1
|
Han X, Li Y, Wang E, Zhu H, Huang X, Pu W, Zhang M, Liu K, Zhao H, Liu Z, Zhao Y, Shen L, Li Y, Yang X, Wang QD, Ma X, Shen R, O Lui K, Wang L, He B, Zhou B. Exploring Origin-Dependent Susceptibility of Smooth Muscle Cells to Aortic Diseases Through Intersectional Genetics. Circulation 2025; 151:1248-1267. [PMID: 39925267 DOI: 10.1161/circulationaha.124.070782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND The developmental diversity among smooth muscle cells (SMCs) plays a crucial role in segment-specific aortic diseases. However, traditional genetic approaches are inadequate for enabling in vivo analysis of disease susceptibility associated with cellular origin. There is an urgent need to build genetic technologies that target different developmental origins to investigate the mechanisms of aortopathies, thereby facilitating the development of effective therapeutics. METHODS To address this challenge, we developed an advanced dual recombinase-mediated intersectional genetic system, specifically designed to precisely target SMCs from various developmental origins in mice. Specifically, we used Isl1-Dre, Wnt1-Dre, Meox1-DreER, and Upk3b-Dre to target SMC progenitors from the second heart field, cardiac neural crest, somites, and mesothelium, respectively. This system was combined with single-cell RNA sequencing to investigate the impact of TGF-β (transforming growth factor-β) signaling in different segments of the aorta by selectively knocking out Tgfbr2 in the ascending aorta and Smad4 in the aortic arch, respectively. RESULTS Through intersectional genetic approaches, we use the Myh11-Cre(ER) driver along with origin-specific Dre drivers to trace cells of diverse developmental origins within the SMC population. We found that a deficiency of Tgfbr2 in SMCs of the ascending aorta leads to aneurysm formation in this specific region. We also demonstrate the critical role of Smad4 in preserving aortic wall integrity and homeostasis in SMCs of the aortic arch. CONCLUSIONS Our approach to genetically targeting SMC subtypes provides a novel platform for exploring origin-dependent or location-specific aortic vascular diseases. This genetic system enables comprehensive analysis of contributions from different cell lineages to SMC behavior and pathology, thereby paving the way for targeted research and therapeutic interventions in the future.
Collapse
MESH Headings
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptor, Transforming Growth Factor-beta Type II/deficiency
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Genetic Predisposition to Disease
- Aorta/metabolism
- Aorta/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Huan Zhu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Xiuzhen Huang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Wenjuan Pu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Mingjun Zhang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
| | - Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Zixin Liu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yufei Zhao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Yan Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China (Y.L.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China (Y.L.)
| | - Xiao Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, China (X.Y.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.-D.W.)
| | - Xin Ma
- Department of Pharmacology, Wuxi School of Medicine, Jiangnan University, China (X.M.)
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, China (R.S.)
| | - Kathy O Lui
- CAS CEMCS-CUHK Joint Laboratories, Department of Chemical Pathology; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Chinese University of Hong Kong, China (K.O.L.)
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
- Department of Vascular Surgery (Xiamen), Zhongshan Hospital, Fudan University, Xiamen, China (L.W.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (B.Z.)
| |
Collapse
|
2
|
Torii S, Nagaharu K, Nakanishi N, Usui H, Hori Y, Hirose K, Toyosawa S, Morii E, Narushima M, Kubota Y, Nakagawa O, Imanaka-Yoshida K, Maruyama K. Embryological cellular origins and hypoxia-mediated mechanisms in PIK3CA-driven refractory vascular malformations. EMBO Mol Med 2025:10.1038/s44321-025-00235-1. [PMID: 40234712 DOI: 10.1038/s44321-025-00235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025] Open
Abstract
Congenital vascular malformations, affecting 0.5% of the population, often occur in the head and neck, complicating treatment due to the critical functions in these regions. Our previous research identified distinct developmental origins for blood and lymphatic vessels in these areas, tracing them to the cardiopharyngeal mesoderm (CPM), which contributes to the development of the head, neck, and cardiovascular system in both mouse and human embryos. In this study, we investigated the pathogenesis of these malformations by expressing Pik3caH1047R in the CPM. Mice expressing Pik3caH1047R in the CPM developed vascular abnormalities restricted to the head and neck. Single-cell RNA sequencing revealed that Pik3caH1047R upregulates Vegf-a expression in endothelial cells through HIF-mediated hypoxia signaling. Human samples supported these findings, showing elevated HIF-1α and VEGF-A in malformed vessels. Notably, inhibition of HIF-1α and VEGF-A in the mouse model significantly reduced abnormal vasculature. These results highlight the role of embryonic origins and hypoxia-driven mechanisms in vascular malformations, providing a foundation for the development of therapies targeting these difficult-to-treat conditions.
Collapse
Affiliation(s)
- Sota Torii
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Keiki Nagaharu
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, 514-8507, Japan
| | - Nanako Nakanishi
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Hidehito Usui
- Department of Surgery, Kanagawa Children's Medical Center, 2-138-4, Mutsukawa, Minami-ku, Yokohama, Kanagawa, Japan
| | - Yumiko Hori
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Central Laboratory and Surgical Pathology, NHO Osaka National Hospital, 2-1-14 Hoenzaka, Chuo-ku, Osaka, 540-0006, Japan
| | - Katsutoshi Hirose
- Department of Oral and Maxillofacial Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Toyosawa
- Department of Oral and Maxillofacial Pathology, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eiichi Morii
- Department of Pathology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Mitsunaga Narushima
- Department of Plastic and Reconstructive Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shimmachi, Suita, Osaka, 564-8565, Japan
| | - Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Kazuaki Maruyama
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
3
|
Burnicka-Turek O, Trampel KA, Laforest B, Broman MT, Yang XH, Khan Z, Rytkin E, Li B, Schaffer E, Gadek M, Shen KM, Efimov IR, Moskowitz IP. Coordinated Tbx3/Tbx5 transcriptional control of the adult ventricular conduction system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.29.610377. [PMID: 39257760 PMCID: PMC11383707 DOI: 10.1101/2024.08.29.610377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The cardiac conduction system (CCS) orchestrates the electrical impulses that enable coordinated contraction of the cardiac chambers. The T-box transcription factors TBX3 and TBX5 are required for cardiac conduction system development and associated with overlapping and distinct human cardiac conduction system diseases. We evaluated the coordinated role of Tbx3 and Tbx5 in the murine ventricular conduction system (VCS). We engineered a compound Tbx3:Tbx5 conditional knockout allele for both genes located in cis on mouse chromosome 5. Conditional deletion of both T-box transcriptional factors in the ventricular conduction system, using the VCS-specific MinK:Cre, caused loss of VCS function and molecular identity. Combined Tbx3 and Tbx5 deficiency in the adult VCS led to conduction defects, including prolonged PR and QRS intervals and elevated susceptibility to ventricular tachycardia. These electrophysiological defects occurred prior to detectable alterations in cardiac contractility or histologic morphology, indicative of a primary conduction system defect. Tbx3:Tbx5 double knockout VCS cardiomyocytes revealed a transcriptional shift towards non-CCS-specialized working myocardium, indicating a change to their cellular identity. Furthermore, optical mapping revealed a loss of VCS-specific conduction system propagation. Collectively, these findings indicate that Tbx3 and Tbx5 coordinate to control VCS molecular fate and function, with implications for understanding cardiac conduction disorders in humans.
Collapse
Affiliation(s)
- Ozanna Burnicka-Turek
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Katy A. Trampel
- Departments of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Brigitte Laforest
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Michael T. Broman
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, IL, 60637, USA
| | - Xinan H. Yang
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Zoheb Khan
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Eric Rytkin
- Departments of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Binjie Li
- Departments of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Ella Schaffer
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Margaret Gadek
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Kaitlyn M. Shen
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Igor R. Efimov
- Departments of Biomedical Engineering, Northwestern University, Chicago, IL 60611, USA
| | - Ivan P. Moskowitz
- Departments of Pediatrics, Pathology, and Human Genetics, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Shukla S, Jana S, Sanford N, Lee CY, Liu L, Cheng P, Quertermous T, Dichek DA. Single-Cell Transcriptomics Identifies Selective Lineage-Specific Regulation of Genes in Aortic Smooth Muscle Cells in Mice. Arterioscler Thromb Vasc Biol 2025; 45:e15-e29. [PMID: 39744838 PMCID: PMC11875902 DOI: 10.1161/atvbaha.124.321482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
BACKGROUND Smooth muscle cells (SMCs) of the proximal thoracic aorta are derived from second heart field (SHF) and cardiac neural crest (CNC) lineages. Recent studies, both in vitro and in vivo, have implied relevance of lineage-specific SMC functions in the pathophysiology of thoracic aortic diseases; however, whether 2 lineage-derived SMCs have any predisposed transcriptional differences in the control aorta remains unexplored. METHODS Single-cell RNA sequencing and single-nucleus assay for transposase-accessible chromatin sequencing were performed on isolated cells from the aortic root and ascending aortas of 14-week-old SHF-traced (Mef2c-Cre+/0-Yfp+/0) and CNC-traced (Wnt1-Cre+/0-Yfp+/0) male mice. RNA in situ hybridization was performed for spatial expression of selected differentially expressed genes (DEGs) of both lineages. RESULTS Lineage stratification of SMCs in the proximal thoracic aorta was identified using antibody-based immunofluorescence staining. Single-cell RNA sequencing recognized 12 consistently upregulated DEGs (Des, Tnnt2, Hand2os1, Psd, Gpc3, Meis2, Dcn, Gm34030, Palld, Nrtn, Lum, and Cfh) in SHF-derived SMCs and 9 consistently upregulated DEGs (Ccn5, Ccdc42, Tes, Eln, Aebp1, Galnt6, Ccn2, Aopep, and Wtip) in CNC-derived SMCs. RNA in situ hybridization validated upregulated expressions of selective SHF-specific DEGs at the aortic root. We found SHF-derived SMCs contain a distinct, large subpopulation of SMCs that is enriched with Des and Tnnt2 expressions. Single-nucleus assay for transposase-accessible chromatin analysis further confirmed higher chromosomal accessibility for upregulated DEGs of SHF-derived SMCs. CONCLUSIONS The present study recognizes the presence of limited but distinct transcriptomic differences between CNC-derived and SHF-derived SMCs in the control proximal thoracic aorta.
Collapse
Affiliation(s)
- Shalabh Shukla
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Sayantan Jana
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Nicole Sanford
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Chloe Y Lee
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Li Liu
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, CA, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, CA, USA
| | - David A Dichek
- Division of Cardiology, Department of Medicine, University of Washington, WA, USA
| |
Collapse
|
5
|
Vitali HE, Kuschel B, Sherpa C, Jones BW, Jacob N, Madiha SA, Elliott S, Dziennik E, Kreun L, Conatser C, Bhetwal BP, Sharma B. Hypoxia regulate developmental coronary angiogenesis potentially through VEGF-R2- and SOX17-mediated signaling. Dev Dyn 2025; 254:174-188. [PMID: 39360476 PMCID: PMC11810610 DOI: 10.1002/dvdy.750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/13/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND The development of coronary vessels in embryonic mouse heart involves various progenitor populations, including sinus venosus (SV), endocardium, and proepicardium. ELA/APJ signaling is known to regulate coronary growth from the SV, whereas VEGF-A/VEGF-R2 signaling controls growth from the endocardium. Previous studies suggest hypoxia might regulate coronary growth, but its specific downstream pathways are unclear. In this study, we further investigated the role of hypoxia and have identified SOX17- and VEGF-R2-mediated signaling as the potential downstream pathways in its regulation of developmental coronary angiogenesis. RESULTS HIF-1α stabilization by knocking out von Hippel Lindau (VHL) protein in the myocardium (cKO) disrupted normal coronary angiogenesis in embryonic mouse hearts, resembling patterns of accelerated coronary growth. VEGF-R2 expression was increased in coronary endothelial cells under hypoxia in vitro and in VHL cKO hearts in vivo. Similarly, SOX17 expression was increased in the VHL cKO hearts, while its knockout in the endocardium disrupted normal coronary growth. CONCLUSION These findings provide further evidence that hypoxia regulates developmental coronary growth potentially through VEGF-R2 and SOX17 pathways, shedding light on mechanisms of coronary vessel development.
Collapse
Affiliation(s)
- Halie E. Vitali
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Bryce Kuschel
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Chhiring Sherpa
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | - Nisha Jacob
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Syeda A. Madiha
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Sam Elliott
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Eddie Dziennik
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Lily Kreun
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Cora Conatser
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Bhupal P. Bhetwal
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ 07110
| | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN 47306
| |
Collapse
|
6
|
Arriagada C, Lin E, Schonning M, Astrof S. Mesodermal fibronectin controls cell shape, polarity, and mechanotransduction in the second heart field during cardiac outflow tract development. Dev Cell 2025; 60:62-84.e7. [PMID: 39413783 PMCID: PMC11706711 DOI: 10.1016/j.devcel.2024.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024]
Abstract
Failure in the elongation of the cardiac outflow tract (OFT) results in congenital heart disease due to the misalignment of the great arteries with the left and right ventricles. The OFT lengthens via the accretion of progenitors from the second heart field (SHF). SHF cells are exquisitely regionalized and organized into an epithelial-like layer, forming the dorsal pericardial wall (DPW). Tissue tension, cell polarity, and proliferation within the DPW are important for the addition of SHF-derived cells to the heart and OFT elongation. However, the genes controlling these processes are not completely characterized. Using conditional mutagenesis in the mouse, we show that fibronectin (FN1) synthesized by the mesoderm coordinates multiple cellular behaviors in the anterior DPW. FN1 is enriched in the anterior DPW and plays a role in OFT elongation by maintaining a balance between pro- and anti-adhesive cell-extracellular matrix (ECM) interactions and controlling DPW cell shape, polarity, cohesion, proliferation, and mechanotransduction.
Collapse
Affiliation(s)
- Cecilia Arriagada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Evan Lin
- Princeton Day School, Princeton, NJ, USA
| | - Michael Schonning
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers Biomedical and Health Sciences, 185 South Orange Ave., Newark, NJ 07103, USA.
| |
Collapse
|
7
|
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: 4] [Impact Index Per Article: 4.0] [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
|
8
|
Argiro L, Chevalier C, Choquet C, Nandkishore N, Ghata A, Baudot A, Zaffran S, Lescroart F. Gastruloids are competent to specify both cardiac and skeletal muscle lineages. Nat Commun 2024; 15:10172. [PMID: 39580459 PMCID: PMC11585638 DOI: 10.1038/s41467-024-54466-w] [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] [Received: 05/17/2023] [Accepted: 11/12/2024] [Indexed: 11/25/2024] Open
Abstract
Cardiopharyngeal mesoderm contributes to the formation of the heart and head muscles. However, the mechanisms governing cardiopharyngeal mesoderm specification remain unclear. Here, we reproduce cardiopharyngeal mesoderm specification towards cardiac and skeletal muscle lineages with gastruloids from mouse embryonic stem cells. By conducting a comprehensive temporal analysis of cardiopharyngeal mesoderm development and differentiation in gastruloids compared to mouse embryos, we present the evidence for skeletal myogenesis in gastruloids. We identify different subpopulations of cardiomyocytes and skeletal muscles, the latter of which most likely correspond to different states of myogenesis with "head-like" and "trunk-like" skeletal myoblasts. In this work, we unveil the potential of gastruloids to undergo specification into both cardiac and skeletal muscle lineages, allowing the investigation of the mechanisms of cardiopharyngeal mesoderm differentiation in development and how this could be affected in congenital diseases.
Collapse
Affiliation(s)
- Laurent Argiro
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Céline Chevalier
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Caroline Choquet
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Nitya Nandkishore
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
- Department of Biotechnology, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu, India
| | - Adeline Ghata
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Anaïs Baudot
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France
| | - Stéphane Zaffran
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| | - Fabienne Lescroart
- Aix-Marseille Univ, INSERM, Marseille Medical Genetics (MMG), Marseille, France.
| |
Collapse
|
9
|
Guijarro C, Song S, Aigouy B, Clément R, Villoutreix P, Kelly RG. Single-cell morphometrics reveals T-box gene-dependent patterns of epithelial tension in the Second Heart field. Nat Commun 2024; 15:9512. [PMID: 39496595 PMCID: PMC11535409 DOI: 10.1038/s41467-024-53612-8] [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] [Received: 07/28/2023] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
The vertebrate heart tube extends by progressive addition of epithelial second heart field (SHF) progenitor cells from the dorsal pericardial wall. The interplay between epithelial mechanics and genetic mechanisms during SHF deployment is unknown. Here, we present a quantitative single-cell morphometric analysis of SHF cells during heart tube extension, including force inference analysis of epithelial stress. Joint spatial Principal Component Analysis reveals that cell orientation and stress direction are the main parameters defining apical cell morphology and distinguishes cells adjacent to the arterial and venous poles. Cell shape and mechanical forces display a dynamic relationship during heart tube formation. Moreover, while the T-box transcription factor Tbx1 is necessary for cell orientation towards the arterial pole, activation of Tbx5 in the posterior SHF correlates with the establishment of epithelial stress and SHF deletion of Tbx5 relaxes the progenitor epithelium. Integrating findings from cell-scale feature patterning and mechanical stress provides new insights into cardiac morphogenesis.
Collapse
Affiliation(s)
- Clara Guijarro
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Solène Song
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Benoit Aigouy
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Raphaël Clément
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Paul Villoutreix
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France.
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France.
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
10
|
Henderson DJ, Alqahtani A, Chaudhry B, Cook A, Eley L, Houyel L, Hughes M, Keavney B, de la Pompa JL, Sled J, Spielmann N, Teboul L, Zaffran S, Mill P, Liu KJ. Beyond genomic studies of congenital heart defects through systematic modelling and phenotyping. Dis Model Mech 2024; 17:dmm050913. [PMID: 39575509 PMCID: PMC11603121 DOI: 10.1242/dmm.050913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/29/2024] [Indexed: 12/01/2024] Open
Abstract
Congenital heart defects (CHDs), the most common congenital anomalies, are considered to have a significant genetic component. However, despite considerable efforts to identify pathogenic genes in patients with CHDs, few gene variants have been proven as causal. The complexity of the genetic architecture underlying human CHDs likely contributes to this poor genetic discovery rate. However, several other factors are likely to contribute. For example, the level of patient phenotyping required for clinical care may be insufficient for research studies focused on mechanistic discovery. Although several hundred mouse gene knockouts have been described with CHDs, these are generally not phenotyped and described in the same way as CHDs in patients, and thus are not readily comparable. Moreover, most patients with CHDs carry variants of uncertain significance of crucial cardiac genes, further complicating comparisons between humans and mouse mutants. In spite of major advances in cardiac developmental biology over the past 25 years, these advances have not been well communicated to geneticists and cardiologists. As a consequence, the latest data from developmental biology are not always used in the design and interpretation of studies aimed at discovering the genetic causes of CHDs. In this Special Article, while considering other in vitro and in vivo models, we create a coherent framework for accurately modelling and phenotyping human CHDs in mice, thereby enhancing the translation of genetic and genomic studies into the causes of CHDs in patients.
Collapse
Affiliation(s)
- Deborah J. Henderson
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Ahlam Alqahtani
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Bill Chaudhry
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Andrew Cook
- University College London, Zayed Centre for Research, London WC1N 1DZ, UK
| | - Lorraine Eley
- Biosciences Institute, Newcastle University, Centre for Life, Newcastle upon Tyne NE1 3BZ, UK
| | - Lucile Houyel
- Congenital and Pediatric Cardiology Unit, M3C-Necker, Hôpital Universitaire Necker-Enfants Malades, APHP, Université Paris Cité, 149 Rue de Sèvres, 75015 Paris, France
| | - Marina Hughes
- Cardiology Department, Norfolk and Norwich University Hospital, Norwich NR4 7UY, UK
| | - Bernard Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9PT, UK
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - John Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto M5G 1XS, Canada. Department of Medical Biophysics, University of Toronto, Toronto M5G 1XS, Canada
| | - Nadine Spielmann
- Institute of Experimental Genetics, German Mouse Clinic, Helmholtz Center Munich (GmbH), German Research Center for Environmental Health, D-85764 Neuherberg, Germany
| | - Lydia Teboul
- Mary Lyon Centre, MRC Harwell, Oxfordshire OX11 0RD, UK
| | - Stephane Zaffran
- Aix Marseille Université, INSERM, Marseille Medical Genetics, U1251, 13005 Marseille, France
| | - Pleasantine Mill
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- MRC Human Genetics Unit, Institute for Genetics and Cancer, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Karen J. Liu
- MRC National Mouse Genetics Network, Congenital Anomalies Cluster, Harwell, OX11 0RD, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| |
Collapse
|
11
|
Feng H, Yang S, Zhang L, Zhu J, Li J, Yang Z. A new Prdm1-Cre line is suitable for studying the second heart field development. Dev Biol 2024; 514:78-86. [PMID: 38880275 DOI: 10.1016/j.ydbio.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024]
Abstract
The second heart field (SHF) plays a pivotal role in heart development, particularly in outflow tract (OFT) morphogenesis and septation, as well as in the expansion of the right ventricle (RV). Two mouse Cre lines, the Mef2c-AHF-Cre (Mef2c-Cre) and Isl1-Cre, have been widely used to study the SHF development. However, Cre activity is triggered not only in the SHF but also in the RV in the Mef2c-Cre mice, and in the Isl1-Cre mice, Cre activation is not SHF-specific. Therefore, a more suitable SHF-Cre line is desirable for better understanding SHF development. Here, we generated and characterized the Prdm1-Cre knock-in mice. In comparison with Mef2c-Cre mice, the Cre activity is similar in the pharyngeal and splanchnic mesoderm, and in the OFT of the Prdm1-Cre mice. Nonetheless, it was noticed that Cre expression is largely reduced in the RV of Prdm1-Cre mice compared to the Mef2c-Cre mice. Furthermore, we deleted Hand2, Nkx2-5, Pdk1 and Tbx20 using both Mef2c-Cre and Prdm1-Cre mice to study OFT morphogenesis and septation, making a comparison between these two Cre lines. New insights were obtained in understanding SHF development including differentiation into cardiomyocytes in the OFT using Prdm1-Cre mice. In conclusion, we found that Prdm1-Cre mouse line is a more appropriate tool to monitor SHF development, while the Mef2c-Cre mice are excellent in studying the role and function of the SHF in OFT morphogenesis and septation.
Collapse
Affiliation(s)
- Haiyue Feng
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Suming Yang
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijun Zhang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Jingai Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Jinsong Li
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
12
|
Moran HR, Nyarko OO, O’Rourke R, Ching RCK, Riemslagh FW, Peña B, Burger A, Sucharov CC, Mosimann C. The pericardium forms as a distinct structure during heart formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613484. [PMID: 39345600 PMCID: PMC11429720 DOI: 10.1101/2024.09.18.613484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The heart integrates diverse cell lineages into a functional unit, including the pericardium, a mesothelial sac that supports heart movement, homeostasis, and immune responses. However, despite its critical roles, the developmental origins of the pericardium remain uncertain due to disparate models. Here, using live imaging, lineage tracking, and single-cell transcriptomics in zebrafish, we find the pericardium forms within the lateral plate mesoderm from dedicated anterior mesothelial progenitors and distinct from the classic heart field. Imaging of transgenic reporters in zebrafish documents lateral plate mesoderm cells that emerge lateral of the classic heart field and among a continuous mesothelial progenitor field. Single-cell transcriptomics and trajectories of hand2-expressing lateral plate mesoderm reveal distinct populations of mesothelial and cardiac precursors, including pericardial precursors that are distinct from the cardiomyocyte lineage. The mesothelial gene expression signature is conserved in mammals and carries over to post-natal development. Light sheet-based live-imaging and machine learning-supported cell tracking documents that during heart tube formation, pericardial precursors that reside at the anterior edge of the heart field migrate anteriorly and medially before fusing, enclosing the embryonic heart to form a single pericardial cavity. Pericardium formation proceeds even upon genetic disruption of heart tube formation, uncoupling the two structures. Canonical Wnt/β-catenin signaling modulates pericardial cell number, resulting in a stretched pericardial epithelium with reduced cell number upon canonical Wnt inhibition. We connect the pathological expression of secreted Wnt antagonists of the SFRP family found in pediatric dilated cardiomyopathy to increased pericardial stiffness: sFRP1 in the presence of increased catecholamines causes cardiomyocyte stiffness in neonatal rats as measured by atomic force microscopy. Altogether, our data integrate pericardium formation as an independent process into heart morphogenesis and connect disrupted pericardial tissue properties such as pericardial stiffness to pediatric cardiomyopathies.
Collapse
Affiliation(s)
- Hannah R. Moran
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Obed O. Nyarko
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rebecca O’Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ryenne-Christine K. Ching
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Frederike W. Riemslagh
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Brisa Peña
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Cardiovascular Institute, Division of Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Bioengineering Department, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Alexa Burger
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
13
|
Piñeiro-Sabarís R, MacGrogan D, de la Pompa JL. Deficient GATA6-CXCR7 signaling leads to bicuspid aortic valve. Dis Model Mech 2024; 17:dmm050934. [PMID: 39253784 PMCID: PMC11413932 DOI: 10.1242/dmm.050934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/31/2024] [Indexed: 09/11/2024] Open
Abstract
The cardiac outflow tract (OFT) transiently links the ventricles to the aortic sac and forms the arterial valves. Abnormalities in these valves, such as bicuspid aortic valve (BAV), are common congenital anomalies. GATA6-inactivating variants cause cardiac OFT defects and BAV, but their mechanisms are unclear. We generated Gata6STOP/+ mice using CRISPR-Cas9, which show highly penetrant BAV (70%) and membranous ventricular septal defects (43%). These mice exhibited decreased proliferation and increased ISL1-positive progenitor cells in the OFT, indicating abnormal cardiovascular differentiation. Gata6 deletion with the Mef2cCre driver line recapitulated Gata6STOP/+ phenotypes, indicating a cell-autonomous role for Gata6 in the second heart field. Gata6STOP/+ mice showed reduced OFT length and caliber, associated with deficient cardiac neural crest cell contribution, which may cause valvulo-septal defects. RNA-sequencing analysis showed depletion in pathways related to cell proliferation and migration, highlighting Cxcr7 (also known as Ackr3) as a candidate gene. Reduced mesenchymal cell migration and invasion were observed in Gata6STOP/+ OFT tissue. CXCR7 agonists reduced mesenchymal cell migration and increased invasion in wild-type but not in Gata6STOP/+ explants, indicating the GATA6-dependent role of CXCR7 in OFT development and its potential link to BAV.
Collapse
Affiliation(s)
- Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
14
|
Dumas CE, Rousset C, De Bono C, Cortés C, Jullian E, Lescroart F, Zaffran S, Adachi N, Kelly RG. Retinoic acid signalling regulates branchiomeric neck muscle development at the head/trunk interface. Development 2024; 151:dev202905. [PMID: 39082789 DOI: 10.1242/dev.202905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/18/2024] [Indexed: 08/30/2024]
Abstract
Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.
Collapse
Affiliation(s)
- Camille E Dumas
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Célia Rousset
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Claudio Cortés
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Estelle Jullian
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | | | - Stéphane Zaffran
- Aix-Marseille Université, INSERM, MMG U1251, 13005 Marseille, France
| | - Noritaka Adachi
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, 13009 Marseille, France
| |
Collapse
|
15
|
Piñeiro-Sabarís R, MacGrogan D, de la Pompa JL. Intricate MIB1-NOTCH-GATA6 Interactions in Cardiac Valvular and Septal Development. J Cardiovasc Dev Dis 2024; 11:223. [PMID: 39057643 PMCID: PMC11277162 DOI: 10.3390/jcdd11070223] [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: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Genome-wide association studies and experimental mouse models implicate the MIB1 and GATA6 genes in congenital heart disease (CHD). Their close physical proximity and conserved synteny suggest that these two genes might be involved in analogous cardiac developmental processes. Heterozygous Gata6 loss-of-function mutations alone or humanized Mib1 mutations in a NOTCH1-sensitized genetic background cause bicuspid aortic valve (BAV) and a membranous ventricular septal defect (VSD), consistent with MIB1 and NOTCH1 functioning in the same pathway. To determine if MIB1-NOTCH and GATA6 interact in valvular and septal development, we generated compound heterozygote mice carrying different Mib1 missense (Mib1K735R and Mib1V943F) or nonsense (Mib1R530X) mutations with the Gata6STOP/+ heterozygous null mutation. Combining Mib1R530X/+ or Mib1K735R/+ with Gata6STOP/+ does not affect Gata6STOP/+ single mutant phenotypes. In contrast, combining Mib1V943F/+ with Gata6STOP/+ decreases the incidence of BAV and VSD by 50%, suggesting a suppressive effect of Mib1V943F/+ on Gata6STOP/+. Transcriptomic and functional analyses revealed that while the EMT pathway term is depleted in the Gata6STOP/+ mutant, introducing the Mib1V943F variant robustly enriches this term, consistent with the Mib1V943F/+ phenotypic suppression of Gata6STOP/+. Interestingly, combined Notch1 and Gata6 insufficiency led to a nearly fully penetrant VSD but did not affect the BAV phenotype, underscoring the complex functional relationship between MIB1, NOTCH, and GATA6 in valvular and septal development.
Collapse
Affiliation(s)
- Rebeca Piñeiro-Sabarís
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | - José Luis de la Pompa
- Intercellular Signaling in Cardiovascular Development & Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain;
- Ciber de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Melchor Fernández Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
16
|
En A, Bogireddi H, Thomas B, Stutzman AV, Ikegami S, LaForest B, Almakki O, Pytel P, Moskowitz IP, Ikegami K. Pervasive nuclear envelope ruptures precede ECM signaling and disease onset without activating cGAS-STING in Lamin-cardiomyopathy mice. Cell Rep 2024; 43:114284. [PMID: 38814785 PMCID: PMC11290591 DOI: 10.1016/j.celrep.2024.114284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 04/14/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we test a prevailing hypothesis that NE ruptures trigger the pathological cGAS-STING cytosolic DNA-sensing pathway using a mouse model of Lamin cardiomyopathy. The reduction of Lamin A/C in cardio-myocyte of adult mice causes pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures are followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remains inactive. Deleting cGas or Sting does not rescue cardiomyopathy in the mouse model. The lack of cGAS-STING activation is likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling is activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin cardiomyopathy.
Collapse
Affiliation(s)
- Atsuki En
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa 236-0027, Japan
| | - Hanumakumar Bogireddi
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Briana Thomas
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alexis V Stutzman
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Sachie Ikegami
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Brigitte LaForest
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Omar Almakki
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA
| | - Peter Pytel
- Department of Pathology, the University of Chicago, Chicago, IL 60637, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, the University of Chicago, Chicago, IL 60637, USA; Department of Pathology, the University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, the University of Chicago, Chicago, IL 60637, USA
| | - Kohta Ikegami
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
17
|
Lu J, Qian S, Sun Z. Targeting histone deacetylase in cardiac diseases. Front Physiol 2024; 15:1405569. [PMID: 38983721 PMCID: PMC11232433 DOI: 10.3389/fphys.2024.1405569] [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: 03/23/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Histone deacetylases (HDAC) catalyze the removal of acetylation modifications on histones and non-histone proteins, which regulates gene expression and other cellular processes. HDAC inhibitors (HDACi), approved anti-cancer agents, emerge as a potential new therapy for heart diseases. Cardioprotective effects of HDACi are observed in many preclinical animal models of heart diseases. Genetic mouse models have been developed to understand the role of each HDAC in cardiac functions. Some of the findings are controversial. Here, we provide an overview of how HDACi and HDAC impact cardiac functions under physiological or pathological conditions. We focus on in vivo studies of zinc-dependent classical HDACs, emphasizing disease conditions involving cardiac hypertrophy, myocardial infarction (MI), ischemic reperfusion (I/R) injury, and heart failure. In particular, we review how non-biased omics studies can help our understanding of the mechanisms underlying the cardiac effects of HDACi and HDAC.
Collapse
Affiliation(s)
- Jiao Lu
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, United States
| | - Sichong Qian
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, United States
| | - Zheng Sun
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine, Houston, TX, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
18
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Buffering Mechanism in Aortic Arch Artery Formation and Congenital Heart Disease. Circ Res 2024; 134:e112-e132. [PMID: 38618720 PMCID: PMC11081845 DOI: 10.1161/circresaha.123.322767] [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: 03/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease arising from defective morphogenesis of pharyngeal arch arteries (PAAs) and their derivatives. METHODS Mouse genetics, lineage tracing, confocal microscopy, and quantitative image analyses were used to investigate mechanisms of PAA formation and repair. RESULTS The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived endothelial cells (ECs) is regulated by VEGFR2 (vascular endothelial growth factor receptor 2) and Tbx1. Remarkably, when the SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated 3-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of 1 VEGFR2 allele (VEGFR2SHF-HET) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2SHF-KO) abolishes it. The decrease in SHF-derived ECs in VEGFR2SHF-HET and VEGFR2SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2SHF-KO mutants. Blocking the compensatory response in VEGFR2SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1+/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and congenital heart disease. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. CONCLUSIONS Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling.
Collapse
Affiliation(s)
- AnnJosette Ramirez
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Christina A. Vyzas
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Kevin Eng
- Department of Statistics, Rutgers University, School of Arts and Sciences, Piscataway, NJ 08854
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
19
|
En A, Bogireddi H, Thomas B, Stutzman A, Ikegami S, LaForest B, Almakki O, Pytel P, Moskowitz IP, Ikegami K. Pervasive nuclear envelope ruptures precede ECM signaling and disease onset without activating cGAS-STING in Lamin-cardiomyopathy mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.28.555134. [PMID: 37693381 PMCID: PMC10491116 DOI: 10.1101/2023.08.28.555134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Nuclear envelope (NE) ruptures are emerging observations in Lamin-related dilated cardiomyopathy, an adult-onset disease caused by loss-of-function mutations in Lamin A/C, a nuclear lamina component. Here, we tested a prevailing hypothesis that NE ruptures trigger pathological cGAS-STING cytosolic DNA-sensing pathway, using a mouse model of Lamin-cardiomyopathy. Reduction of Lamin A/C in cardiomyocytes of adult mice caused pervasive NE ruptures in cardiomyocytes, preceding inflammatory transcription, fibrosis, and fatal dilated cardiomyopathy. NE ruptures were followed by DNA damage accumulation without causing immediate cardiomyocyte death. However, cGAS-STING-dependent inflammatory signaling remained inactive. Deleting cGas or Sting did not rescue cardiomyopathy. The lack of cGAS-STING activation was likely due to the near absence of cGAS expression in adult cardiomyocytes at baseline. Instead, extracellular matrix (ECM) signaling was activated and predicted to initiate pro-inflammatory communication from Lamin-reduced cardiomyocytes to fibroblasts. Our work nominates ECM signaling, not cGAS-STING, as a potential inflammatory contributor in Lamin-cardiomyopathy.
Collapse
Affiliation(s)
- Atsuki En
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Hanumakumar Bogireddi
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Briana Thomas
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Alexis Stutzman
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Sachie Ikegami
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Brigitte LaForest
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Omar Almakki
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Peter Pytel
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, The University of Chicago, Chicago, IL, USA
- Department of Pathology, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Kohta Ikegami
- Division of Molecular Cardvascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
20
|
Guijarro C, Kelly RG. On the involvement of the second heart field in congenital heart defects. C R Biol 2024; 347:9-18. [PMID: 38488639 DOI: 10.5802/crbiol.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 03/19/2024]
Abstract
Congenital heart defects (CHD) affect 1 in 100 live births and result from defects in cardiac development. Growth of the early heart tube occurs by the progressive addition of second heart field (SHF) progenitor cells to the cardiac poles. The SHF gives rise to ventricular septal, right ventricular and outflow tract myocardium at the arterial pole, and atrial, including atrial septal myocardium, at the venous pole. SHF deployment creates the template for subsequent cardiac septation and has been implicated in cardiac looping and in orchestrating outflow tract development with neural crest cells. Genetic or environmental perturbation of SHF deployment thus underlies a spectrum of common forms of CHD affecting conotruncal and septal morphogenesis. Here we review the major properties of SHF cells as well as recent insights into the developmental programs that drive normal cardiac progenitor cell addition and the origins of CHD.
Collapse
|
21
|
Kathiriya IS, Dominguez MH, Rao KS, Muncie-Vasic JM, Devine WP, Hu KM, Hota SK, Garay BI, Quintero D, Goyal P, Matthews MN, Thomas R, Sukonnik T, Miguel-Perez D, Winchester S, Brower EF, Forjaz A, Wu PH, Wirtz D, Kiemen AL, Bruneau BG. A disrupted compartment boundary underlies abnormal cardiac patterning and congenital heart defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.05.578995. [PMID: 38370632 PMCID: PMC10871243 DOI: 10.1101/2024.02.05.578995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Failure of septation of the interventricular septum (IVS) is the most common congenital heart defect (CHD), but mechanisms for patterning the IVS are largely unknown. We show that a Tbx5+/Mef2cAHF+ progenitor lineage forms a compartment boundary bisecting the IVS. This coordinated population originates at a first- and second heart field interface, subsequently forming a morphogenetic nexus. Ablation of Tbx5+/Mef2cAHF+ progenitors cause IVS disorganization, right ventricular hypoplasia and mixing of IVS lineages. Reduced dosage of the CHD transcription factor TBX5 disrupts boundary position and integrity, resulting in ventricular septation defects (VSDs) and patterning defects, including Slit2 and Ntn1 misexpression. Reducing NTN1 dosage partly rescues cardiac defects in Tbx5 mutant embryos. Loss of Slit2 or Ntn1 causes VSDs and perturbed septal lineage distributions. Thus, we identify essential cues that direct progenitors to pattern a compartment boundary for proper cardiac septation, revealing new mechanisms for cardiac birth defects.
Collapse
Affiliation(s)
- Irfan S Kathiriya
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
| | - Martin H Dominguez
- Gladstone Institutes, San Francisco, CA
- Department of Medicine, University of California, San Francisco, San Francisco, CA
- Current address: Department of Medicine (Cardiovascular Medicine), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kavitha S Rao
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
| | | | - W Patrick Devine
- Gladstone Institutes, San Francisco, CA
- Current address: Department of Pathology, University of California, San Francisco, San Francisco, CA
| | - Kevin M Hu
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
- Current address: Creighton University School of Medicine, Omaha, NE
| | - Swetansu K Hota
- Gladstone Institutes, San Francisco, CA
- Current address: Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
| | - Bayardo I Garay
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Current address: University of Minnesota Medical Scientist Training Program, Minneapolis, MN
| | - Diego Quintero
- Gladstone Institutes, San Francisco, CA
- Current address: Department of Human Genetics, Emory University School of Medicine, Atlanta, GA
| | - Piyush Goyal
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
- Gladstone Institutes, San Francisco, CA
- Current address: Touro University California, Vallejo, CA
| | - Megan N Matthews
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA
| | | | | | | | | | | | - André Forjaz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Ashley L Kiemen
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA
- Roddenberry Center for Stem Cell Biology and Medicine, Gladstone Institutes, San Francisco, CA
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
22
|
Padmanabhan A, de Soysa TY, Pelonero A, Sapp V, Shah PP, Wang Q, Li L, Lee CY, Sadagopan N, Nishino T, Ye L, Yang R, Karnay A, Poleshko A, Bolar N, Linares-Saldana R, Ranade SS, Alexanian M, Morton SU, Jain M, Haldar SM, Srivastava D, Jain R. A genome-wide CRISPR screen identifies BRD4 as a regulator of cardiomyocyte differentiation. NATURE CARDIOVASCULAR RESEARCH 2024; 3:317-331. [PMID: 39196112 PMCID: PMC11361716 DOI: 10.1038/s44161-024-00431-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/19/2024] [Indexed: 08/29/2024]
Abstract
Human induced pluripotent stem cell (hiPSC) to cardiomyocyte (CM) differentiation has reshaped approaches to studying cardiac development and disease. In this study, we employed a genome-wide CRISPR screen in a hiPSC to CM differentiation system and reveal here that BRD4, a member of the bromodomain and extraterminal (BET) family, regulates CM differentiation. Chemical inhibition of BET proteins in mouse embryonic stem cell (mESC)-derived or hiPSC-derived cardiac progenitor cells (CPCs) results in decreased CM differentiation and persistence of cells expressing progenitor markers. In vivo, BRD4 deletion in second heart field (SHF) CPCs results in embryonic or early postnatal lethality, with mutants demonstrating myocardial hypoplasia and an increase in CPCs. Single-cell transcriptomics identified a subpopulation of SHF CPCs that is sensitive to BRD4 loss and associated with attenuated CM lineage-specific gene programs. These results highlight a previously unrecognized role for BRD4 in CM fate determination during development and a heterogenous requirement for BRD4 among SHF CPCs.
Collapse
Affiliation(s)
- Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | | | | | - Valerie Sapp
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, CA, USA
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Parisha P Shah
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Qiaohong Wang
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Li
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Clara Youngna Lee
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Nandhini Sadagopan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | | | - Lin Ye
- Gladstone Institutes, San Francisco, CA, USA
| | - Rachel Yang
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashley Karnay
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrey Poleshko
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Nikhita Bolar
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Ricardo Linares-Saldana
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Michael Alexanian
- Gladstone Institutes, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
| | - Sarah U Morton
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mohit Jain
- Department of Medicine, University of California, San Diego, School of Medicine, San Diego, CA, USA
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Saptarsi M Haldar
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, School of Medicine, San Francisco, CA, USA
- Amgen Research, Cardiometabolic Disorders, South San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institutes, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, School of Medicine, San Francisco, CA, USA.
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone Institutes, San Francisco, CA, USA.
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA.
| | - Rajan Jain
- Cardiovascular Institute, Epigenetics Institute, and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
Lin Y, Yang Q, Lin X, Liu X, Qian Y, Xu D, Cao N, Han X, Zhu Y, Hu W, He X, Yu Z, Kong X, Zhu L, Zhong Z, Liu K, Zhou B, Wang Y, Peng J, Zhu W, Wang J. Extracellular Matrix Disorganization Caused by ADAMTS16 Deficiency Leads to Bicuspid Aortic Valve With Raphe Formation. Circulation 2024; 149:605-626. [PMID: 38018454 DOI: 10.1161/circulationaha.123.065458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND A better understanding of the molecular mechanism of aortic valve development and bicuspid aortic valve (BAV) formation would significantly improve and optimize the therapeutic strategy for BAV treatment. Over the past decade, the genes involved in aortic valve development and BAV formation have been increasingly recognized. On the other hand, ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) gene family members have been reported to be able to modulate cardiovascular development and diseases. The present study aimed to further investigate the roles of ADAMTS family members in aortic valve development and BAV formation. METHODS Morpholino-based ADAMTS family gene-targeted screening for zebrafish heart outflow tract phenotypes combined with DNA sequencing in a 304 cohort BAV patient registry study was initially carried out to identify potentially related genes. Both ADAMTS gene-specific fluorescence in situ hybridization assay and genetic tracing experiments were performed to evaluate the expression pattern in the aortic valve. Accordingly, related genetic mouse models (both knockout and knockin) were generated using the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9) method to further study the roles of ADAMTS family genes. The lineage-tracing technique was used again to evaluate how the cellular activity of specific progenitor cells was regulated by ADAMTS genes. Bulk RNA sequencing was used to investigate the signaling pathways involved. Inducible pluripotent stem cells derived from both BAV patients and genetic mouse tissue were used to study the molecular mechanism of ADAMTS. Immunohistochemistry was performed to examine the phenotype of cardiac valve anomalies, especially in the extracellular matrix components. RESULTS ADAMTS genes targeting and phenotype screening in zebrafish and targeted DNA sequencing on a cohort of patients with BAV identified ADAMTS16 (a disintegrin and metalloproteinase with thrombospondin motifs 16) as a BAV-causing gene and found the ADAMTS16 p. H357Q variant in an inherited BAV family. Both in situ hybridization and genetic tracing studies described a unique spatiotemporal pattern of ADAMTS16 expression during aortic valve development. Adamts16+/- and Adamts16+/H355Q mouse models both exhibited a right coronary cusp-noncoronary cusp fusion-type BAV phenotype, with progressive aortic valve thickening associated with raphe formation (fusion of the commissure). Further, ADAMTS16 deficiency in Tie2 lineage cells recapitulated the BAV phenotype. This was confirmed in lineage-tracing mouse models in which Adamts16 deficiency affected endothelial and second heart field cells, not the neural crest cells. Accordingly, the changes were mainly detected in the noncoronary and right coronary leaflets. Bulk RNA sequencing using inducible pluripotent stem cells-derived endothelial cells and genetic mouse embryonic heart tissue unveiled enhanced FAK (focal adhesion kinase) signaling, which was accompanied by elevated fibronectin levels. Both in vitro inducible pluripotent stem cells-derived endothelial cells culture and ex vivo embryonic outflow tract explant studies validated the altered FAK signaling. CONCLUSIONS Our present study identified a novel BAV-causing ADAMTS16 p. H357Q variant. ADAMTS16 deficiency led to BAV formation.
Collapse
Affiliation(s)
- Ying Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Qifan Yang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaoping Lin
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xianbao Liu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Yi Qian
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Dilin Xu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Naifang Cao
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, China (X.H.)
| | - Yanqing Zhu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wangxing Hu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiaopeng He
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhengyang Yu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Xiangmin Kong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Lianlian Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Zhiwei Zhong
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Kai Liu
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Bin Zhou
- New Cornerstone Investigator Institute, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences (B.Z.)
| | - Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University Health Science Center, China (Y.W.)
| | - Jinrong Peng
- Ministry of Education Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network (Y.Z., K.L., J.P.), Hangzhou, China
| | - Wei Zhu
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| | - Jian'an Wang
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University School of Medicine (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.), Hangzhou, China
- Research Center for Life Science and Human Health, Binjiang Institute (J.W.), Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, China (Y.L., Q.Y., X. Lin, X. Liu, Y.Q., D.X., N.C., W.H., X.H., Z.Y., X.K., L.Z., Z.Z., W.Z., J.W.)
| |
Collapse
|
24
|
Ramirez A, Vyzas CA, Zhao H, Eng K, Degenhardt K, Astrof S. Identification of novel buffering mechanisms in aortic arch artery development and congenital heart disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.02.530833. [PMID: 38370627 PMCID: PMC10871175 DOI: 10.1101/2023.03.02.530833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Rationale The resiliency of embryonic development to genetic and environmental perturbations has been long appreciated; however, little is known about the mechanisms underlying the robustness of developmental processes. Aberrations resulting in neonatal lethality are exemplified by congenital heart disease (CHD) arising from defective morphogenesis of pharyngeal arch arteries (PAA) and their derivatives. Objective To uncover mechanisms underlying the robustness of PAA morphogenesis. Methods and Results The second heart field (SHF) gives rise to the PAA endothelium. Here, we show that the number of SHF-derived ECs is regulated by VEGFR2 and Tbx1 . Remarkably, when SHF-derived EC number is decreased, PAA development can be rescued by the compensatory endothelium. Blocking such compensatory response leads to embryonic demise. To determine the source of compensating ECs and mechanisms regulating their recruitment, we investigated three-dimensional EC connectivity, EC fate, and gene expression. Our studies demonstrate that the expression of VEGFR2 by the SHF is required for the differentiation of SHF-derived cells into PAA ECs. The deletion of one VEGFR2 allele (VEGFR2 SHF-HET ) reduces SHF contribution to the PAA endothelium, while the deletion of both alleles (VEGFR2 SHF-KO ) abolishes it. The decrease in SHF-derived ECs in VEGFR2 SHF-HET and VEGFR2 SHF-KO embryos is complemented by the recruitment of ECs from the nearby veins. Compensatory ECs contribute to PAA derivatives, giving rise to the endothelium of the aortic arch and the ductus in VEGFR2 SHF-KO mutants. Blocking the compensatory response in VEGFR2 SHF-KO mutants results in embryonic lethality shortly after mid-gestation. The compensatory ECs are absent in Tbx1 +/- embryos, a model for 22q11 deletion syndrome, leading to unpredictable arch artery morphogenesis and CHD. Tbx1 regulates the recruitment of the compensatory endothelium in an SHF-non-cell-autonomous manner. Conclusions Our studies uncover a novel buffering mechanism underlying the resiliency of PAA development and remodeling. Nonstandard Abbreviations and Acronyms in Alphabetical Order CHD - congenital heart disease; ECs - endothelial cells; IAA-B - interrupted aortic arch type B; PAA - pharyngeal arch arteries; RERSA - retro-esophageal right subclavian artery; SHF - second heart field; VEGFR2 - Vascular endothelial growth factor receptor 2.
Collapse
|
25
|
Racedo SE, Liu Y, Shi L, Zheng D, Morrow BE. Dgcr8 functions in the secondary heart field for outflow tract and right ventricle development in mammals. Dev Biol 2024; 506:72-84. [PMID: 38110169 PMCID: PMC10793380 DOI: 10.1016/j.ydbio.2023.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/28/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
The DGCR8 gene, encoding a critical miRNA processing protein, maps within the hemizygous region in patients with 22q11.2 deletion syndrome. Most patients have malformations of the cardiac outflow tract that is derived in part from the anterior second heart field (aSHF) mesoderm. To understand the function of Dgcr8 in the aSHF, we inactivated it in mice using Mef2c-AHF-Cre. Inactivation resulted in a fully penetrant persistent truncus arteriosus and a hypoplastic right ventricle leading to lethality by E14.5. To understand the molecular mechanism for this phenotype, we performed gene expression profiling of the aSHF and the cardiac outflow tract with right ventricle in conditional null versus normal mouse littermates at stage E9.5 prior to morphology changes. We identified dysregulation of mRNA gene expression, of which some are relevant to cardiogenesis. Many pri-miRNA genes were strongly increased in expression in mutant embryos along with reduced expression of mature miRNA genes. We further examined the individual, mature miRNAs that were decreased in expression along with pri-miRNAs that were accumulated that could be direct effects due to loss of Dgcr8. Among these genes, were miR-1a, miR-133a, miR-134, miR143 and miR145a, which have known functions in heart development. These early mRNA and miRNA changes may in part, explain the first steps that lead to the resulting phenotype in Dgcr8 aSHF conditional mutant embryos.
Collapse
Affiliation(s)
- Silvia E Racedo
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yang Liu
- Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Bell Buckle, TN, USA
| | - Lijie Shi
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA; Departments of Pediatrics and Ob/Gyn & Population Health, USA.
| |
Collapse
|
26
|
Alexander BE, Zhao H, Astrof S. SMAD4: A critical regulator of cardiac neural crest cell fate and vascular smooth muscle development. Dev Dyn 2024; 253:119-143. [PMID: 37650555 PMCID: PMC10842824 DOI: 10.1002/dvdy.652] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/07/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND During embryogenesis, cardiac neural crest-derived cells (NCs) migrate into the pharyngeal arches and give rise to the vascular smooth muscle cells (vSMCs) of the pharyngeal arch arteries (PAAs). vSMCs are critical for the remodeling of the PAAs into their final adult configuration, giving rise to the aortic arch and its arteries (AAAs). RESULTS We investigated the role of SMAD4 in NC-to-vSMC differentiation using lineage-specific inducible mouse strains. We found that the expression of SMAD4 in the NC is indelible for regulating the survival of cardiac NCs. Although the ablation of SMAD4 at E9.5 in the NC lineage led to a near-complete absence of NCs in the pharyngeal arches, PAAs became invested with vSMCs derived from a compensatory source. Analysis of AAA development at E16.5 showed that the alternative vSMC source compensated for the lack of NC-derived vSMCs and rescued AAA morphogenesis. CONCLUSIONS Our studies uncovered the requisite role of SMAD4 in the contribution of the NC to the pharyngeal arch mesenchyme. We found that in the absence of SMAD4+ NCs, vSMCs around the PAAs arose from a different progenitor source, rescuing AAA morphogenesis. These findings shed light on the remarkable plasticity of developmental mechanisms governing AAA development.
Collapse
Affiliation(s)
- Brianna E. Alexander
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Molecular Biology, Genetics, and Cancer Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
27
|
Grunert M, Dorn C, Rickert-Sperling S. Cardiac Transcription Factors and Regulatory Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:295-311. [PMID: 38884718 DOI: 10.1007/978-3-031-44087-8_16] [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
Cardiac development is a fine-tuned process governed by complex transcriptional networks, in which transcription factors (TFs) interact with other regulatory layers. In this chapter, we introduce the core cardiac TFs including Gata, Hand, Nkx2, Mef2, Srf, and Tbx. These factors regulate each other's expression and can also act in a combinatorial manner on their downstream targets. Their disruption leads to various cardiac phenotypes in mice, and mutations in humans have been associated with congenital heart defects. In the second part of the chapter, we discuss different levels of regulation including cis-regulatory elements, chromatin structure, and microRNAs, which can interact with transcription factors, modulate their function, or are downstream targets. Finally, examples of disturbances of the cardiac regulatory network leading to congenital heart diseases in human are provided.
Collapse
Affiliation(s)
- Marcel Grunert
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cornelia Dorn
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
28
|
Eley L, Richardson RV, Alqahtani A, Chaudhry B, Henderson DJ. eNOS plays essential roles in the developing heart and aorta linked to disruption of Notch signalling. Dis Model Mech 2024; 17:dmm050265. [PMID: 38111957 PMCID: PMC10846539 DOI: 10.1242/dmm.050265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 12/12/2023] [Indexed: 12/20/2023] Open
Abstract
eNOS (NOS3) is the enzyme that generates nitric oxide, a signalling molecule and regulator of vascular tone. Loss of eNOS function is associated with increased susceptibility to atherosclerosis, hypertension, thrombosis and stroke. Aortopathy and cardiac hypertrophy have also been found in eNOS null mice, but their aetiology is unclear. We evaluated eNOS nulls before and around birth for cardiac defects, revealing severe abnormalities in the ventricular myocardium and pharyngeal arch arteries. Moreover, in the aortic arch, there were fewer baroreceptors, which sense changes in blood pressure. Adult eNOS null survivors showed evidence of cardiac hypertrophy, aortopathy and cartilaginous metaplasia in the periductal region of the aortic arch. Notch1 and neuregulin were dysregulated in the forming pharyngeal arch arteries and ventricles, suggesting that these pathways may be relevant to the defects observed. Dysregulation of eNOS leads to embryonic and perinatal death, suggesting mutations in eNOS are candidates for causing congenital heart defects in humans. Surviving eNOS mutants have a deficiency of baroreceptors that likely contributes to high blood pressure and may have relevance to human patients who suffer from hypertension associated with aortic arch abnormalities.
Collapse
Affiliation(s)
- Lorraine Eley
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Rachel V. Richardson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Ahlam Alqahtani
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Bill Chaudhry
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - Deborah J. Henderson
- Bioscience Institute, Newcastle University, Centre for Life, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| |
Collapse
|
29
|
Buckingham M, Kelly RG. Cardiac Progenitor Cells of the First and Second Heart Fields. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:103-124. [PMID: 38884707 DOI: 10.1007/978-3-031-44087-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
The heart forms from the first and second heart fields, which contribute to distinct regions of the myocardium. This is supported by clonal analyses, which identify corresponding first and second cardiac cell lineages in the heart. Progenitor cells of the second heart field and its sub-domains are controlled by a gene regulatory network and signaling pathways, which determine their behavior. Multipotent cells in this field can also contribute cardiac endothelial and smooth muscle cells. Furthermore, the skeletal muscles of the head and neck are clonally related to myocardial cells that form the arterial and venous poles of the heart. These lineage relationships, together with the genes that regulate the heart fields, have major implications for congenital heart disease.
Collapse
Affiliation(s)
- Margaret Buckingham
- Department of Developmental and Stem Cell Biology, CNRS UMR 3738, Institut Pasteur, Paris, France.
| | - Robert G Kelly
- Aix Marseille Université, Institut de Biologie du Dévelopment de Marseille, Marseille, France.
| |
Collapse
|
30
|
Wessels A. Inflow Tract Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:145-153. [PMID: 38884709 DOI: 10.1007/978-3-031-44087-8_7] [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
The development of the inflow tract is undoubtedly one of the most complex remodeling events in the formation of the four-chambered heart. It involves the creation of two separate atrial chambers, the formation of an atrial/atrioventricular (AV) septal complex, the incorporation of the caval veins and coronary sinus into the right atrium, and the remodeling events that result in pulmonary venous return draining into the left atrium. In these processes, the atrioventricular mesenchymal complex, consisting of the major atrioventricular (AV) cushions, the mesenchymal cap on the primary atrial septum (pAS), and the dorsal mesenchymal protrusion (DMP), plays a crucial role.
Collapse
Affiliation(s)
- Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
31
|
Coppiello G, Barlabé P, Moya-Jódar M, Abizanda G, Pogontke C, Barreda C, Iglesias E, Linares J, Arellano-Viera E, Larequi E, San Martín-Úriz P, Carvajal-Vergara X, Pelacho B, Mazo MM, Pérez-Pomares JM, Ruiz-Villalba A, Ullate-Agote A, Prósper F, Aranguren XL. Generation of heart and vascular system in rodents by blastocyst complementation. Dev Cell 2023; 58:2881-2895.e7. [PMID: 37967560 DOI: 10.1016/j.devcel.2023.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/10/2023] [Accepted: 10/23/2023] [Indexed: 11/17/2023]
Abstract
Generating organs from stem cells through blastocyst complementation is a promising approach to meet the clinical need for transplants. In order to generate rejection-free organs, complementation of both parenchymal and vascular cells must be achieved, as endothelial cells play a key role in graft rejection. Here, we used a lineage-specific cell ablation system to produce mouse embryos unable to form both the cardiac and vascular systems. By mouse intraspecies blastocyst complementation, we rescued heart and vascular system development separately and in combination, obtaining complemented hearts with cardiomyocytes and endothelial cells of exogenous origin. Complemented chimeras were viable and reached adult stage, showing normal cardiac function and no signs of histopathological defects in the heart. Furthermore, we implemented the cell ablation system for rat-to-mouse blastocyst complementation, obtaining xenogeneic hearts whose cardiomyocytes were completely of rat origin. These results represent an advance in the experimentation towards the in vivo generation of transplantable organs.
Collapse
Affiliation(s)
- Giulia Coppiello
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain.
| | - Paula Barlabé
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Marta Moya-Jódar
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Gloria Abizanda
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain; Cell Therapy Area, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - Cristina Pogontke
- Department of Animal Biology, University of Málaga, Málaga 29010, Spain; Biomedical Research Institute of Málaga (IBIMA-Plataforma BIONAND), Málaga 29590, Spain
| | - Carolina Barreda
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Elena Iglesias
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Javier Linares
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, 01307 Dresden, Germany
| | | | - Eduardo Larequi
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Patxi San Martín-Úriz
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Xonia Carvajal-Vergara
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Beatriz Pelacho
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Manuel Maria Mazo
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain; Cell Therapy Area, Clínica Universidad de Navarra, Pamplona 31008, Spain
| | - José Maria Pérez-Pomares
- Department of Animal Biology, University of Málaga, Málaga 29010, Spain; Biomedical Research Institute of Málaga (IBIMA-Plataforma BIONAND), Málaga 29590, Spain
| | - Adrián Ruiz-Villalba
- Department of Animal Biology, University of Málaga, Málaga 29010, Spain; Biomedical Research Institute of Málaga (IBIMA-Plataforma BIONAND), Málaga 29590, Spain
| | - Asier Ullate-Agote
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain
| | - Felipe Prósper
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain; Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona 31008, Spain; Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid 28029, Spain; Red Española de Terapias Avanzadas (RICORS-TERAV), Madrid 28029, Spain
| | - Xabier L Aranguren
- Program of Regenerative Medicine, Centre for Applied Medical Research (CIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31008, Spain.
| |
Collapse
|
32
|
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
|
33
|
Nishino T, Ranade SS, Pelonero A, van Soldt BJ, Ye L, Alexanian M, Koback F, Huang Y, Sadagopan N, Lam A, Zholudeva LV, Li F, Padmanabhan A, Thomas R, van Bemmel JG, Gifford CA, Costa MW, Srivastava D. Single Cell Multimodal Analyses Reveal Epigenomic and Transcriptomic Basis for Birth Defects in Maternal Diabetes. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1190-1203. [PMID: 39183978 PMCID: PMC11343316 DOI: 10.1038/s44161-023-00367-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/19/2023] [Indexed: 08/27/2024]
Abstract
Maternal diabetes mellitus is among the most frequent environmental contributors to congenital birth defects, including heart defects and craniofacial anomalies, yet the cell types affected and mechanisms of disruption are largely unknown. Using multi-modal single cell analyses, here we show that maternal diabetes affects the epigenomic landscape of specific subsets of cardiac and craniofacial progenitors during embryogenesis. A previously unrecognized cardiac progenitor subpopulation expressing the homeodomain-containing protein ALX3 showed prominent chromatin accessibility changes and acquired a more posterior identity. Similarly, a subpopulation of neural crest-derived cells in the second pharyngeal arch, which contributes to craniofacial structures, displayed abnormalities in the epigenetic landscape and axial patterning defects. Chromatin accessibility changes in both populations were associated with increased retinoic acid signaling, known to establish anterior-posterior identity. This work highlights how an environmental insult can have highly selective epigenomic consequences on discrete cell types leading to developmental patterning defects.
Collapse
Affiliation(s)
- Tomohiro Nishino
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Sanjeev S. Ranade
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Angelo Pelonero
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Benjamin J. van Soldt
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Lin Ye
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Michael Alexanian
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Frances Koback
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Yu Huang
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Nandhini Sadagopan
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Adrienne Lam
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Lyandysha V. Zholudeva
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Feiya Li
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Arun Padmanabhan
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | | | - Joke G. van Bemmel
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Casey A. Gifford
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Mauro W. Costa
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institutes; San Francisco, CA, USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone; San Francisco, CA, USA
- Division of Cardiology, Department of Pediatrics, University of California, San Francisco; San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco; San Francisco, CA, USA
| |
Collapse
|
34
|
Francis RJB, San Agustin JT, Szabo Rogers HL, Cui C, Jonassen JA, Eguether T, Follit JA, Lo CW, Pazour GJ. Autonomous and non-cell autonomous role of cilia in structural birth defects in mice. PLoS Biol 2023; 21:e3002425. [PMID: 38079449 PMCID: PMC10735189 DOI: 10.1371/journal.pbio.3002425] [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: 06/26/2023] [Revised: 12/21/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023] Open
Abstract
Ciliopathies are associated with wide spectrum of structural birth defects (SBDs), indicating important roles for cilia in development. Here, we provide novel insights into the temporospatial requirement for cilia in SBDs arising from deficiency in Ift140, an intraflagellar transport (IFT) protein regulating ciliogenesis. Ift140-deficient mice exhibit cilia defects accompanied by wide spectrum of SBDs including macrostomia (craniofacial defects), exencephaly, body wall defects, tracheoesophageal fistula (TEF), randomized heart looping, congenital heart defects (CHDs), lung hypoplasia, renal anomalies, and polydactyly. Tamoxifen inducible CAGGCre-ER deletion of a floxed Ift140 allele between E5.5 to 9.5 revealed early requirement for Ift140 in left-right heart looping regulation, mid to late requirement for cardiac outflow septation and alignment, and late requirement for craniofacial development and body wall closure. Surprisingly, CHD were not observed with 4 Cre drivers targeting different lineages essential for heart development, but craniofacial defects and omphalocele were observed with Wnt1-Cre targeting neural crest and Tbx18-Cre targeting epicardial lineage and rostral sclerotome through which trunk neural crest cells migrate. These findings revealed cell autonomous role of cilia in cranial/trunk neural crest-mediated craniofacial and body wall closure defects, while non-cell autonomous multi-lineage interactions underlie CHD pathogenesis, revealing unexpected developmental complexity for CHD associated with ciliopathies.
Collapse
Affiliation(s)
- Richard J. B. Francis
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
- Discipline of Biomedical Sciences and Molecular Biology; College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, Australia
| | - Jovenal T. San Agustin
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Heather L. Szabo Rogers
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Cheng Cui
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Julie A. Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Thibaut Eguether
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - John A. Follit
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Cecilia W. Lo
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
35
|
Edwards W, Bussey OK, Conlon FL. The Tbx20-TLE interaction is essential for the maintenance of the second heart field. Development 2023; 150:dev201677. [PMID: 37756602 PMCID: PMC10629681 DOI: 10.1242/dev.201677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
T-box transcription factor 20 (Tbx20) plays a multifaceted role in cardiac morphogenesis and controls a broad gene regulatory network. However, the mechanism by which Tbx20 activates and represses target genes in a tissue-specific and temporal manner remains unclear. Studies show that Tbx20 directly interacts with the Transducin-like Enhancer of Split (TLE) family of proteins to mediate transcriptional repression. However, a function for the Tbx20-TLE transcriptional repression complex during heart development has yet to be established. We created a mouse model with a two amino acid substitution in the Tbx20 EH1 domain, thereby disrupting the Tbx20-TLE interaction. Disruption of this interaction impaired crucial morphogenic events, including cardiac looping and chamber formation. Transcriptional profiling of Tbx20EH1Mut hearts and analysis of putative direct targets revealed misexpression of the retinoic acid pathway and cardiac progenitor genes. Further, we show that altered cardiac progenitor development and function contribute to the severe cardiac defects in our model. Our studies indicate that TLE-mediated repression is a primary mechanism by which Tbx20 controls gene expression.
Collapse
Affiliation(s)
- Whitney Edwards
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Olivia K. Bussey
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Frank L. Conlon
- Department of Biology and Genetics, McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Integrative Program for Biological & Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
36
|
Luo H, Yang Z, Li J, Jin H, Jiang M, Shan C. Deletion of PDK 1 Caused Cardiac Malmorphogenesis and Heart Defects Due to Profound Protein Phosphorylation Changes Mediated by SHP 2. J Cardiovasc Transl Res 2023; 16:1220-1231. [PMID: 36988860 DOI: 10.1007/s12265-023-10380-y] [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: 02/17/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023]
Abstract
Phosphoinositide-dependent protein kinase-1 (PDK1), a master kinase and involved in multiple signaling transduction, participates in regulating embryonic cardiac development and postnatal cardiac remodeling. Germline PDK1 knockout mice displayed no heart development; in this article, we deleted PDK1 in heart tissue with different cre to characterize the temporospatial features and find the relevance with congenital heart disease(CHD), furthermore to investigate the underlying mechanism. Knocking out PDK1 with Nkx2.5-cre, the heart showed prominent pulmonic stenosis. Ablated PDK1 with Mef2cSHF-cre, the second heart field (SHF) exhibited severe hypoplasia. And deleted PDK1 with αMHC-cre, the mice displayed dilated heart disease, protein analysis indicated PI3K and ERK were activated; meanwhile, PDK1-AKT-GSK3, and S6K-S6 were disrupted; phosphorylation level of Akt473, S6k421/424, and Gsk3α21 enhanced; however, Akt308, S6k389, and Gsk3β9 decreased. In mechanism investigation, we found SHP2 membrane localization and phosphorylation level of SHP2542 elevated, which suggested SHP2 likely mediated the disruption.
Collapse
Affiliation(s)
- Hongmei Luo
- Guangdong Medical University, Guangdong Dongguan, 523808, China.
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China.
| | - Zhongzhou Yang
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Jie Li
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Hengwei Jin
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Mingyang Jiang
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| | - Congjia Shan
- Model Animal Research Center, Nanjing University, Jiangsu Nanjing, 210028, China
| |
Collapse
|
37
|
Harvey DC, Verma R, Sedaghat B, Hjelm BE, Morton SU, Seidman JG, Kumar SR. Mutations in genes related to myocyte contraction and ventricular septum development in non-syndromic tetralogy of Fallot. Front Cardiovasc Med 2023; 10:1249605. [PMID: 37840956 PMCID: PMC10569225 DOI: 10.3389/fcvm.2023.1249605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 09/08/2023] [Indexed: 10/17/2023] Open
Abstract
Objective Eighty percent of patients with a diagnosis of tetralogy of Fallot (TOF) do not have a known genetic etiology or syndrome. We sought to identify key molecular pathways and biological processes that are enriched in non-syndromic TOF, the most common form of cyanotic congenital heart disease, rather than single driver genes to elucidate the pathogenesis of this disease. Methods We undertook exome sequencing of 362 probands with non-syndromic TOF and their parents within the Pediatric Cardiac Genomics Consortium (PCGC). We identified rare (minor allele frequency <1 × 10-4), de novo variants to ascertain pathways and processes affected in this population to better understand TOF pathogenesis. Pathways and biological processes enriched in the PCGC TOF cohort were compared to 317 controls without heart defects (and their parents) from the Simons Foundation Autism Research Initiative (SFARI). Results A total of 120 variants in 117 genes were identified as most likely to be deleterious, with CHD7, CLUH, UNC13C, and WASHC5 identified in two probands each. Gene ontology analyses of these variants using multiple bioinformatic tools demonstrated significant enrichment in processes including cell cycle progression, chromatin remodeling, myocyte contraction and calcium transport, and development of the ventricular septum and ventricle. There was also a significant enrichment of target genes of SOX9, which is critical in second heart field development and whose loss results in membranous ventricular septal defects related to disruption of the proximal outlet septum. None of these processes was significantly enriched in the SFARI control cohort. Conclusion Innate molecular defects in cardiac progenitor cells and genes related to their viability and contractile function appear central to non-syndromic TOF pathogenesis. Future research utilizing our results is likely to have significant implications in stratification of TOF patients and delivery of personalized clinical care.
Collapse
Affiliation(s)
- Drayton C. Harvey
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Riya Verma
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Brandon Sedaghat
- Department of Medicine, Rosalind Franklin University School of Medicine and Science, Chicago, IL, United States
| | - Brooke E. Hjelm
- Translational Genomics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Sarah U. Morton
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA, United States
| | - Jon G. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, United States
| | - S. Ram Kumar
- Departments of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
38
|
Wu B, Wu B, Benkaci S, Shi L, Lu P, Park T, Morrow BE, Wang Y, Zhou B. Crk and Crkl Are Required in the Endocardial Lineage for Heart Valve Development. J Am Heart Assoc 2023; 12:e029683. [PMID: 37702066 PMCID: PMC10547300 DOI: 10.1161/jaha.123.029683] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/02/2023] [Indexed: 09/14/2023]
Abstract
Background Endocardial cells are a major progenitor population that gives rise to heart valves through endocardial cushion formation by endocardial to mesenchymal transformation and the subsequent endocardial cushion remodeling. Genetic variants that affect these developmental processes can lead to congenital heart valve defects. Crk and Crkl are ubiquitously expressed genes encoding cytoplasmic adaptors essential for cell signaling. This study aims to explore the specific role of Crk and Crkl in the endocardial lineage during heart valve development. Methods and Results We deleted Crk and Crkl specifically in the endocardial lineage. The resultant heart valve morphology was evaluated by histological analysis, and the underlying cellular and molecular mechanisms were investigated by immunostaining and quantitative reverse transcription polymerase chain reaction. We found that the targeted deletion of Crk and Crkl impeded the remodeling of endocardial cushions at the atrioventricular canal into the atrioventricular valves. We showed that apoptosis was temporally increased in the remodeling atrioventricular endocardial cushions, and this developmentally upregulated apoptosis was repressed by deletion of Crk and Crkl. Loss of Crk and Crkl also resulted in altered extracellular matrix production and organization in the remodeling atrioventricular endocardial cushions. These morphogenic defects were associated with altered expression of genes in BMP (bone morphogenetic protein), connective tissue growth factor, and WNT signaling pathways, and reduced extracellular signal-regulated kinase signaling activities. Conclusions Our findings support that Crk and Crkl have shared functions in the endocardial lineage that critically regulate atrioventricular valve development; together, they likely coordinate the morphogenic signals involved in the remodeling of the atrioventricular endocardial cushions.
Collapse
Affiliation(s)
- Bingruo Wu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Brian Wu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Sonia Benkaci
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Lijie Shi
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Pengfei Lu
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| | - Taeju Park
- Children’s Mercy Research Institute, Children’s Mercy Kansas City and Department of Pediatrics, University of Missouri‐Kansas City School of MedicineKansas CityMO
| | | | - Yidong Wang
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
- Cardiovascular Research Center, School of Basic Medical SciencesXi’an Jiaotong University Health Science CenterXi’anChina
| | - Bin Zhou
- Department of GeneticsAlbert Einstein College of MedicineBronxNY
| |
Collapse
|
39
|
Stathopoulou A, Wang P, Thellier C, Kelly RG, Zheng D, Scambler PJ. CHARGE syndrome-associated CHD7 acts at ISL1-regulated enhancers to modulate second heart field gene expression. Cardiovasc Res 2023; 119:2089-2105. [PMID: 37052590 PMCID: PMC10478754 DOI: 10.1093/cvr/cvad059] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/20/2022] [Accepted: 04/12/2023] [Indexed: 04/14/2023] Open
Abstract
AIMS Haploinsufficiency of the chromo-domain protein CHD7 underlies most cases of CHARGE syndrome, a multisystem birth defect including congenital heart malformation. Context specific roles for CHD7 in various stem, progenitor, and differentiated cell lineages have been reported. Previously, we showed severe defects when Chd7 is absent from cardiopharyngeal mesoderm (CPM). Here, we investigate altered gene expression in the CPM and identify specific CHD7-bound target genes with known roles in the morphogenesis of affected structures. METHODS AND RESULTS We generated conditional KO of Chd7 in CPM and analysed cardiac progenitor cells using transcriptomic and epigenomic analyses, in vivo expression analysis, and bioinformatic comparisons with existing datasets. We show CHD7 is required for correct expression of several genes established as major players in cardiac development, especially within the second heart field (SHF). We identified CHD7 binding sites in cardiac progenitor cells and found strong association with histone marks suggestive of dynamically regulated enhancers during the mesodermal to cardiac progenitor transition of mESC differentiation. Moreover, CHD7 shares a subset of its target sites with ISL1, a pioneer transcription factor in the cardiogenic gene regulatory network, including one enhancer modulating Fgf10 expression in SHF progenitor cells vs. differentiating cardiomyocytes. CONCLUSION We show that CHD7 interacts with ISL1, binds ISL1-regulated cardiac enhancers, and modulates gene expression across the mesodermal heart fields during cardiac morphogenesis.
Collapse
Affiliation(s)
- Athanasia Stathopoulou
- Developmental Biology of Birth Defects, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | | | - Robert G Kelly
- Aix-Marseille University, CNRS UMR 7288, IBDM, Marseille, France
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Neurology and Neurosciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Peter J Scambler
- Developmental Biology of Birth Defects, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| |
Collapse
|
40
|
Alcalde M, Toro R, Bonet F, Córdoba-Caballero J, Martínez-Barrios E, Ranea JA, Vallverdú-Prats M, Brugada R, Meraviglia V, Bellin M, Sarquella-Brugada G, Campuzano O. Role of microRNAs in arrhythmogenic cardiomyopathy: translation as biomarkers into clinical practice. Transl Res 2023; 259:72-82. [PMID: 37105319 DOI: 10.1016/j.trsl.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/11/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023]
Abstract
Arrhythmogenic cardiomyopathy is a rare inherited entity, characterized by a progressive fibro-fatty replacement of the myocardium. It leads to malignant arrhythmias and a high risk of sudden cardiac death. Incomplete penetrance and variable expressivity are hallmarks of this arrhythmogenic cardiac disease, where the first manifestation may be syncope and sudden cardiac death, often triggered by physical exercise. Early identification of individuals at risk is crucial to adopt protective and ideally personalized measures to prevent lethal episodes. The genetic analysis identifies deleterious rare variants in nearly 70% of cases, mostly in genes encoding proteins of the desmosome. However, other factors may modulate the phenotype onset and outcome of disease, such as microRNAs. These small noncoding RNAs play a key role in gene expression regulation and the network of cellular processes. In recent years, data focused on the role of microRNAs as potential biomarkers in arrhythmogenic cardiomyopathy have progressively increased. A better understanding of the functions and interactions of microRNAs will likely have clinical implications. Herein, we propose an exhaustive review of the literature regarding these noncoding RNAs, their versatile mechanisms of gene regulation and present novel targets in arrhythmogenic cardiomyopathy.
Collapse
Affiliation(s)
- Mireia Alcalde
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain
| | - Rocío Toro
- Medicine Department, School of Medicine, Cadiz, Spain; Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain.
| | - Fernando Bonet
- Medicine Department, School of Medicine, Cadiz, Spain; Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - José Córdoba-Caballero
- Medicine Department, School of Medicine, Cadiz, Spain; Research Unit, Biomedical Research and Innovation Institute of Cadiz (INiBICA), Puerta del Mar University Hospital, Cádiz, Spain
| | - Estefanía Martínez-Barrios
- Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, Barcelona, Spain; European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands; Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Juan Antonio Ranea
- Departamento de Biología Molecular y Bioquímica, Universidad de Málaga, Málaga, Spain; Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, Spain; Centro de Investigación Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Marta Vallverdú-Prats
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Cardiology Department, Hospital Josep Trueta, Girona, Spain
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, the Netherlands; Department of Biology, University of Padua, Padua, Italy; Veneto Institute of Molecular Medicine, Padua, Italy
| | - Georgia Sarquella-Brugada
- Pediatric Arrhythmias, Inherited Cardiac Diseases and Sudden Death Unit, Cardiology Department, Sant Joan de Déu Hospital, Barcelona, Spain; European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands; Arrítmies Pediàtriques, Cardiologia Genètica i Mort Sobtada, Malalties Cardiovasculars en el Desenvolupament, Institut de Recerca Sant Joan de Déu, Barcelona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Centro de Investigación Biomédica en Red. Enfermedades Cardiovasculares, Madrid, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain.
| |
Collapse
|
41
|
Francis R, San Agustin JT, Szabo Rogers HL, Cui C, Jonassen JA, Eguether T, Follit JA, Lo CW, Pazour GJ. Autonomous and non-cell autonomous etiology of ciliopathy associated structural birth defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544132. [PMID: 37333142 PMCID: PMC10274801 DOI: 10.1101/2023.06.07.544132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Ciliopathies are associated with wide spectrum of structural birth defects (SBD), indicating important roles for cilia in development. Here we provide novel insights into the temporospatial requirement for cilia in SBDs arising from deficiency in Ift140 , an intraflagellar transport protein regulating ciliogenesis. Ift140 deficient mice exhibit cilia defects accompanied by wide spectrum of SBDs including macrostomia (craniofacial defects), exencephaly, body wall defects, tracheoesophageal fistula, randomized heart looping, congenital heart defects (CHD), lung hypoplasia, renal anomalies, and polydactyly. Tamoxifen inducible CAG-Cre deletion of a floxed Ift140 allele between E5.5 to 9.5 revealed early requirement for Ift140 in left-right heart looping regulation, mid to late requirement for cardiac outflow septation and alignment, and late requirement for craniofacial development and body wall closure. Surprisingly, CHD was not observed with four Cre drivers targeting different lineages essential for heart development, but craniofacial defects and omphalocele were observed with Wnt1-Cre targeting neural crest and Tbx18-Cre targeting epicardial lineage and rostral sclerotome through which trunk neural crest cells migrate. These findings revealed cell autonomous role of cilia in cranial/trunk neural crest mediated craniofacial and body wall closure defects, while non-cell autonomous multi-lineage interactions underlie CHD pathogenesis, revealing unexpected developmental complexity for CHD associated with ciliopathy.
Collapse
|
42
|
Smallwood K, Watt KEN, Ide S, Baltrunaite K, Brunswick C, Inskeep K, Capannari C, Adam MP, Begtrup A, Bertola DR, Demmer L, Demo E, Devinsky O, Gallagher ER, Guillen Sacoto MJ, Jech R, Keren B, Kussmann J, Ladda R, Lansdon LA, Lunke S, Mardy A, McWalters K, Person R, Raiti L, Saitoh N, Saunders CJ, Schnur R, Skorvanek M, Sell SL, Slavotinek A, Sullivan BR, Stark Z, Symonds JD, Wenger T, Weber S, Whalen S, White SM, Winkelmann J, Zech M, Zeidler S, Maeshima K, Stottmann RW, Trainor PA, Weaver KN. POLR1A variants underlie phenotypic heterogeneity in craniofacial, neural, and cardiac anomalies. Am J Hum Genet 2023; 110:809-825. [PMID: 37075751 PMCID: PMC10183370 DOI: 10.1016/j.ajhg.2023.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/21/2023] [Indexed: 04/21/2023] Open
Abstract
Heterozygous pathogenic variants in POLR1A, which encodes the largest subunit of RNA Polymerase I, were previously identified as the cause of acrofacial dysostosis, Cincinnati-type. The predominant phenotypes observed in the cohort of 3 individuals were craniofacial anomalies reminiscent of Treacher Collins syndrome. We subsequently identified 17 additional individuals with 12 unique heterozygous variants in POLR1A and observed numerous additional phenotypes including neurodevelopmental abnormalities and structural cardiac defects, in combination with highly prevalent craniofacial anomalies and variable limb defects. To understand the pathogenesis of this pleiotropy, we modeled an allelic series of POLR1A variants in vitro and in vivo. In vitro assessments demonstrate variable effects of individual pathogenic variants on ribosomal RNA synthesis and nucleolar morphology, which supports the possibility of variant-specific phenotypic effects in affected individuals. To further explore variant-specific effects in vivo, we used CRISPR-Cas9 gene editing to recapitulate two human variants in mice. Additionally, spatiotemporal requirements for Polr1a in developmental lineages contributing to congenital anomalies in affected individuals were examined via conditional mutagenesis in neural crest cells (face and heart), the second heart field (cardiac outflow tract and right ventricle), and forebrain precursors in mice. Consistent with its ubiquitous role in the essential function of ribosome biogenesis, we observed that loss of Polr1a in any of these lineages causes cell-autonomous apoptosis resulting in embryonic malformations. Altogether, our work greatly expands the phenotype of human POLR1A-related disorders and demonstrates variant-specific effects that provide insights into the underlying pathogenesis of ribosomopathies.
Collapse
Affiliation(s)
- Kelly Smallwood
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Satoru Ide
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan; Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies), Mishima, Shizuoka, Japan
| | - Kristina Baltrunaite
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Chad Brunswick
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Katherine Inskeep
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Corrine Capannari
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Margaret P Adam
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | | | - Laurie Demmer
- Atrium Health's Levine Children's Hospital, Charlotte, NC, USA
| | - Erin Demo
- Sibley Heart Center, Atlanta, GA, USA
| | - Orrin Devinsky
- Department of Neurology, Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Emily R Gallagher
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Robert Jech
- Department of Neurology, Charles University, 1st Faculty of Medicine and General University Hospital in Prague, Prague, Czech Republic
| | - Boris Keren
- Genetic Department, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Jennifer Kussmann
- Division of Clinical Genetics, Department of Pediatrics, Children's Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA
| | - Roger Ladda
- Department of Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Lisa A Lansdon
- Department of Pathology and Laboratory Medicine, Children's Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA; Genomic Medicine Center, Children's Mercy Research Institute, 2401 Gillham Road, Kansas City, MO, USA; School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, USA
| | - Sebastian Lunke
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia; Australian Genomics, Melbourne, VIC, Australia
| | - Anne Mardy
- Department of Women's Health, University of Texas Austin Dell Medical Center, Austin, TX, USA
| | | | | | - Laura Raiti
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia
| | | | - Carol J Saunders
- Department of Pathology and Laboratory Medicine, Children's Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA; Genomic Medicine Center, Children's Mercy Research Institute, 2401 Gillham Road, Kansas City, MO, USA; School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, USA
| | | | - Matej Skorvanek
- Department of Neurology, P.J. Safarik University, Kosice, Slovak Republic; Department of Neurology, University Hospital of L. Pasteur, Kosice, Slovak Republic
| | - Susan L Sell
- Department of Pediatrics, Penn State Health Children's Hospital, Hershey, PA, USA
| | - Anne Slavotinek
- Division of Medical Genetics, Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Bonnie R Sullivan
- Division of Clinical Genetics, Department of Pediatrics, Children's Mercy Kansas City, 2401 Gillham Road, Kansas City, MO, USA
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia; Australian Genomics, Melbourne, VIC, Australia
| | - Joseph D Symonds
- Paediatric Neuroscience Research Group, Royal Hospital for Children, Glasgow G667AB, UK
| | - Tara Wenger
- Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Sacha Weber
- CCA-AHU de génétique clinique et de neurogénétique, Service de Génétique et de Neurologie, CHU de Caen, Caen, France
| | - Sandra Whalen
- Genetic Department, APHP, Sorbonne Université, Pitié-Salpêtrière Hospital, 47-83 Boulevard de l'Hôpital, 75013 Paris, France
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Flemington Road, Melbourne, VIC, Australia; University of Melbourne, Melbourne, VIC, Australia
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany; Lehrstuhl für Neurogenetik, Technische Universität München, Munich, Germany; Munich Cluster for Systems Neurology, SyNergy, Munich, Germany
| | - Michael Zech
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Shimriet Zeidler
- Department of Clinical Genetics, Erasmus MC, Rotterdam, the Netherlands
| | - Kazuhiro Maeshima
- Genome Dynamics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan; Department of Genetics, School of Life Science, Sokendai (Graduate University for Advanced Studies), Mishima, Shizuoka, Japan
| | - Rolf W Stottmann
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - K Nicole Weaver
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
43
|
Baral K, D'amato G, Kuschel B, Bogan F, Jones BW, Large CL, Whatley JD, Red-Horse K, Sharma B. APJ+ cells in the SHF contribute to the cells of aorta and pulmonary trunk through APJ signaling. Dev Biol 2023; 498:77-86. [PMID: 37037405 DOI: 10.1016/j.ydbio.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
Outflow tract develops from cardiac progenitor cells in the second heart field (SHF) domain. APJ, a G-Protein Coupled Receptor, is expressed by cardiac progenitor cells in the SHF. By lineage tracing APJ + SHF cells, we show that these cardiac progenitor cell contribute to the cells of outflow tract (OFT), which eventually give rise to aorta and pulmonary trunk/artery upon its morphogenesis. Furthermore, we show that early APJ + cells give rise to both aorta and pulmonary cells but late APJ + cells predominantly give rise to pulmonary cells. APJ is expressed by the outflow tract progenitors but its role in the SHF is unclear. We performed knockout studies to determine the role of APJ in SHF cell proliferation and survival. Our data suggested that APJ knockout in the SHF reduced the proliferation of SHF progenitors, while there was no significant impact on survival of the SHF progenitors. In addition, we show that ectopic overexpression of WNT in these cells disrupted aorta and pulmonary morphogenesis from outflow tract. Overall, our study have identified APJ + progenitor population within the SHF that give rise to aorta and pulmonary trunk/artery cells. Furthermore, we show that APJ signaling stimulate proliferation of these cells in the SHF.
Collapse
Affiliation(s)
- Kamal Baral
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Gaetano D'amato
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Bryce Kuschel
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Frank Bogan
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Brendan W Jones
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Colton L Large
- Department of Biology, Ball State University, Muncie, IN, USA
| | | | | | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN, USA.
| |
Collapse
|
44
|
Zawada D, Kornherr J, Meier AB, Santamaria G, Dorn T, Nowak-Imialek M, Ortmann D, Zhang F, Lachmann M, Dreßen M, Ortiz M, Mascetti VL, Harmer SC, Nobles M, Tinker A, De Angelis MT, Pedersen RA, Grote P, Laugwitz KL, Moretti A, Goedel A. Retinoic acid signaling modulation guides in vitro specification of human heart field-specific progenitor pools. Nat Commun 2023; 14:1722. [PMID: 37012244 PMCID: PMC10070453 DOI: 10.1038/s41467-023-36764-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 02/15/2023] [Indexed: 04/05/2023] Open
Abstract
Cardiogenesis relies on the precise spatiotemporal coordination of multiple progenitor populations. Understanding the specification and differentiation of these distinct progenitor pools during human embryonic development is crucial for advancing our knowledge of congenital cardiac malformations and designing new regenerative therapies. By combining genetic labelling, single-cell transcriptomics, and ex vivo human-mouse embryonic chimeras we uncovered that modulation of retinoic acid signaling instructs human pluripotent stem cells to form heart field-specific progenitors with distinct fate potentials. In addition to the classical first and second heart fields, we observed the appearance of juxta-cardiac field progenitors giving rise to both myocardial and epicardial cells. Applying these findings to stem-cell based disease modelling we identified specific transcriptional dysregulation in first and second heart field progenitors derived from stem cells of patients with hypoplastic left heart syndrome. This highlights the suitability of our in vitro differentiation platform for studying human cardiac development and disease.
Collapse
Affiliation(s)
- Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Jessica Kornherr
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Catanzaro, Italy
| | - Tatjana Dorn
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Monika Nowak-Imialek
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Daniel Ortmann
- Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Fangfang Zhang
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Mark Lachmann
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Martina Dreßen
- German Heart Center Munich, Department of Cardiovascular Surgery, Institute Insure - Technical University of Munich, School of Medicine and Health, Munich, Germany
| | | | - Victoria L Mascetti
- Bristol Heart Institute, Bristol Medical School, Translational Health Sciences, Bristol, UK
| | - Stephen C Harmer
- School of Physiology, Pharmacology and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Muriel Nobles
- Clinical Pharmacology & Precision Medicine, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrew Tinker
- Clinical Pharmacology & Precision Medicine, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Maria Teresa De Angelis
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Experimental and Clinical Medicine, University "Magna Graecia", Catanzaro, Italy
| | - Roger A Pedersen
- Department of Obstetrics and Gynecology, Stanford School of Medicine, Stanford University, Stanford, USA
| | - Phillip Grote
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- Department of Surgery, Yale University School of Medicine, New Haven, USA.
| | - Alexander Goedel
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
45
|
Kelly RG. The heart field transcriptional landscape at single-cell resolution. Dev Cell 2023; 58:257-266. [PMID: 36809764 DOI: 10.1016/j.devcel.2023.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/06/2022] [Accepted: 01/27/2023] [Indexed: 02/22/2023]
Abstract
Organogenesis requires the orchestrated development of multiple cell lineages that converge, interact, and specialize to generate coherent functional structures, exemplified by transformation of the cardiac crescent into a four-chambered heart. Cardiomyocytes originate from the first and second heart fields, which make different regional contributions to the definitive heart. In this review, a series of recent single-cell transcriptomic analyses, together with genetic tracing experiments, are discussed, providing a detailed panorama of the cardiac progenitor cell landscape. These studies reveal that first heart field cells originate in a juxtacardiac field adjacent to extraembryonic mesoderm and contribute to the ventrolateral side of the cardiac primordium. In contrast, second heart field cells are deployed dorsomedially from a multilineage-primed progenitor population via arterial and venous pole pathways. Refining our knowledge of the origin and developmental trajectories of cells that build the heart is essential to address outstanding challenges in cardiac biology and disease.
Collapse
Affiliation(s)
- Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Marseille, France.
| |
Collapse
|
46
|
Genetic lineage tracing identifies cardiac mesenchymal-to-adipose transition in an arrhythmogenic cardiomyopathy model. SCIENCE CHINA. LIFE SCIENCES 2023; 66:51-66. [PMID: 36322324 DOI: 10.1007/s11427-022-2176-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is one of the most common inherited cardiomyopathies, characterized by progressive fibrofatty replacement in the myocardium. However, the cellular origin of cardiac adipocytes in ACM remains largely unknown. Unraveling the cellular source of cardiac adipocytes in ACM would elucidate the underlying pathological process and provide a potential target for therapy. Herein, we generated an ACM mouse model by inactivating desmosomal gene desmoplakin in cardiomyocytes; and examined the adipogenic fates of several cell types in the disease model. The results showed that SOX9+, PDGFRa+, and PDGFRb+ mesenchymal cells, but not cardiomyocytes or smooth muscle cells, contribute to the intramyocardial adipocytes in the ACM model. Mechanistically, Bmp4 was highly expressed in the ACM mouse heart and functionally promoted cardiac mesenchymal-to-adipose transition in vitro.
Collapse
|
47
|
Yasuhara J, Schultz K, Bigelow AM, Garg V. Congenital aortic valve stenosis: from pathophysiology to molecular genetics and the need for novel therapeutics. Front Cardiovasc Med 2023; 10:1142707. [PMID: 37187784 PMCID: PMC10175644 DOI: 10.3389/fcvm.2023.1142707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Congenital aortic valve stenosis (AVS) is one of the most common valve anomalies and accounts for 3%-6% of cardiac malformations. As congenital AVS is often progressive, many patients, both children and adults, require transcatheter or surgical intervention throughout their lives. While the mechanisms of degenerative aortic valve disease in the adult population are partially described, the pathophysiology of adult AVS is different from congenital AVS in children as epigenetic and environmental risk factors play a significant role in manifestations of aortic valve disease in adults. Despite increased understanding of genetic basis of congenital aortic valve disease such as bicuspid aortic valve, the etiology and underlying mechanisms of congenital AVS in infants and children remain unknown. Herein, we review the pathophysiology of congenitally stenotic aortic valves and their natural history and disease course along with current management strategies. With the rapid expansion of knowledge of genetic origins of congenital heart defects, we also summarize the literature on the genetic contributors to congenital AVS. Further, this increased molecular understanding has led to the expansion of animal models with congenital aortic valve anomalies. Finally, we discuss the potential to develop novel therapeutics for congenital AVS that expand on integration of these molecular and genetic advances.
Collapse
Affiliation(s)
- Jun Yasuhara
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Correspondence: Jun Yasuhara Vidu Garg
| | - Karlee Schultz
- Medical Student Research Program, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Amee M. Bigelow
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| | - Vidu Garg
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, United States
- Heart Center, Nationwide Children’s Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, United States
- Correspondence: Jun Yasuhara Vidu Garg
| |
Collapse
|
48
|
Zhao K, Yang Z. The second heart field: the first 20 years. Mamm Genome 2022:10.1007/s00335-022-09975-8. [PMID: 36550326 DOI: 10.1007/s00335-022-09975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022]
Abstract
In 2001, three independent groups reported the identification of a novel cluster of progenitor cells that contribute to heart development in mouse and chicken embryos. This population of progenitor cells was designated as the second heart field (SHF), and a new research direction in heart development was launched. Twenty years have since passed and a comprehensive understanding of the SHF has been achieved. This review provides retrospective insights in to the contribution, the signaling regulatory networks and the epithelial properties of the SHF. It also includes the spatiotemporal characteristics of SHF development and interactions between the SHF and other types of cells during heart development. Although considerable efforts will be required to investigate the cellular heterogeneity of the SHF, together with its intricate regulatory networks and undefined mechanisms, it is expected that the burgeoning new technology of single-cell sequencing and precise lineage tracing will advance the comprehension of SHF function and its molecular signals. The advances in SHF research will translate to clinical applications and to the treatment of congenital heart diseases, especially conotruncal defects, as well as to regenerative medicine.
Collapse
Affiliation(s)
- Ke Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, and Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, 210093, China.
| |
Collapse
|
49
|
Garcia-Canadilla P, Mohun TJ, Bijnens B, Cook AC. Detailed quantification of cardiac ventricular myocardial architecture in the embryonic and fetal mouse heart by application of structure tensor analysis to high resolution episcopic microscopic data. Front Cell Dev Biol 2022; 10:1000684. [DOI: 10.3389/fcell.2022.1000684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/31/2022] [Indexed: 11/18/2022] Open
Abstract
The mammalian heart, which is one of the first organs to form and function during embryogenesis, develops from a simple tube into a complex organ able to efficiently pump blood towards the rest of the body. The progressive growth of the compact myocardium during embryonic development is accompanied by changes in its structural complexity and organisation. However, how myocardial myoarchitecture develops during embryogenesis remain poorly understood. To date, analysis of heart development has focused mainly on qualitative descriptions using selected 2D histological sections. High resolution episcopic microscopy (HREM) is a novel microscopic imaging technique that enables to obtain high-resolution three-dimensional images of the heart and perform detailed quantitative analyses of heart development. In this work, we performed a detailed characterization of the development of myocardial architecture in wildtype mice, from E14.5 to E18.5, by means of structure tensor analysis applied to HREM images of the heart. Our results shows that even at E14.5, myocytes are already aligned, showing a gradual change in their helical angle from positive angulation in the endocardium towards negative angulation in the epicardium. Moreover, there is gradual increase in the degree of myocardial organisation concomitant with myocardial growth. However, the development of the myoarchitecture is heterogeneous showing regional differences between ventricles, ventricular walls as well as between myocardial layers, with different growth patterning between the endocardium and epicardium. We also found that the percentage of circumferentially arranged myocytes within the LV significantly increases with gestational age. Finally, we found that fractional anisotropy (FA) within the LV gradually increases with gestational age, while the FA within RV remains unchanged.
Collapse
|
50
|
Deepe RN, Drummond JR, Wolters RA, Fitzgerald EA, Tarolli HG, Harvey AB, Wessels A. Sox9 Expression in the Second Heart Field; A Morphological Assessment of the Importance to Cardiac Development with Emphasis on Atrioventricular Septation. J Cardiovasc Dev Dis 2022; 9:376. [PMID: 36354775 PMCID: PMC9699451 DOI: 10.3390/jcdd9110376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Failure to form the septal structures that separate the left and right cardiac chambers results in defects that allow shunting of blood from one side of the heart to the other, leading to the mixing of oxygenated and de-oxygenated blood. The atrioventricular (AV) mesenchymal complex, consisting of the AV cushions, the Dorsal Mesenchymal Protrusion (DMP), and the mesenchymal cap, plays a crucial role in AV septation. Cells found in these structures derive from different cell lineages. In this study we have investigated the role of the transcription factor Sox9 in the Second Heart Field (SHF) with the emphasis on the formation of the atrioventricular septal complex. Using a mouse model in which Sox9 is conditionally deleted from the SHF we demonstrate that in this model virtually all mouse embryos develop septal abnormalities, including complete atrioventricular septal defects (cAVSDs) and isolated ventricular septal defects. Our morphological analyses indicate that perturbation of the development of the mesenchymal cap appears to play a crucial role in the pathogenesis of the atrial septal defects observed in the AVSDs and suggests that this component of the AV mesenchymal complex might play a more important role in AV septation than previously appreciated.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|