key regulatory roles of this transcription factor during cardiogenesis and thus congenital heart diseases. More recently, impaired Pitx2 function has also been linked to arrhythmogenic processes, providing novel roles in the adult heart. In this manuscript, we provide a state-of-the-art review of the fundamental roles of Pitx2 during cardiogenesis, arrhythmogenesis and its contribution to congenital heart diseases.
Collapse
|
30
|
van Vliet PP, Lin L, Boogerd CJ, Martin JF, Andelfinger G, Grossfeld PD, Evans SM. Tissue specific requirements for WNT11 in developing outflow tract and dorsal mesenchymal protrusion. Dev Biol 2017; 429:249-259. [PMID: 28669819 PMCID: PMC5580348 DOI: 10.1016/j.ydbio.2017.06.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 06/04/2017] [Accepted: 06/18/2017] [Indexed: 12/29/2022]
Abstract
Correct cardiac development is essential for fetal and adult life. Disruptions in a variety of signaling pathways result in congenital heart defects, including outflow and inflow tract defects. We previously found that WNT11 regulates outflow tract development. However, tissue specific requirements for WNT11 in this process remain unknown and whether WNT11 is required for inflow tract development has not been addressed. Here we find that germline Wnt11 null mice also show hypoplasia of the dorsal mesenchymal protrusion (DMP), which is required for atrioventricular septation. Ablation of Wnt11 with myocardial cTnTCre recapitulated outflow tract defects observed in germline Wnt11 null mice, but DMP development was unaffected. In contrast, ablation of Wnt11 with Isl1Cre fully recapitulated both outflow tract and DMP defects of Wnt11 germline nulls. DMP hypoplasia in Wnt11 mutants was associated with reduced proliferation within the DMP, but no evident defects in myocardial differentiation of the DMP. Examination of Pitx2-, Axin2-, or Patched-lacZ reporter mice revealed no alterations in reporter expression, suggesting that WNT11 was required downstream of, or in parallel to, these signaling pathways to regulate DMP formation. These studies revealed a previously unappreciated role for WNT11 for DMP formation and distinct tissue-specific requirements for WNT11 in outflow tract and DMP development.
Collapse
Affiliation(s)
| | - Lizhu Lin
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, USA; Department of Pediatrics, School of Medicine, UCSD, La Jolla, USA
| | - Cornelis J Boogerd
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, USA
| | - James F Martin
- Baylor College of Medicine, Texas Heart Institute, Houston, USA
| | | | - Paul D Grossfeld
- Department of Pediatrics, School of Medicine, UCSD, La Jolla, USA.
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, USA; Department of Medicine, UCSD, La Jolla, USA; Department of Pharmacology, UCSD, La Jolla, USA.
| |
Collapse
|
31
|
Grimes DT, Burdine RD. Left-Right Patterning: Breaking Symmetry to Asymmetric Morphogenesis. Trends Genet 2017; 33:616-628. [PMID: 28720483 DOI: 10.1016/j.tig.2017.06.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 10/19/2022]
Abstract
Vertebrates exhibit striking left-right (L-R) asymmetries in the structure and position of the internal organs. Symmetry is broken by motile cilia-generated asymmetric fluid flow, resulting in a signaling cascade - the Nodal-Pitx2 pathway - being robustly established within mesodermal tissue on the left side only. This pathway impinges upon various organ primordia to instruct their side-specific development. Recently, progress has been made in understanding both the breaking of embryonic L-R symmetry and how the Nodal-Pitx2 pathway controls lateralized cell differentiation, migration, and other aspects of cell behavior, as well as tissue-level mechanisms, that drive asymmetries in organ formation. Proper execution of asymmetric organogenesis is critical to health, making furthering our understanding of L-R development an important concern.
Collapse
Affiliation(s)
- Daniel T Grimes
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| | - Rebecca D Burdine
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
32
|
Pitcairn E, Harris H, Epiney J, Pai VP, Lemire JM, Ye B, Shi NQ, Levin M, McLaughlin KA. Coordinating heart morphogenesis: A novel role for hyperpolarization-activated cyclic nucleotide-gated (HCN) channels during cardiogenesis in Xenopus laevis. Commun Integr Biol 2017; 10:e1309488. [PMID: 28702127 PMCID: PMC5501196 DOI: 10.1080/19420889.2017.1309488] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 12/18/2022] Open
Abstract
Hyperpolarization-activated cyclic-nucleotide gated channel (HCN) proteins are important regulators of both neuronal and cardiac excitability. Among the 4 HCN isoforms, HCN4 is known as a pacemaker channel, because it helps control the periodicity of contractions in vertebrate hearts. Although the physiological role of HCN4 channel has been studied in adult mammalian hearts, an earlier role during embryogenesis has not been clearly established. Here, we probe the embryonic roles of HCN4 channels, providing the first characterization of the expression profile of any of the HCN isoforms during Xenopus laevis development and investigate the consequences of altering HCN4 function on embryonic pattern formation. We demonstrate that both overexpression of HCN4 and injection of dominant-negative HCN4 mRNA during early embryogenesis results in improper expression of key patterning genes and severely malformed hearts. Our results suggest that HCN4 serves to coordinate morphogenetic control factors that provide positional information during heart morphogenesis in Xenopus.
Collapse
Affiliation(s)
- Emily Pitcairn
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Hannah Harris
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Justine Epiney
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Vaibhav P Pai
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Joan M Lemire
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Bin Ye
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Nian-Qing Shi
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Levin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| | - Kelly A McLaughlin
- Department of Biology and Allen Discovery Center at Tufts University, Medford, MA, USA
| |
Collapse
|
33
|
Grant MG, Patterson VL, Grimes DT, Burdine RD. Modeling Syndromic Congenital Heart Defects in Zebrafish. Curr Top Dev Biol 2017; 124:1-40. [DOI: 10.1016/bs.ctdb.2016.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
Current Perspectives in Cardiac Laterality. J Cardiovasc Dev Dis 2016; 3:jcdd3040034. [PMID: 29367577 PMCID: PMC5715725 DOI: 10.3390/jcdd3040034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/23/2016] [Accepted: 12/05/2016] [Indexed: 12/16/2022] Open
Abstract
The heart is the first organ to break symmetry in the developing embryo and onset of dextral looping is the first indication of this event. Looping is a complex process that progresses concomitantly to cardiac chamber differentiation and ultimately leads to the alignment of the cardiac regions in their final topology. Generation of cardiac asymmetry is crucial to ensuring proper form and consequent functionality of the heart, and therefore it is a highly regulated process. It has long been known that molecular left/right signals originate far before morphological asymmetry and therefore can direct it. The use of several animal models has led to the characterization of a complex regulatory network, which invariably converges on the Tgf-β signaling molecule Nodal and its downstream target, the homeobox transcription factor Pitx2. Here, we review current data on the cellular and molecular bases of cardiac looping and laterality, and discuss the contribution of Nodal and Pitx2 to these processes. A special emphasis will be given to the morphogenetic role of Pitx2 and to its modulation of transcriptional and functional properties, which have also linked laterality to atrial fibrillation.
Collapse
|
35
|
Development of the cardiac pacemaker. Cell Mol Life Sci 2016; 74:1247-1259. [PMID: 27770149 DOI: 10.1007/s00018-016-2400-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 01/11/2023]
Abstract
The sinoatrial node (SAN) is the dominant pacemaker of the heart. Abnormalities in SAN formation and function can cause sinus arrhythmia, including sick sinus syndrome and sudden death. A better understanding of genes and signaling pathways that regulate SAN development and function is essential to develop more effective treatment to sinus arrhythmia, including biological pacemakers. In this review, we briefly summarize the key processes of SAN morphogenesis during development, and focus on the transcriptional network that drives SAN development.
Collapse
|
36
|
Ji Y, Buel SM, Amack JD. Mutations in zebrafish pitx2 model congenital malformations in Axenfeld-Rieger syndrome but do not disrupt left-right placement of visceral organs. Dev Biol 2016; 416:69-81. [PMID: 27297886 DOI: 10.1016/j.ydbio.2016.06.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 06/05/2016] [Accepted: 06/06/2016] [Indexed: 12/18/2022]
Abstract
Pitx2 is a conserved homeodomain transcription factor that has multiple functions during embryonic development. Mutations in human PITX2 cause autosomal dominant Axenfeld-Rieger syndrome (ARS), characterized by congenital eye and tooth malformations. Pitx2(-/-) knockout mouse models recapitulate aspects of ARS, but are embryonic lethal. To date, ARS treatments remain limited to managing individual symptoms due to an incomplete understanding of PITX2 function. In addition to regulating eye and tooth development, Pitx2 is a target of a conserved Nodal (TGFβ) signaling pathway that mediates left-right (LR) asymmetry of visceral organs. Based on its highly conserved asymmetric expression domain, the Nodal-Pitx2 axis has long been considered a common denominator of LR development in vertebrate embryos. However, functions of Pitx2 during asymmetric organ morphogenesis are not well understood. To gain new insight into Pitx2 function we used genome editing to create mutations in the zebrafish pitx2 gene. Mutations in the pitx2 homeodomain caused phenotypes reminiscent of ARS, including aberrant development of the cornea and anterior chamber of the eye and reduced or absent teeth. Intriguingly, LR asymmetric looping of the heart and gut was normal in pitx2 mutants. These results suggest conserved roles for Pitx2 in eye and tooth development and indicate Pitx2 is not required for asymmetric looping of zebrafish visceral organs. This work establishes zebrafish pitx2 mutants as a new animal model for investigating mechanisms underlying congenital malformations in ARS and high-throughput drug screening for ARS therapeutics. Additionally, pitx2 mutants present a unique opportunity to identify new genes involved in vertebrate LR patterning. We show Nodal signaling-independent of Pitx2-controls asymmetric expression of the fatty acid elongase elovl6 in zebrafish, pointing to a potential novel pathway during LR organogenesis.
Collapse
Affiliation(s)
- Yongchang Ji
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Sharleen M Buel
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Jeffrey D Amack
- Department of Cell and Developmental Biology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
37
|
Barnes RM, Harris IS, Jaehnig EJ, Sauls K, Sinha T, Rojas A, Schachterle W, McCulley DJ, Norris RA, Black BL. MEF2C regulates outflow tract alignment and transcriptional control of Tdgf1. Development 2016; 143:774-9. [PMID: 26811383 DOI: 10.1242/dev.126383] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 01/19/2016] [Indexed: 01/24/2023]
Abstract
Congenital heart defects are the most common birth defects in humans, and those that affect the proper alignment of the outflow tracts and septation of the ventricles are a highly significant cause of morbidity and mortality in infants. A late differentiating population of cardiac progenitors, referred to as the anterior second heart field (AHF), gives rise to the outflow tract and the majority of the right ventricle and provides an embryological context for understanding cardiac outflow tract alignment and membranous ventricular septal defects. However, the transcriptional pathways controlling AHF development and their roles in congenital heart defects remain incompletely elucidated. Here, we inactivated the gene encoding the transcription factor MEF2C in the AHF in mice. Loss of Mef2c function in the AHF results in a spectrum of outflow tract alignment defects ranging from overriding aorta to double-outlet right ventricle and dextro-transposition of the great arteries. We identify Tdgf1, which encodes a Nodal co-receptor (also known as Cripto), as a direct transcriptional target of MEF2C in the outflow tract via an AHF-restricted Tdgf1 enhancer. Importantly, both the MEF2C and TDGF1 genes are associated with congenital heart defects in humans. Thus, these studies establish a direct transcriptional pathway between the core cardiac transcription factor MEF2C and the human congenital heart disease gene TDGF1. Moreover, we found a range of outflow tract alignment defects resulting from a single genetic lesion, supporting the idea that AHF-derived outflow tract alignment defects may constitute an embryological spectrum rather than distinct anomalies.
Collapse
Affiliation(s)
- Ralston M Barnes
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Ian S Harris
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric J Jaehnig
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Kimberly Sauls
- Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Anabel Rojas
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - William Schachterle
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - David J McCulley
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA
| | - Russell A Norris
- Cardiovascular Developmental Biology Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Brian L Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-3120, USA Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Ye W, Song Y, Huang Z, Zhang Y, Chen Y. Genetic Regulation of Sinoatrial Node Development and Pacemaker Program in the Venous Pole. J Cardiovasc Dev Dis 2015; 2:282-298. [PMID: 26682210 PMCID: PMC4679406 DOI: 10.3390/jcdd2040282] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 11/24/2015] [Indexed: 12/23/2022] Open
Abstract
The definitive sinoatrial node (SAN), the primary pacemaker of the mammalian heart, develops from part of pro-pacemaking embryonic venous pole that expresses both Hcn4 and the transcriptional factor Shox2. It is noted that ectopic pacemaking activities originated from the myocardial sleeves of the pulmonary vein and systemic venous return, both derived from the Shox2+ pro-pacemaking cells in the venous pole, cause atrial fibrillation. However, the developmental link between the pacemaker properties in the embryonic venous pole cells and the SAN remains largely uncharacterized. Furthermore, the genetic program for the development of heterogeneous populations of the SAN is also under-appreciated. Here, we review the literature for a better understanding of the heterogeneous development of the SAN in relation to that of the sinus venosus myocardium and pulmonary vein myocardium. We also attempt to revisit genetic models pertinent to the development of pacemaker activities in the perspective of a Shox2-Nkx2-5 epistatic antagonism. Finally, we describe recent efforts in deciphering the regulatory networks for pacemaker development by genome-wide approaches.
Collapse
Affiliation(s)
- Wenduo Ye
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; E-Mails: (Y.S.); (Z.H.)
| | - Yingnan Song
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; E-Mails: (Y.S.); (Z.H.)
- Southern Center for Biomedical Research, Fujian Normal University, Fuzhou 350108, China; E-Mail:
- Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou 350108, China
| | - Zhen Huang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; E-Mails: (Y.S.); (Z.H.)
- Southern Center for Biomedical Research, Fujian Normal University, Fuzhou 350108, China; E-Mail:
- Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou 350108, China
| | - Yanding Zhang
- Southern Center for Biomedical Research, Fujian Normal University, Fuzhou 350108, China; E-Mail:
- Fujian Key Laboratory of Developmental and Neural Biology, College of Life Science, Fujian Normal University, Fuzhou 350108, China
| | - Yiping Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA; E-Mails: (Y.S.); (Z.H.)
- Southern Center for Biomedical Research, Fujian Normal University, Fuzhou 350108, China; E-Mail:
| |
Collapse
|
39
|
Chamber identity programs drive early functional partitioning of the heart. Nat Commun 2015; 6:8146. [PMID: 26306682 PMCID: PMC4560818 DOI: 10.1038/ncomms9146] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022] Open
Abstract
The vertebrate heart muscle (myocardium) develops from the first heart field (FHF) and expands by adding second heart field (SHF) cells. While both lineages exist already in teleosts, the primordial contributions of FHF and SHF to heart structure and function remain incompletely understood. Here we delineate the functional contribution of the FHF and SHF to the zebrafish heart using the cis-regulatory elements of the draculin (drl) gene. The drl reporters initially delineate the lateral plate mesoderm, including heart progenitors. Subsequent myocardial drl reporter expression restricts to FHF descendants. We harnessed this unique feature to uncover that loss of tbx5a and pitx2 affect relative FHF versus SHF contributions to the heart. High-resolution physiology reveals distinctive electrical properties of each heart field territory that define a functional boundary within the single zebrafish ventricle. Our data establish that the transcriptional program driving cardiac septation regulates physiologic ventricle partitioning, which successively provides mechanical advantages of sequential contraction. The heart forms from combining the first with the second heart field, which in mammals creates left and right ventricle. Here transgenic zebrafish and physiology studies reveal that transcription factors controlling septation in mammals already in teleosts guide muscle coupling by controlling the relative contribution of the two fields to the heart.
Collapse
|
40
|
The pattern of congenital heart defects arising from reduced Tbx5 expression is altered in a Down syndrome mouse model. BMC DEVELOPMENTAL BIOLOGY 2015. [PMID: 26208718 PMCID: PMC4514943 DOI: 10.1186/s12861-015-0080-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Nearly half of all individuals with Down Syndrome (DS) have some type of congenital heart defect (CHD), suggesting that DS sensitizes to CHD but does not cause it. We used a common mouse model of DS, the Ts65Dn mouse, to study the contribution of Tbx5, a known modifier of CHD, to heart defects on a trisomic backgroun. Mice that were heterozygous for a Tbx5 null allele were crossed with Ts65Dn mice. Thoraxes of progeny were fixed in 10% formalin, embedded in paraffin, and sectioned for analysis of CHD. Gene expression in embryonic hearts was examined by quantitative PCR and in situ hybridization. A TBX5 DNA binding site was verified by luciferase assays. METHODS Mice that were heterozygous for a Tbx5 null allele were crossed with Ts65Dn mice. Thoraxes of progeny were fixed in 10% formalin, embedded in paraffin, and sectioned for analysis of CHD. Gene expression in embryonic hearts was examined by quantitative PCR and in situ hybridization. A TBX5 DNA binding site was verified by luciferase assays. RESULTS We crossed mice that were heterozygous for a Tbx5 null allele with Ts65Dn mice. Mice that were trisomic and carried the Tbx5 mutation (Ts65Dn;Tbx5 (+/-) ) had a significantly increased incidence of overriding aorta compared to their euploid littermates. Ts65Dn;Tbx5 (+/-) mice also showed reduced expression of Pitx2, a molecular marker for the left atrium. Transcript levels of the trisomic Adamts1 gene were decreased in Tbx5 (+/-) mice compared to their euploid littermates. Evidence of a valid binding site for TBX5 upstream of the trisomic Adamts1 locus was also shown. CONCLUSION Haploinsufficiency of Tbx5 and trisomy affects alignment of the aorta and this effect may stem from deviations from normal left-right patterning in the heart. We have unveiled a previously unknown interaction between the Tbx5 gene and trisomy, suggesting a connection between Tbx5 and trisomic genes important during heart development.
Collapse
|
41
|
Narematsu M, Kamimura T, Yamagishi T, Fukui M, Nakajima Y. Impaired development of left anterior heart field by ectopic retinoic acid causes transposition of the great arteries. J Am Heart Assoc 2015; 4:jah3958. [PMID: 25929268 PMCID: PMC4599416 DOI: 10.1161/jaha.115.001889] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Background Transposition of the great arteries is one of the most commonly diagnosed conotruncal heart defects at birth, but its etiology is largely unknown. The anterior heart field (AHF) that resides in the anterior pharyngeal arches contributes to conotruncal development, during which heart progenitors that originated from the left and right AHF migrate to form distinct conotruncal regions. The aim of this study is to identify abnormal AHF development that causes the morphology of transposition of the great arteries. Methods and Results We placed a retinoic acid–soaked bead on the left or the right or on both sides of the AHF of stage 12 to 14 chick embryos and examined the conotruncal heart defect at stage 34. Transposition of the great arteries was diagnosed at high incidence in embryos for which a retinoic acid–soaked bead had been placed in the left AHF at stage 12. Fluorescent dye tracing showed that AHF exposed to retinoic acid failed to contribute to conotruncus development. FGF8 and Isl1 expression were downregulated in retinoic acid–exposed AHF, and differentiation and expansion of cardiomyocytes were suppressed in cultured AHF in medium supplemented with retinoic acid. Conclusions The left AHF at the early looped heart stage, corresponding to Carnegie stages 10 to 11 (28 to 29 days after fertilization) in human embryos, is the region of the impediment that causes the morphology of transposition of the great arteries.
Collapse
Affiliation(s)
- Mayu Narematsu
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan (M.N., T.K., T.Y., Y.N.)
| | - Tatsuya Kamimura
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan (M.N., T.K., T.Y., Y.N.)
| | - Toshiyuki Yamagishi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan (M.N., T.K., T.Y., Y.N.)
| | - Mitsuru Fukui
- Laboratory of Statics, Graduate School of Medicine, Osaka City University, Osaka, Japan (M.F.)
| | - Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan (M.N., T.K., T.Y., Y.N.)
| |
Collapse
|
42
|
Zhao CM, Peng LY, Li L, Liu XY, Wang J, Zhang XL, Yuan F, Li RG, Qiu XB, Yang YQ. PITX2 Loss-of-Function Mutation Contributes to Congenital Endocardial Cushion Defect and Axenfeld-Rieger Syndrome. PLoS One 2015; 10:e0124409. [PMID: 25893250 PMCID: PMC4404345 DOI: 10.1371/journal.pone.0124409] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 03/13/2015] [Indexed: 12/17/2022] Open
Abstract
Congenital heart disease (CHD), the most common type of birth defect, is still the leading non-infectious cause of infant morbidity and mortality in humans. Aggregating evidence demonstrates that genetic defects are involved in the pathogenesis of CHD. However, CHD is genetically heterogeneous and the genetic components underpinning CHD in an overwhelming majority of patients remain unclear. In the present study, the coding exons and flanking introns of the PITX2 gene, which encodes a paired-like homeodomain transcription factor 2essential for cardiovascular morphogenesis as well as maxillary facial development, was sequenced in 196 unrelated patients with CHD and subsequently in the mutation carrier's family members available. As a result, a novel heterozygous PITX2 mutation, p.Q102X for PITX2a, or p.Q148X for PITX2b, or p.Q155X for PITX2c, was identified in a family with endocardial cushion defect (ECD) and Axenfeld-Rieger syndrome (ARS). Genetic analysis of the pedigree showed that the nonsense mutation co-segregated with ECD and ARS transmitted in an autosomal dominant pattern with complete penetrance. The mutation was absent in 800 control chromosomes from an ethnically matched population. Functional analysis by using a dual-luciferase reporter assay system revealed that the mutant PITX2 had no transcriptional activity and that the mutation eliminated synergistic transcriptional activation between PITX2 and NKX2.5, another transcription factor pivotal for cardiogenesis. To our knowledge, this is the first report on the association of PITX2 loss-of-function mutation with increased susceptibility to ECD and ARS. The findings provide novel insight into the molecular mechanisms underpinning ECD and ARS, suggesting the potential implications for the antenatal prophylaxis and personalized treatment of CHD and ARS.
Collapse
Affiliation(s)
- Cui-Mei Zhao
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
- Division of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Lu-Ying Peng
- Division of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Li Li
- Division of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Xing-Yuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juan Wang
- Department of Cardiology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xian-Ling Zhang
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fang Yuan
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ruo-Gu Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xing-Biao Qiu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi-Qing Yang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Cardiovascular Research Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
43
|
Kong P, Racedo SE, Macchiarulo S, Hu Z, Carpenter C, Guo T, Wang T, Zheng D, Morrow BE. Tbx1 is required autonomously for cell survival and fate in the pharyngeal core mesoderm to form the muscles of mastication. Hum Mol Genet 2014; 23:4215-31. [PMID: 24705356 DOI: 10.1093/hmg/ddu140] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Velo-cardio-facial/DiGeorge syndrome, also known as 22q11.2 deletion syndrome, is a congenital anomaly disorder characterized by craniofacial anomalies including velo-pharyngeal insufficiency, facial muscle hypotonia and feeding difficulties, in part due to hypoplasia of the branchiomeric muscles. Inactivation of both alleles of mouse Tbx1, encoding a T-box transcription factor, deleted on chromosome 22q11.2, results in reduction or loss of branchiomeric muscles. To identify downstream pathways, we performed gene profiling of microdissected pharyngeal arch one (PA1) from Tbx1(+/+) and Tbx1(-/-) embryos at stages E9.5 (somites 20-25) and E10.5 (somites 30-35). Basic helix-loop-helix (bHLH) transcription factors were reduced, while secondary heart field genes were increased in expression early and were replaced by an increase in expression of cellular stress response genes later, suggesting a change in gene expression patterns or cell populations. Lineage tracing studies using Mesp1(Cre) and T-Cre drivers showed that core mesoderm cells within PA1 were present at E9.5 but were greatly reduced by E10.5 in Tbx1(-/-) embryos. Using Tbx1(Cre) knock-in mice, we found that cells are lost due to apoptosis, consistent with increase in expression of cellular stress response genes at E10.5. To determine whether Tbx1 is required autonomously in the core mesoderm, we used Mesp1(Cre) and T-Cre mesodermal drivers in combination with inactivate Tbx1 and found reduction or loss of branchiomeric muscles from PA1. These mechanistic studies inform us that Tbx1 is required upstream of key myogenic genes needed for core mesoderm cell survival and fate, between E9.5 and E10.5, resulting in formation of the branchiomeric muscles.
Collapse
Affiliation(s)
- Ping Kong
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Stephania Macchiarulo
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Zunju Hu
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Courtney Carpenter
- Department of Surgery, Montefiore Medical Center, 111 East 210th Street, Bronx, NY 10467, USA
| | - Tingwei Guo
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA
| | - Tao Wang
- Department of Epidemiology and Population Health, Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA and
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA, Departments of Neurology and Neuroscience, Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, 1301 Morris Park Avenue, Bronx, NY 10461, USA,
| |
Collapse
|
44
|
Vela I, Morrissey C, Zhang X, Chen S, Corey E, Strutton GM, Nelson CC, Nicol DL, Clements JA, Gardiner EM. PITX2 and non-canonical Wnt pathway interaction in metastatic prostate cancer. Clin Exp Metastasis 2014; 31:199-211. [PMID: 24162257 DOI: 10.1007/s10585-013-9620-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 10/06/2013] [Indexed: 10/26/2022]
Abstract
The non-canonical Wnt pathway, a regulator of cellular motility and morphology, is increasingly implicated in cancer metastasis. In a quantitative PCR array analysis of 84 Wnt pathway associated genes, both non-canonical and canonical pathways were activated in primary and metastatic tumors relative to normal prostate. Expression of the Wnt target gene PITX2 in a prostate cancer (PCa) bone metastasis was strikingly elevated over normal prostate (over 2,000-fold) and primary prostate cancer (over 200-fold). The elevation of PITX2 protein was also evident on tissue microarrays, with strong PITX2 immunostaining in PCa skeletal and, to a lesser degree, soft tissue metastases. PITX2 is associated with cell migration during normal tissue morphogenesis. In our studies, overexpression of individual PITX2A/B/C isoforms stimulated PC-3 PCa cell motility, with the PITX2A isoform imparting a specific motility advantage in the presence of non-canonical Wnt5a stimulation. Furthermore, PITX2 specific shRNA inhibited PC-3 cell migration toward bone cell derived chemoattractant. These experimental results support a pivotal role of PITX2A and non-canonical Wnt signaling in enhancement of PCa cell motility, suggest PITX2 involvement in homing of PCa to the skeleton, and are consistent with a role for PITX2 in PCa metastasis to soft and bone tissues. Our findings, which significantly expand previous evidence that PITX2 is associated with risk of PCa biochemical recurrence, indicate that variation in PITX2 expression accompanies and may promote prostate tumor progression and metastasis.
Collapse
Affiliation(s)
- I Vela
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Franco D, Christoffels VM, Campione M. Homeobox transcription factor Pitx2: The rise of an asymmetry gene in cardiogenesis and arrhythmogenesis. Trends Cardiovasc Med 2014; 24:23-31. [PMID: 23953978 DOI: 10.1016/j.tcm.2013.06.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 01/05/2023]
Abstract
The homeobox transcription factor Pitx2 displays a highly specific expression pattern during embryogenesis. Gain and loss of function experiments have unraveled its pivotal role in left-right signaling. Conditional deletion in mice has demonstrated a complex and intricate role for Pitx2 in distinct aspects of cardiac development and more recently a link to atrial fibrillation has been proposed based on genome-wide association studies. In this review we will revise the role of Pitx2 in the developing heart, starting from the early events of left-right determination followed by its role in cardiac morphogenesis and ending with its role in cardiac arrhythmogenesis.
Collapse
Affiliation(s)
- Diego Franco
- Department of Experimental Biology B3-362, University of Jaén, Jaen 23071, Spain.
| | | | - Marina Campione
- CNR-Institute of Neurosciences, Department of Biomedical Sciences, University of Padua, Padua, Italy
| |
Collapse
|
46
|
Ma HY, Xu J, Eng D, Gross MK, Kioussi C. Pitx2-mediated cardiac outflow tract remodeling. Dev Dyn 2013; 242:456-68. [PMID: 23361844 PMCID: PMC3673775 DOI: 10.1002/dvdy.23934] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2013] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Heart morphogenesis involves sequential anatomical changes from a linear tube of a single channel peristaltic pump to a four-chamber structure with two channels controlled by one-way valves. The developing heart undergoes continuous remodeling, including septation. RESULTS Pitx2-null mice are characterized by cardiac septational defects of the atria, ventricles, and outflow tract. Pitx2-null mice also exhibited a short outflow tract, including unseptated conus and deformed endocardial cushions. Cushions were characterized with a jelly-like structure, rather than the distinct membrane-looking leaflets, indicating that endothelial mesenchymal transition was impaired in Pitx2(-/-) embryos. Mesoderm cells from the branchial arches and neural crest cells from the otic region contribute to the development of the endocardial cushions, and both were reduced in number. Members of the Fgf and Bmp families exhibited altered expression levels in the mutants. CONCLUSIONS We suggest that Pitx2 is involved in the cardiac outflow tract septation by promoting and/or maintaining the number and the remodeling process of the mesoderm progenitor cells. Pitx2 influences the expression of transcription factors and signaling molecules involved in the differentiation of the cushion mesenchyme during heart development.
Collapse
Affiliation(s)
- Hsiao-Yen Ma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Jun Xu
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Diana Eng
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Michael K. Gross
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Chrissa Kioussi
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
47
|
Knopp P, Figeac N, Fortier M, Moyle L, Zammit PS. Pitx genes are redeployed in adult myogenesis where they can act to promote myogenic differentiation in muscle satellite cells. Dev Biol 2013; 377:293-304. [PMID: 23438814 DOI: 10.1016/j.ydbio.2013.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 01/29/2013] [Accepted: 02/15/2013] [Indexed: 11/17/2022]
Abstract
Skeletal muscle retains a resident stem cell population called satellite cells. Although mitotically quiescent in mature muscle, satellite cells can be activated to produce myoblast progeny to generate myonuclei for skeletal muscle homoeostasis, hypertrophy and repair. Regulation of satellite cell function in adult requires redeployment of many of the regulatory networks fundamental to developmental myogenesis. Involved in such control of muscle stem cell fate in embryos are members of the Pitx gene family of bicoid-class homeodomain proteins. Here, we investigated the expression and function of all three Pitx genes in muscle satellite cells of adult mice. Endogenous Pitx1 was undetectable, whilst Pitx2a, Pitx2b and Pitx2c were at low levels in proliferating satellite cells, but increased during the early stages of myogenic differentiation. By contrast, proliferating satellite cells expressed robust amounts of Pitx3, with levels then decreasing as cells differentiated, although Pitx3 remained expressed in unfused myoblasts. To examine the role of Pitx genes in satellite cell function, retroviral-mediated expression of Pitx1, all Pitx2 isoforms or Pitx3, was used. Constitutive expression of any Pitx isoform suppressed satellite cell proliferation, with the cells undergoing enhanced myogenic differentiation. Conversely, myogenic differentiation into multinucleated myotubes was decreased when Pitx2 or Pitx3 levels were reduced using siRNA. Together, our results show that Pitx genes play a role in regulating satellite cell function during myogenesis in adult.
Collapse
Affiliation(s)
- Paul Knopp
- King's College London, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, London, SE1 1UL, England, UK
| | | | | | | | | |
Collapse
|
48
|
Neeb Z, Lajiness JD, Bolanis E, Conway SJ. Cardiac outflow tract anomalies. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2013; 2:499-530. [PMID: 24014420 DOI: 10.1002/wdev.98] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mature outflow tract (OFT) is, in basic terms, a short conduit. It is a simple, although vital, connection situated between contracting muscular heart chambers and a vast embryonic vascular network. Unfortunately, it is also a focal point underlying many multifactorial congenital heart defects (CHDs). Through the use of various animal models combined with human genetic investigations, we are beginning to comprehend the molecular and cellular framework that controls OFT morphogenesis. Clear roles of neural crest cells (NCC) and second heart field (SHF) derivatives have been established during OFT formation and remodeling. The challenge now is to determine how the SHF and cardiac NCC interact, the complex reciprocal signaling that appears to be occurring at various stages of OFT morphogenesis, and finally how endocardial progenitors and primary heart field (PHF) communicate with both these colonizing extra-cardiac lineages. Although we are beginning to understand that this dance of progenitor populations is wonderfully intricate, the underlying pathogenesis and the spatiotemporal cell lineage interactions remain to be fully elucidated. What is now clear is that OFT alignment and septation are independent processes, invested via separate SHF and cardiac neural crest (CNC) lineages. This review will focus on our current understanding of the respective contributions of the SHF and CNC lineage during OFT development and pathogenesis.
Collapse
Affiliation(s)
- Zachary Neeb
- Developmental Biology and Neonatal Medicine Program, HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
49
|
Pharyngeal mesoderm regulatory network controls cardiac and head muscle morphogenesis. Proc Natl Acad Sci U S A 2012; 109:18839-44. [PMID: 23112163 DOI: 10.1073/pnas.1208690109] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The search for developmental mechanisms driving vertebrate organogenesis has paved the way toward a deeper understanding of birth defects. During embryogenesis, parts of the heart and craniofacial muscles arise from pharyngeal mesoderm (PM) progenitors. Here, we reveal a hierarchical regulatory network of a set of transcription factors expressed in the PM that initiates heart and craniofacial organogenesis. Genetic perturbation of this network in mice resulted in heart and craniofacial muscle defects, revealing robust cross-regulation between its members. We identified Lhx2 as a previously undescribed player during cardiac and pharyngeal muscle development. Lhx2 and Tcf21 genetically interact with Tbx1, the major determinant in the etiology of DiGeorge/velo-cardio-facial/22q11.2 deletion syndrome. Furthermore, knockout of these genes in the mouse recapitulates specific cardiac features of this syndrome. We suggest that PM-derived cardiogenesis and myogenesis are network properties rather than properties specific to individual PM members. These findings shed new light on the developmental underpinnings of congenital defects.
Collapse
|
50
|
Scherptong RWC, Jongbloed MRM, Wisse LJ, Vicente-Steijn R, Bartelings MM, Poelmann RE, Schalij MJ, Gittenberger-De Groot AC. Morphogenesis of outflow tract rotation during cardiac development: the pulmonary push concept. Dev Dyn 2012; 241:1413-22. [PMID: 22826212 DOI: 10.1002/dvdy.23833] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2012] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Understanding of cardiac outflow tract (OFT) remodeling is essential to explain repositioning of the aorta and pulmonary orifice. In wild type embryos (E9.5-14.5), second heart field contribution (SHF) to the OFT was studied using expression patterns of Islet 1, Nkx2.5, MLC-2a, WT-1, and 3D-reconstructions. Abnormal remodeling was studied in VEGF120/120 embryos. RESULTS In wild type, Islet 1 and Nkx2.5 positive myocardial precursors formed an asymmetric elongated column almost exclusively at the pulmonary side of the OFT up to the pulmonary orifice. In VEGF120/120 embryos, the Nkx2.5-positive mesenchymal population was disorganized with a short extension along the pulmonary OFT. CONCLUSIONS We postulate that normally the pulmonary trunk and orifice are pushed in a higher and more frontal position relative to the aortic orifice by asymmetric addition of SHF-myocardium. Deficient or disorganized right ventricular OFT expansion might explain cardiac malformations with abnormal position of the great arteries, such as double outlet right ventricle.
Collapse
|