1
|
Wang S, Kang Y, Xie H. PKD2: An Important Membrane Protein in Organ Development. Cells 2024; 13:1722. [PMID: 39451240 PMCID: PMC11506562 DOI: 10.3390/cells13201722] [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: 08/25/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
PKD2 was first identified as the pathogenic protein for autosomal dominant polycystic kidney disease (ADPKD) and is widely recognized as an ion channel. Subsequent studies have shown that PKD2 is widely expressed in various animal tissues and plays a crucial role in tissue and organ development. Additionally, PKD2 is conserved from single-celled organisms to vertebrates. Here, we provide an overview of recent advances in the function of PKD2 in key model animals, focusing on the establishment of left-right organ asymmetry, renal homeostasis, cardiovascular development, and signal transduction in reproduction and mating. We specifically focus on the roles of PKD2 in development and highlight future prospects for PKD2 research.
Collapse
Affiliation(s)
- Shuo Wang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Haibo Xie
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
2
|
El-Nashar H, Sabry M, Tseng YT, Francis N, Latif N, Parker KH, Moore JE, Yacoub MH. Multiscale structure and function of the aortic valve apparatus. Physiol Rev 2024; 104:1487-1532. [PMID: 37732828 PMCID: PMC11495199 DOI: 10.1152/physrev.00038.2022] [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: 12/07/2022] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/22/2023] Open
Abstract
Whereas studying the aortic valve in isolation has facilitated the development of life-saving procedures and technologies, the dynamic interplay of the aortic valve and its surrounding structures is vital to preserving their function across the wide range of conditions encountered in an active lifestyle. Our view is that these structures should be viewed as an integrated functional unit, here referred to as the aortic valve apparatus (AVA). The coupling of the aortic valve and root, left ventricular outflow tract, and blood circulation is crucial for AVA's functions: unidirectional flow out of the left ventricle, coronary perfusion, reservoir function, and support of left ventricular function. In this review, we explore the multiscale biological and physical phenomena that underlie the simultaneous fulfillment of these functions. A brief overview of the tools used to investigate the AVA, such as medical imaging modalities, experimental methods, and computational modeling, specifically fluid-structure interaction (FSI) simulations, is included. Some pathologies affecting the AVA are explored, and insights are provided on treatments and interventions that aim to maintain quality of life. The concepts explained in this article support the idea of AVA being an integrated functional unit and help identify unanswered research questions. Incorporating phenomena through the molecular, micro, meso, and whole tissue scales is crucial for understanding the sophisticated normal functions and diseases of the AVA.
Collapse
Affiliation(s)
- Hussam El-Nashar
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Malak Sabry
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Biomedical Engineering, King's College London, London, United Kingdom
| | - Yuan-Tsan Tseng
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nadine Francis
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Najma Latif
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Kim H Parker
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - James E Moore
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Magdi H Yacoub
- Aswan Heart Research Centre, Magdi Yacoub Foundation, Cairo, Egypt
- Heart Science Centre, Magdi Yacoub Institute, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Liu Z, Liu Y, Yu Z, Tan C, Pek N, O'Donnell A, Wu A, Glass I, Winlaw DS, Guo M, Spence JR, Chen YW, Yutzey KE, Miao Y, Gu M. APOE-NOTCH axis governs elastogenesis during human cardiac valve remodeling. NATURE CARDIOVASCULAR RESEARCH 2024; 3:933-950. [PMID: 39196035 DOI: 10.1038/s44161-024-00510-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/19/2024] [Indexed: 08/29/2024]
Abstract
Valve remodeling is a process involving extracellular matrix organization and elongation of valve leaflets. Here, through single-cell RNA sequencing of human fetal valves, we identified an elastin-producing valve interstitial cell (VIC) subtype (apolipoprotein E (APOE)+, elastin-VICs) spatially located underneath valve endothelial cells (VECs) sensing unidirectional flow. APOE knockdown in fetal VICs resulted in profound elastogenesis defects. In valves with pulmonary stenosis (PS), we observed elastin fragmentation and decreased expression of APOE along with other genes regulating elastogenesis. Cell-cell interaction analysis revealed that jagged 1 (JAG1) from unidirectional VECs activates elastogenesis in elastin-VICs through NOTCH2. Similar observations were made in VICs cocultured with VECs under unidirectional flow. Notably, a drastic reduction of JAG1-NOTCH2 was also observed in PS valves. Lastly, we found that APOE controls JAG1-induced NOTCH activation and elastogenesis in VICs through the extracellular signal-regulated kinase pathway. Our study suggests important roles of both APOE and NOTCH in regulating elastogenesis during human valve remodeling.
Collapse
Affiliation(s)
- Ziyi Liu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Yu Liu
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford School of Medicine, Stanford, CA, USA
| | - Zhiyun Yu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Cheng Tan
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Nicole Pek
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Anna O'Donnell
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angeline Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ian Glass
- Department of Pediatrics, Genetic Medicine, University of Washington, Seattle, WA, USA
| | - David S Winlaw
- Cardiothoracic Surgery, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Surgery, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Minzhe Guo
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jason R Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Ya-Wen Chen
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institute for Airway Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine E Yutzey
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yifei Miao
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA.
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA.
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA.
- Cardiovascular Institute, Stanford School of Medicine, Stanford, CA, USA.
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Jiang X, Ly OT, Chen H, Zhang Z, Ibarra BA, Pavel MA, Brown GE, Sridhar A, Tofovic D, Swick A, Marszalek R, Vanoye CG, Navales F, George AL, Khetani SR, Rehman J, Gao Y, Darbar D, Saxena A. Transient titin-dependent ventricular defects during development lead to adult atrial arrhythmia and impaired contractility. iScience 2024; 27:110395. [PMID: 39100923 PMCID: PMC11296057 DOI: 10.1016/j.isci.2024.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/28/2024] [Accepted: 06/25/2024] [Indexed: 08/06/2024] Open
Abstract
Developmental causes of the most common arrhythmia, atrial fibrillation (AF), are poorly defined, with compensation potentially masking arrhythmic risk. Here, we delete 9 amino acids (Δ9) within a conserved domain of the giant protein titin's A-band in zebrafish and human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs). We find that ttna Δ9/Δ9 zebrafish embryos' cardiac morphology is perturbed and accompanied by reduced functional output, but ventricular function recovers within days. Despite normal ventricular function, ttna Δ9/Δ9 adults exhibit AF and atrial myopathy, which are recapitulated in TTN Δ9/Δ9-hiPSC-aCMs. Additionally, action potential is shortened and slow delayed rectifier potassium current (I Ks) is increased due to aberrant atrial natriuretic peptide (ANP) levels. Strikingly, suppression of I Ks in both models prevents AF and improves atrial contractility. Thus, a small internal deletion in titin causes developmental abnormalities that increase the risk of AF via ion channel remodeling, with implications for patients who harbor disease-causing variants in sarcomeric proteins.
Collapse
Affiliation(s)
- Xinghang Jiang
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Olivia T. Ly
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Hanna Chen
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ziwei Zhang
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Beatriz A. Ibarra
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Mahmud A. Pavel
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Grace E. Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - David Tofovic
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Abigail Swick
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Richard Marszalek
- Department of Physiology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Carlos G. Vanoye
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Fritz Navales
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Alfred L. George
- Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, IL 60607, USA
| | - Yu Gao
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
- Department of Medicine, Jesse Brown Veterans Administration, Chicago, IL 60612, USA
| | - Ankur Saxena
- Department of Cell, Developmental, and Integrative Biology, UAB Heersink School of Medicine, Birmingham, AL 35233, USA
- Department of Biological Sciences, University of Illinois Chicago, Chicago, IL 60607, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
- O'Neal Comprehensive Cancer Center, Birmingham, AL 35233, USA
| |
Collapse
|
5
|
Andrews TGR, Priya R. The Mechanics of Building Functional Organs. Cold Spring Harb Perspect Biol 2024:a041520. [PMID: 38886066 PMCID: PMC7616527 DOI: 10.1101/cshperspect.a041520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Organ morphogenesis is multifaceted, multiscale, and fundamentally a robust process. Despite the complex and dynamic nature of embryonic development, organs are built with reproducible size, shape, and function, allowing them to support organismal growth and life. This striking reproducibility of tissue form exists because morphogenesis is not entirely hardwired. Instead, it is an emergent product of mechanochemical information flow, operating across spatial and temporal scales-from local cellular deformations to organ-scale form and function, and back. In this review, we address the mechanical basis of organ morphogenesis, as understood by observations and experiments in living embryos. To this end, we discuss how mechanical information controls the emergence of a highly conserved set of structural motifs that shape organ architectures across the animal kingdom: folds and loops, tubes and lumens, buds, branches, and networks. Moving forward, we advocate for a holistic conceptual framework for the study of organ morphogenesis, which rests on an interdisciplinary toolkit and brings the embryo center stage.
Collapse
Affiliation(s)
| | - Rashmi Priya
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| |
Collapse
|
6
|
Abello J, Yin Y, Zhao Y, Maurer J, Lee J, Bodell C, Clevenger AJ, Burton Z, Goeckel ME, Lin M, Grainger S, Halabi CM, Raghavan SA, Sah R, Stratman AN. Endothelial cell Piezo1 promotes vascular smooth muscle cell differentiation on large arteries. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598539. [PMID: 38915529 PMCID: PMC11195244 DOI: 10.1101/2024.06.11.598539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Vascular stabilization is a mechanosensitive process, in part driven by blood flow. Here, we demonstrate the involvement of the mechanosensitive ion channel, Piezo1, in promoting arterial accumulation of vascular smooth muscle cells (vSMCs) during zebrafish development. Using a series of small molecule antagonists or agonists to temporally regulate Piezo1 activity, we identified a role for the Piezo1 channel in regulating klf2a levels and altered targeting of vSMCs between arteries and veins. Increasing Piezo1 activity suppressed klf2a and increased vSMC association with the cardinal vein, while inhibition of Piezo1 activity increased klf2a levels and decreased vSMC association with arteries. We supported the small molecule data with in vivo genetic suppression of piezo1 and 2 in zebrafish, resulting in loss of transgelin+ vSMCs on the dorsal aorta. Further, endothelial cell (EC)-specific Piezo1 knockout in mice was sufficient to decrease vSMC accumulation along the descending dorsal aorta during development, thus phenocopying our zebrafish data, and supporting functional conservation of Piezo1 in mammals. To determine mechanism, we used in vitro modeling assays to demonstrate that differential sensing of pulsatile versus laminar flow forces across endothelial cells changes the expression of mural cell differentiation genes. Together, our findings suggest a crucial role for EC Piezo1 in sensing force within large arteries to mediate mural cell differentiation and stabilization of the arterial vasculature.
Collapse
Affiliation(s)
- Javier Abello
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ying Yin
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yonghui Zhao
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Josh Maurer
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Jihui Lee
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Cherokee Bodell
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Abigail J. Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Zarek Burton
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Megan E. Goeckel
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Michelle Lin
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Carmen M. Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Shreya A. Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Rajan Sah
- Department of Internal Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
- Center for Cardiovascular Research, Washington University, St Louis, MO, USA
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
Yang D, Jian Z, Tang C, Chen Z, Zhou Z, Zheng L, Peng X. Zebrafish Congenital Heart Disease Models: Opportunities and Challenges. Int J Mol Sci 2024; 25:5943. [PMID: 38892128 PMCID: PMC11172925 DOI: 10.3390/ijms25115943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Congenital heart defects (CHDs) are common human birth defects. Genetic mutations potentially cause the exhibition of various pathological phenotypes associated with CHDs, occurring alone or as part of certain syndromes. Zebrafish, a model organism with a strong molecular conservation similar to humans, is commonly used in studies on cardiovascular diseases owing to its advantageous features, such as a similarity to human electrophysiology, transparent embryos and larvae for observation, and suitability for forward and reverse genetics technology, to create various economical and easily controlled zebrafish CHD models. In this review, we outline the pros and cons of zebrafish CHD models created by genetic mutations associated with single defects and syndromes and the underlying pathogenic mechanism of CHDs discovered in these models. The challenges of zebrafish CHD models generated through gene editing are also discussed, since the cardiac phenotypes resulting from a single-candidate pathological gene mutation in zebrafish might not mirror the corresponding human phenotypes. The comprehensive review of these zebrafish CHD models will facilitate the understanding of the pathogenic mechanisms of CHDs and offer new opportunities for their treatments and intervention strategies.
Collapse
|
8
|
da Silva AR, Gunawan F, Boezio GLM, Faure E, Théron A, Avierinos JF, Lim S, Jha SG, Ramadass R, Guenther S, Looso M, Zaffran S, Juan T, Stainier DYR. egr3 is a mechanosensitive transcription factor gene required for cardiac valve morphogenesis. SCIENCE ADVANCES 2024; 10:eadl0633. [PMID: 38748804 PMCID: PMC11095463 DOI: 10.1126/sciadv.adl0633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 04/11/2024] [Indexed: 05/19/2024]
Abstract
Biomechanical forces, and their molecular transducers, including key mechanosensitive transcription factor genes, such as KLF2, are required for cardiac valve morphogenesis. However, klf2 mutants fail to completely recapitulate the valveless phenotype observed under no-flow conditions. Here, we identify the transcription factor EGR3 as a conserved biomechanical force transducer critical for cardiac valve formation. We first show that egr3 null zebrafish display a complete and highly penetrant loss of valve leaflets, leading to severe blood regurgitation. Using tissue-specific loss- and gain-of-function tools, we find that during cardiac valve formation, Egr3 functions cell-autonomously in endothelial cells, and identify one of its effectors, the nuclear receptor Nr4a2b. We further find that mechanical forces up-regulate egr3/EGR3 expression in the developing zebrafish heart and in porcine valvular endothelial cells, as well as during human aortic valve remodeling. Altogether, these findings reveal that EGR3 is necessary to transduce the biomechanical cues required for zebrafish cardiac valve morphogenesis, and potentially for pathological aortic valve remodeling in humans.
Collapse
Affiliation(s)
- Agatha Ribeiro da Silva
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Felix Gunawan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Giulia L. M. Boezio
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Emilie Faure
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Alexis Théron
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Chirurgie Cardiaque, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - Jean-François Avierinos
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
- Service de Cardiologie, AP-HM, Hôpital de la Timone, 13005 Marseille, France
| | - SoEun Lim
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Shivam Govind Jha
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Radhan Ramadass
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
| | - Stefan Guenther
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stéphane Zaffran
- Aix Marseille Université, INSERM, MMG, U1251, 13005 Marseille, France
| | - Thomas Juan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| |
Collapse
|
9
|
Martin M, Chen CY, McCowan T, Wells S. Differential Development of the Chordae Tendineae and Anterior Leaflet of the Bovine Mitral Valve. J Cardiovasc Dev Dis 2024; 11:106. [PMID: 38667724 PMCID: PMC11050492 DOI: 10.3390/jcdd11040106] [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: 03/04/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
There is increasing evidence that some adult mitral valve pathologies may have developmental origins involving errors in cell signaling and protein deposition during valvulogenesis. While early and late gestational stages are well-documented in zebrafish, chicks, and small mammalian models, longitudinal studies in large mammals with a similar gestational period to humans are lacking. Further, the mechanism of chordae tendineae formation and multiplication remains unclear. The current study presents a comprehensive examination of mitral anterior leaflet and chordae tendineae development in a bovine model (a large mammal with the same gestational period as humans). Remarkably distinct from small mammals, bovine development displayed early branched chordae, with increasing attachments only until birth, while the anterior leaflet grew both during gestation and postnatally. Chordae also exhibited accelerated collagen deposition, maturation, and crimp development during gestation. These findings suggest that the bovine anterior leaflet and chordae tendineae possess unique processes of development despite being a continuous collagenous structure and could provide greater insight into human valve development.
Collapse
Affiliation(s)
- Meghan Martin
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
| | - Chih-Ying Chen
- Medical Sciences Program, Faculties of Science and Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (C.-Y.C.); (T.M.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Timothy McCowan
- Medical Sciences Program, Faculties of Science and Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (C.-Y.C.); (T.M.)
- Integrated Science Program, Faculty of Science, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Sarah Wells
- School of Biomedical Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada;
- Medical Sciences Program, Faculties of Science and Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (C.-Y.C.); (T.M.)
| |
Collapse
|
10
|
Furtado J, Eichmann A. Vascular development, remodeling and maturation. Curr Top Dev Biol 2024; 159:344-370. [PMID: 38729681 DOI: 10.1016/bs.ctdb.2024.02.001] [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] [Indexed: 05/12/2024]
Abstract
The development of the vascular system is crucial in supporting the growth and health of all other organs in the body, and vascular system dysfunction is the major cause of human morbidity and mortality. This chapter discusses three successive processes that govern vascular system development, starting with the differentiation of the primitive vascular system in early embryonic development, followed by its remodeling into a functional circulatory system composed of arteries and veins, and its final maturation and acquisition of an organ specific semi-permeable barrier that controls nutrient uptake into tissues and hence controls organ physiology. Along these steps, endothelial cells forming the inner lining of all blood vessels acquire extensive heterogeneity in terms of gene expression patterns and function, that we are only beginning to understand. These advances contribute to overall knowledge of vascular biology and are predicted to unlock the unprecedented therapeutic potential of the endothelium as an avenue for treatment of diseases associated with dysfunctional vasculature.
Collapse
Affiliation(s)
- Jessica Furtado
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, United States; Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Anne Eichmann
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT, United States; Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States; Paris Cardiovascular Research Center, Inserm U970, Université Paris, Paris, France.
| |
Collapse
|
11
|
Juan T, Bellec M, Cardoso B, Athéa H, Fukuda N, Albu M, Günther S, Looso M, Stainier DYR. Control of cardiac contractions using Cre-lox and degron strategies in zebrafish. Proc Natl Acad Sci U S A 2024; 121:e2309842121. [PMID: 38194447 PMCID: PMC10801847 DOI: 10.1073/pnas.2309842121] [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: 06/12/2023] [Accepted: 11/27/2023] [Indexed: 01/11/2024] Open
Abstract
Cardiac contractions and hemodynamic forces are essential for organ development and homeostasis. Control over cardiac contractions can be achieved pharmacologically or optogenetically. However, these approaches lack specificity or require direct access to the heart. Here, we compare two genetic approaches to control cardiac contractions by modulating the levels of the essential sarcomeric protein Tnnt2a in zebrafish. We first recombine a newly generated tnnt2a floxed allele using multiple lines expressing Cre under the control of cardiomyocyte-specific promoters, and show that it does not recapitulate the tnnt2a/silent heart mutant phenotype in embryos. We show that this lack of early cardiac contraction defects is due, at least in part, to the long half-life of tnnt2a mRNA, which masks the gene deletion effects until the early larval stages. We then generate an endogenous Tnnt2a-eGFP fusion line that we use together with the zGRAD system to efficiently degrade Tnnt2a in all cardiomyocytes. Using single-cell transcriptomics, we find that Tnnt2a depletion leads to cardiac phenotypes similar to those observed in tnnt2a mutants, with a loss of blood and pericardial flow-dependent cell types. Furthermore, we achieve conditional degradation of Tnnt2a-eGFP by splitting the zGRAD protein into two fragments that, when combined with the cpFRB2-FKBP system, can be reassembled upon rapamycin treatment. Thus, this Tnnt2a degradation line enables non-invasive control of cardiac contractions with high spatial and temporal specificity and will help further understand how they shape organ development and homeostasis.
Collapse
Affiliation(s)
- Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
| | - Maëlle Bellec
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Bárbara Cardoso
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Héloïse Athéa
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Nana Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Marga Albu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
| | - Stefan Günther
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
- Bioinformatics Core Unit, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim61231, Germany
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz- Kreislaufforschung), Bad Nauheim61231, Germany
- Cardio-Pulmonary Institute, Bad Nauheim61231, Germany
| |
Collapse
|
12
|
Auman HJ, Fernandes IH, Berríos-Otero CA, Colombo S, Yelon D. Zebrafish smarcc1a mutants reveal requirements for BAF chromatin remodeling complexes in distinguishing the atrioventricular canal from the cardiac chambers. Dev Dyn 2024; 253:157-172. [PMID: 37083132 PMCID: PMC10589389 DOI: 10.1002/dvdy.595] [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: 10/31/2022] [Revised: 03/13/2023] [Accepted: 04/08/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND Essential patterning processes transform the heart tube into a compartmentalized organ with distinct chambers separated by an atrioventricular canal (AVC). This transition involves the refinement of expression of genes that are first found broadly throughout the heart tube and then become restricted to the AVC. Despite the importance of cardiac patterning, we do not fully understand the mechanisms that limit gene expression to the AVC. RESULTS We show that the zebrafish gene smarcc1a, encoding a BAF chromatin remodeling complex subunit homologous to mammalian BAF155, is critical for cardiac patterning. In smarcc1a mutants, myocardial differentiation and heart tube assembly appear to proceed normally. Subsequently, the smarcc1a mutant heart fails to exhibit refinement of gene expression patterns to the AVC, and the persistence of broad gene expression is accompanied by failure of chamber expansion. In addition to their cardiac defects, smarcc1a mutants lack pectoral fins, indicating similarity to tbx5a mutants. However, comparison of smarcc1a and tbx5a mutants suggests that perturbation of tbx5a function is not sufficient to cause the smarcc1a mutant phenotype. CONCLUSIONS Our data indicate an important role for Smarcc1a-containing chromatin remodeling complexes in regulating the changes in gene expression and morphology that distinguish the AVC from the cardiac chambers.
Collapse
Affiliation(s)
- Heidi J. Auman
- Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - Ivy H. Fernandes
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Sophie Colombo
- Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - Deborah Yelon
- Skirball Institute, New York University School of Medicine, New York, NY, 10016, USA
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
13
|
Gregorovicova M, Lashkarinia SS, Yap CH, Tomek V, Sedmera D. Hemodynamics During Development and Postnatal Life. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:201-226. [PMID: 38884713 DOI: 10.1007/978-3-031-44087-8_11] [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
A well-developed heart is essential for embryonic survival. There are constant interactions between cardiac tissue motion and blood flow, which determine the heart shape itself. Hemodynamic forces are a powerful stimulus for cardiac growth and differentiation. Therefore, it is particularly interesting to investigate how the blood flows through the heart and how hemodynamics is linked to a particular species and its development, including human. The appropriate patterns and magnitude of hemodynamic stresses are necessary for the proper formation of cardiac structures, and hemodynamic perturbations have been found to cause malformations via identifiable mechanobiological molecular pathways. There are significant differences in cardiac hemodynamics among vertebrate species, which go hand in hand with the presence of specific anatomical structures. However, strong similarities during development suggest a common pattern for cardiac hemodynamics in human adults. In the human fetal heart, hemodynamic abnormalities during gestation are known to progress to congenital heart malformations by birth. In this chapter, we discuss the current state of the knowledge of the prenatal cardiac hemodynamics, as discovered through small and large animal models, as well as from clinical investigations, with parallels gathered from the poikilotherm vertebrates that emulate some hemodynamically significant human congenital heart diseases.
Collapse
Affiliation(s)
- Martina Gregorovicova
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Choon Hwai Yap
- Department of Bioengineering, Imperial College, London, UK
| | - Viktor Tomek
- Pediatric Cardiology, Motol University Hospital, Prague, Czech Republic
| | - David Sedmera
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czech Republic.
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
14
|
Nagorska A, Zaucker A, Lambert F, Inman A, Toral-Perez S, Gorodkin J, Wan Y, Smutny M, Sampath K. Translational control of furina by an RNA regulon is important for left-right patterning, heart morphogenesis and cardiac valve function. Development 2023; 150:dev201657. [PMID: 38032088 PMCID: PMC10730018 DOI: 10.1242/dev.201657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
Heart development is a complex process that requires asymmetric positioning of the heart, cardiac growth and valve morphogenesis. The mechanisms controlling heart morphogenesis and valve formation are not fully understood. The pro-convertase FurinA functions in heart development across vertebrates. How FurinA activity is regulated during heart development is unknown. Through computational analysis of the zebrafish transcriptome, we identified an RNA motif in a variant FurinA transcript harbouring a long 3' untranslated region (3'UTR). The alternative 3'UTR furina isoform is expressed prior to organ positioning. Somatic deletions in the furina 3'UTR lead to embryonic left-right patterning defects. Reporter localisation and RNA-binding assays show that the furina 3'UTR forms complexes with the conserved RNA-binding translational repressor, Ybx1. Conditional ybx1 mutant embryos show premature and increased Furin reporter expression, abnormal cardiac morphogenesis and looping defects. Mutant ybx1 hearts have an expanded atrioventricular canal, abnormal sino-atrial valves and retrograde blood flow from the ventricle to the atrium. This is similar to observations in humans with heart valve regurgitation. Thus, the furina 3'UTR element/Ybx1 regulon is important for translational repression of FurinA and regulation of heart development.
Collapse
Affiliation(s)
- Agnieszka Nagorska
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Andreas Zaucker
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Finnlay Lambert
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore 138672
| | - Angus Inman
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Sara Toral-Perez
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Jan Gorodkin
- Center for non-coding RNAs in Technology and Health, Department of Veterinary and Animal Sciences, Faculty for Health and Medical Sciences, University of Copenhagen, Grønnega °rdsvej 3, 1870 Frederiksberg C, Denmark
| | - Yue Wan
- Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore 138672
| | - Michael Smutny
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Mechanochemical Cell Biology, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Karuna Sampath
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Mechanochemical Cell Biology, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Early Life, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| |
Collapse
|
15
|
Maung Ye SS, Phng LK. A cell-and-plasma numerical model reveals hemodynamic stress and flow adaptation in zebrafish microvessels after morphological alteration. PLoS Comput Biol 2023; 19:e1011665. [PMID: 38048371 PMCID: PMC10721208 DOI: 10.1371/journal.pcbi.1011665] [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: 05/23/2023] [Revised: 12/14/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
The development of a functional cardiovascular system ensures a sustainable oxygen, nutrient and hormone delivery system for successful embryonic development and homeostasis in adulthood. While early vessels are formed by biochemical signaling and genetic programming, the onset of blood flow provides mechanical cues that participate in vascular remodeling of the embryonic vascular system. The zebrafish is a prolific animal model for studying the quantitative relationship between blood flow and vascular morphogenesis due to a combination of favorable factors including blood flow visualization in optically transparent larvae. In this study, we have developed a cell-and-plasma blood transport model using computational fluid dynamics (CFD) to understand how red blood cell (RBC) partitioning affect lumen wall shear stress (WSS) and blood pressure in zebrafish trunk blood vascular networks with altered rheology and morphology. By performing live imaging of embryos with reduced hematocrit, we discovered that cardiac output and caudal artery flow rates were maintained. These adaptation trends were recapitulated in our CFD models, which showed reduction in network WSS via viscosity reduction in the caudal artery/vein and via pressure gradient weakening in the intersegmental vessels (ISVs). Embryos with experimentally reduced lumen diameter showed reduced cardiac output and caudal artery flow rate. Factoring in this trend into our CFD models, simulations highlighted that lumen diameter reduction increased vessel WSS but this increase was mitigated by flow reduction due to the adaptive network pressure gradient weakening. Additionally, hypothetical network CFD models with different vessel lumen diameter distribution characteristics indicated the significance of axial variation in lumen diameter and cross-sectional shape for establishing physiological WSS gradients along ISVs. In summary, our work demonstrates how both experiment-driven and hypothetical CFD modeling can be employed for the study of blood flow physiology during vascular remodeling.
Collapse
Affiliation(s)
- Swe Soe Maung Ye
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| | - Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, Japan
| |
Collapse
|
16
|
Zhu H, Liao D, Mehmood MA, Huang Y, Yuan W, Zheng J, Ma Y, Peng Y, Tian G, Xiao X, Lan C, Li L, Xu K, Lu H, Wang N. Systolic heart failure induced by butylparaben in zebrafish is caused through oxidative stress and immunosuppression. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 268:115692. [PMID: 37981439 DOI: 10.1016/j.ecoenv.2023.115692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/20/2023] [Accepted: 11/12/2023] [Indexed: 11/21/2023]
Abstract
Due to Butylparaben (BuP) widespread application in cosmetics, food, pharmaceuticals, and its presence as an environmental residue, human and animal exposure to BuP is common, potentially posing hazards to both human and animal health. Congenital heart disease is already a serious problem. However, the effects of BuP on the developing heart and its underlying mechanisms remain unclear. Here, zebrafish embryos were exposed to environmentally and human-relevant concentrations of BuP (0.6 mg/L, 1.2 mg/L, and 1.8 mg/L, calculated but not measured) at 6 h post-fertilization (hpf) and were treated until 72 hpf. Exposure to BuP led to cardiac morphological defects and cardiac dysfunction in zebrafish embryos, manifesting symptoms similar to systolic heart failure. The etiology of BuP-induced systolic heart failure in zebrafish embryos is multifactorial, including cardiomyocyte apoptosis, endocardial and atrioventricular valve damage, insufficient myocardial energy, impaired Ca2+ homeostasis, depletion of cardiac-resident macrophages, cardiac immune non-responsiveness, and cardiac oxidative stress. However, excessive accumulation of reactive oxygen species (ROS) in the cardiac region and cardiac immunosuppression (depletion of cardiac-resident macrophages and cardiac immune non-responsiveness) may be the predominant factors. In conclusion, this study indicates that BuP is a potential hazardous substance that can cause adverse effects on the developing heart and provides evidence and insights into the pathological mechanisms by which BuP leads to cardiac dysfunction. It may help to prevent the BuP-based congenital heart disease heart failure in human through ameliorating strategies and BuP discharge policies, while raising awareness to prevent the misuse of preservatives.
Collapse
Affiliation(s)
- Hui Zhu
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China; Wuliangye Group Co., Ltd., Yibin 644007, China; Engineering Technology Research Center of Special Grain for Wine Making, Yibin 644000, China
| | - Dalong Liao
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China
| | - Muhammad Aamer Mehmood
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China; Bioenergy Research Center, Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330029, Jiangxi, China
| | - Wei Yuan
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Jia Zheng
- Wuliangye Group Co., Ltd., Yibin 644007, China
| | - Yi Ma
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China; Engineering Technology Research Center of Special Grain for Wine Making, Yibin 644000, China
| | - Yuyang Peng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Guiyou Tian
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Xiaoping Xiao
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China
| | - Chaohua Lan
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China
| | - Linman Li
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China
| | - Kewei Xu
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou 341000, China; Affiliated Hospital of Jinggangshan University, Center for Clinical Medicine Research of Jinggangshan University, China.
| | - Ning Wang
- School of Bioengineering, Sichuan University of Science & Engineering, Zigong 643000, China; Chengdu Chongqing Shuangcheng economic circle (Luzhou) advanced technology research institute, Luzhou 646000, China; Engineering Technology Research Center of Special Grain for Wine Making, Yibin 644000, China.
| |
Collapse
|
17
|
Mohl W, Kiseleva Z, Jusic A, Bruckner M, Mader RM. Signs and signals limiting myocardial damage using PICSO: a scoping review decoding paradigm shifts toward a new encounter. Front Cardiovasc Med 2023; 10:1030842. [PMID: 37229230 PMCID: PMC10204926 DOI: 10.3389/fcvm.2023.1030842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 04/14/2023] [Indexed: 05/27/2023] Open
Abstract
Background Inducing recovery in myocardial ischemia is limited to a timely reopening of infarct vessels and clearing the cardiac microcirculation, but additional molecular factors may impact recovery. Objective In this scoping review, we identify the paradigm shifts decoding the branching points of experimental and clinical evidence of pressure-controlled intermittent coronary sinus occlusion (PICSO), focusing on myocardial salvage and molecular implications on infarct healing and repair. Design The reporting of evidence was structured chronologically, describing the evolution of the concept from mainstream research to core findings dictating a paradigm change. All data reported in this scoping review are based on published data, but new evaluations are also included. Results Previous findings relate hemodynamic PICSO effects clearing reperfused microcirculation to myocardial salvage. The activation of venous endothelium opened a new avenue for understanding PICSO. A flow-sensitive signaling molecule, miR-145-5p, showed a five-fold increase in porcine myocardium subjected to PICSO.Verifying our theory of "embryonic recall," an upregulation of miR-19b and miR-101 significantly correlates to the time of pressure increase in cardiac veins during PICSO (r2 = 0.90, p < 0.05; r2 = 0.98, p < 0.03), suggesting a flow- and pressure-dependent secretion of signaling molecules into the coronary circulation. Furthermore, cardiomyocyte proliferation by miR-19b and the protective role of miR-101 against remodeling show another potential interaction of PICSO in myocardial healing. Conclusion Molecular signaling during PICSO may contribute to retroperfusion toward deprived myocardium and clearing the reperfused cardiac microcirculation. A burst of specific miRNA reiterating embryonic molecular pathways may play a role in targeting myocardial jeopardy and will be an essential therapeutic contribution in limiting infarcts in recovering patients.
Collapse
Affiliation(s)
- Werner Mohl
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Zlata Kiseleva
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Alem Jusic
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Matthäus Bruckner
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Robert M. Mader
- Department of Medicine I, Comprehensive Cancer Center of the Medical University of Vienna, Vienna,Austria
| |
Collapse
|
18
|
Brown AL, Gerosa FM, Wang J, Hsiai T, Marsden AL. Recent advances in quantifying the mechanobiology of cardiac development via computational modeling. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100428. [PMID: 36583220 PMCID: PMC9794182 DOI: 10.1016/j.cobme.2022.100428] [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] [Indexed: 11/23/2022]
Abstract
Mechanical forces are essential for coordinating cardiac morphogenesis, but much remains to be discovered about the interactions between mechanical forces and the mechanotransduction pathways they activate. Due to the elaborate and fundamentally multi-physics and multi-scale nature of cardiac mechanobiology, a complete understanding requires multiple experimental and analytical techniques. We identify three fundamental tools used in the field to probe these interactions: high resolution imaging, genetic and molecular analysis, and computational modeling. In this review, we focus on computational modeling and present recent studies employing this tool to investigate the mechanobiological pathways involved with cardiac development. These works demonstrate that understanding the detailed spatial and temporal patterns of biomechanical forces is crucial to building a comprehensive understanding of mechanobiology during cardiac development, and that computational modeling is an effective and efficient tool for obtaining such detail. In this context, multidisciplinary studies combining all three tools present the most compelling results.
Collapse
Affiliation(s)
- Aaron L. Brown
- Stanford University, Department of Mechanical Engineering, Stanford, USA, CA, 94305
| | - Fannie M. Gerosa
- Stanford University, Department of Pediatrics, Stanford, USA, CA 94305
- Stanford University, Institute for Computational & Mathematical Engineering, Stanford, USA, CA 94305
| | - Jing Wang
- University of California Los Angeles, Department of Bioengineering, Los Angeles, CA 90095
| | - Tzung Hsiai
- University of California Los Angeles, Department of Bioengineering, Los Angeles, CA 90095
- University of California Los Angeles, Division of Cardiology, Los Angeles, CA 90095
| | - Alison L. Marsden
- Stanford University, Department of Mechanical Engineering, Stanford, USA, CA, 94305
- Stanford University, Department of Pediatrics, Stanford, USA, CA 94305
- Stanford University, Institute for Computational & Mathematical Engineering, Stanford, USA, CA 94305
- Stanford University, Department of Bioengineering, Stanford, USA, CA 94305
| |
Collapse
|
19
|
Salehin N, Teranikar T, Lee J, Chuong CJ. Ventricular anisotropic deformation and contractile function of the developing heart of zebrafish in vivo. Dev Dyn 2023; 252:247-262. [PMID: 36057940 DOI: 10.1002/dvdy.536] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/10/2022] [Accepted: 08/29/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The developing zebrafish ventricle generates higher intraventricular pressure (IVP) with increasing stroke volume and cardiac output at different developmental stages to meet the metabolic demands of the rapidly growing embryo (Salehin et al. Ann Biomed Eng, 2021;49(9): 2080-2093). To understand the changing role of the developing embryonic heart, we studied its biomechanical characteristics during in vivo cardiac cycles. By combining changes in wall strains and IVP measurements, we assessed ventricular wall stiffness during diastolic filling and the ensuing systolic IVP-generation capacity during 3-, 4-, and 5-day post fertilization (dpf). We further examined the anisotropy of wall deformation, in different regions within the ventricle, throughout a complete cardiac cycle. RESULTS We found the ventricular walls grow increasingly stiff during diastolic filling with a corresponding increase in IVP-generation capacity from 3- to 4- and 5-dpf groups. In addition, we found the corresponding increasing level of anisotropic wall deformation through cardiac cycles that favor the latitudinal direction, with the most pronounced found in the equatorial region of the ventricle. CONCLUSIONS From 3- to 4- and 5-dpf groups, the ventricular wall myocardium undergoes increasing level of anisotropic deformation. This, in combination with the increasing wall stiffness and IVP-generation capacity, allows the developing heart to effectively pump blood to meet the rapidly growing embryo's needs.
Collapse
Affiliation(s)
- Nabid Salehin
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Tanveer Teranikar
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Cheng-Jen Chuong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
20
|
Juan T, Ribeiro da Silva A, Cardoso B, Lim S, Charteau V, Stainier DYR. Multiple pkd and piezo gene family members are required for atrioventricular valve formation. Nat Commun 2023; 14:214. [PMID: 36639367 PMCID: PMC9839778 DOI: 10.1038/s41467-023-35843-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023] Open
Abstract
Cardiac valves ensure unidirectional blood flow through the heart, and altering their function can result in heart failure. Flow sensing via wall shear stress and wall stretching through the action of mechanosensors can modulate cardiac valve formation. However, the identity and precise role of the key mechanosensors and their effectors remain mostly unknown. Here, we genetically dissect the role of Pkd1a and other mechanosensors in atrioventricular (AV) valve formation in zebrafish and identify a role for several pkd and piezo gene family members in this process. We show that Pkd1a, together with Pkd2, Pkd1l1, and Piezo2a, promotes AV valve elongation and cardiac morphogenesis. Mechanistically, Pkd1a, Pkd2, and Pkd1l1 all repress the expression of klf2a and klf2b, transcription factor genes implicated in AV valve development. Furthermore, we find that the calcium-dependent protein kinase Camk2g is required downstream of Pkd function to repress klf2a expression. Altogether, these data identify, and dissect the role of, several mechanosensors required for AV valve formation, thereby broadening our understanding of cardiac valvulogenesis.
Collapse
Affiliation(s)
- Thomas Juan
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany. .,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany. .,Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.
| | - Agatha Ribeiro da Silva
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Bárbara Cardoso
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - SoEun Lim
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
| | - Violette Charteau
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany.,Institute for Molecules and Materials (IMM), Department of Biomolecular Chemistry, Radboud University, Nijmegen, The Netherlands
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany. .,German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany. .,Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.
| |
Collapse
|
21
|
Faubert AC, Larina IV, Wang S. Open-source, highly efficient, post-acquisition synchronization for 4D dual-contrast imaging of the mouse embryonic heart over development with optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2023; 14:163-181. [PMID: 36698661 PMCID: PMC9842004 DOI: 10.1364/boe.475027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 05/28/2023]
Abstract
Dynamic imaging of the beating embryonic heart in 3D is critical for understanding cardiac development and defects. Optical coherence tomography (OCT) plays an important role in embryonic heart imaging with its unique imaging scale and label-free contrasts. In particular, 4D (3D + time) OCT imaging enabled biomechanical analysis of the developing heart in various animal models. While ultrafast OCT systems allow for direct volumetric imaging of the beating heart, the imaging speed remains limited, leading to an image quality inferior to that produced by post-acquisition synchronization. As OCT systems become increasingly available to a wide range of biomedical researchers, a more accessible 4D reconstruction method is required to enable the broader application of OCT in the dynamic, volumetric assessment of embryonic heartbeat. Here, we report an open-source, highly efficient, post-acquisition synchronization method for 4D cardiodynamic and hemodynamic imaging of the mouse embryonic heart. Relying on the difference between images to characterize heart wall movements, the method provides good sensitivity to the cardiac activity when aligning heartbeat phases, even at early stages when the heart wall occupies only a small number of pixels. The method works with a densely sampled single 3D data acquisition, which, unlike the B-M scans required by other methods, is readily available in most commercial OCT systems. Compared with an existing approach for the mouse embryonic heart, this method shows superior reconstruction quality. We present the robustness of the method through results from different embryos with distinct heart rates, ranging from 1.24 Hz to 2.13 Hz. Since the alignment process operates on a 1D signal, the method has a high efficiency, featuring sub-second alignment time while utilizing ∼100% of the original image files. This allows us to achieve repeated, dual-contrast imaging of mouse embryonic heart development. This new, open-source method could facilitate research using OCT to study early cardiogenesis.
Collapse
Affiliation(s)
- Andre C. Faubert
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Irina V. Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shang Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
22
|
Scully DM, Larina IV. Mouse embryo phenotyping with optical coherence tomography. Front Cell Dev Biol 2022; 10:1000237. [PMID: 36158219 PMCID: PMC9500480 DOI: 10.3389/fcell.2022.1000237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/26/2022] [Indexed: 01/25/2023] Open
Abstract
With the explosion of gene editing tools in recent years, there has been a much greater demand for mouse embryo phenotyping, and traditional methods such as histology and histochemistry experienced a methodological renaissance as they became the principal tools for phenotyping. However, it is important to explore alternative phenotyping options to maximize time and resources and implement volumetric structural analysis for enhanced investigation of phenotypes. Cardiovascular phenotyping, in particular, is important to perform in vivo due to the dramatic structural and functional changes that occur in heart development over relatively short periods of time. Optical coherence tomography (OCT) is one of the most exciting advanced imaging techniques emerging within the field of developmental biology, and this review provides a summary of how it is currently being implemented in mouse embryo investigations and phenotyping. This review aims to provide an understanding of the approaches used in optical coherence tomography and how they can be applied in embryology and developmental biology, with the overall aim of bridging the gap between biology and technology.
Collapse
|
23
|
Wang S, Larina IV. Following the Beat: Imaging the Valveless Pumping Function in the Early Embryonic Heart. J Cardiovasc Dev Dis 2022; 9:jcdd9080267. [PMID: 36005431 PMCID: PMC9409458 DOI: 10.3390/jcdd9080267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/26/2022] Open
Abstract
In vertebrates, the coordinated beat of the early heart tube drives cardiogenesis and supports embryonic growth. How the heart pumps at this valveless stage marks a fascinating problem that is of vital significance for understanding cardiac development and defects. The developing heart achieves its function at the same time as continuous and dramatic morphological changes, which in turn modify its pumping dynamics. The beauty of this muti-time-scale process also highlights its complexity that requires interdisciplinary approaches to study. High-resolution optical imaging, particularly fast, four-dimensional (4D) imaging, plays a critical role in revealing the process of pumping, instructing numerical modeling, and enabling biomechanical analyses. In this review, we aim to connect the investigation of valveless pumping mechanisms with the recent advancements in embryonic cardiodynamic imaging, facilitating interactions between these two areas of study, in hopes of encouraging and motivating innovative work to further understand the early heartbeat.
Collapse
Affiliation(s)
- Shang Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
- Correspondence:
| | - Irina V. Larina
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
24
|
Zou X, Zhang Y, Wang N, Shi J, Li Q, Hao W, Zhu W, Han W. HEG1 as a novel potential biomarker for the prognosis of lung adenocarcinoma. Cancer Med 2022; 12:3288-3298. [PMID: 35950222 PMCID: PMC9939152 DOI: 10.1002/cam4.5081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/15/2022] [Accepted: 07/03/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Heart development protein with EGF-like domains 1 (HEG1), generally related to angiogenesis and embryonic development, was reported to participate in the occurrence and progression of some tumors recently. However, the role of HEG1 in lung adenocarcinoma (LUAD) is unclear. PATIENTS AND METHODS To explore the effect of HEG1 on LUAD, GEPIA platform and UALCAN database, as well as Kaplan-Meier plotter were adopted to analyze the association of HEG1 with clinicopathological characteristics and survival outcomes for LUAD firstly. And then the HEG1 in LUAD tissues, blood and cell lines were detected by qRT-PCR, western blot, immunofluorescence, immunohistochemistry, and ELISA. Gene set enrichment analysis (GSEA) was conducted to identify pathways that might be affected by HEG1 in LUAD. RESULTS In this study, HEG1 in lung tissues and cell lines of LUAD were significantly downregulated compared to benign pulmonary disease tissues and alveolar epithelial cells (p < 0.05). Moreover, compared with other groups, patients with advanced tumor stage had lower HEG1 mRNA expression levels (p = 0.025), which were negatively correlated with Ki67 index in tumor tissues (r = -0.427, p = 0.033). On the other hand, the LUAD patients with lower HEG1 had shorter overall survival (OS) (HR = 0.51, 95% CI: 0.40-0.65, p < 0.001) according to Kaplan-Meier plotter. In addition, HEG1 in serum of LUAD patients was negatively associated with CEA (r = -0.636, p < 0.001). GSEA showed that HEG1 was enriched in various metabolic-related pathways, including glucose metabolism, lipid metabolism, and nucleotide metabolism signaling. CONCLUSIONS HEG1 was downregulated in LUAD patients and associated with poor prognosis, which indicating HEG1 may serve as a potential biomarker for diagnosis and prognosis of LUAD.
Collapse
Affiliation(s)
- Xin Zou
- Department of Pathology, Qingdao Municipal HospitalDalian Medical UniversityQingdaoChina,Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina
| | - Yue Zhang
- Department of RespiratoryJilin Provincial People's HospitalJilinChina
| | - Ning Wang
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Jie Shi
- NMPA Key Laboratory for Quality Research and Evaluation of Traditional Marine Chinese MedicineQingdaoChina
| | - Qinghai Li
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Wanming Hao
- Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| | - Wenjing Zhu
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,NMPA Key Laboratory for Quality Research and Evaluation of Traditional Marine Chinese MedicineQingdaoChina,Clinical Research Center, Qingdao Municipal HospitalQingdao UniversityQingdaoChina
| | - Wei Han
- Respiratory Disease Key Laboratory of QingdaoQingdao Municipal HospitalQingdaoChina,Department of Pulmonary and Critical Care Medicine, Qingdao Municipal Hospital, School of MedicineQingdao UniversityQingdaoChina
| |
Collapse
|
25
|
Gurung S, Restrepo NK, Chestnut B, Klimkaite L, Sumanas S. Single-cell transcriptomic analysis of vascular endothelial cells in zebrafish embryos. Sci Rep 2022; 12:13065. [PMID: 35906287 PMCID: PMC9338088 DOI: 10.1038/s41598-022-17127-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular endothelial cells exhibit substantial phenotypic and transcriptional heterogeneity which is established during early embryogenesis. However, the molecular mechanisms involved in establishing endothelial cell diversity are still not well understood. Zebrafish has emerged as an advantageous model to study vascular development. Despite its importance, the single-cell transcriptomic profile of vascular endothelial cells during zebrafish development is still missing. To address this, we applied single-cell RNA-sequencing (scRNA-seq) of vascular endothelial cells isolated from zebrafish embryos at the 24 hpf stage. Six distinct clusters or subclusters related to vascular endothelial cells were identified which include arterial, two venous, cranial, endocardial and endothelial progenitor cell subtypes. Furthermore, we validated our findings by characterizing novel markers for arterial, venous, and endocardial cells. We experimentally confirmed the presence of two transcriptionally different venous cell subtypes, demonstrating heterogeneity among venous endothelial cells at this early developmental stage. This dataset will be a valuable resource for future functional characterization of vascular endothelial cells and interrogation of molecular mechanisms involved in the establishment of their heterogeneity and cell-fate decisions.
Collapse
Affiliation(s)
- Suman Gurung
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA
| | - Nicole K Restrepo
- Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA
| | - Brendan Chestnut
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Laurita Klimkaite
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. .,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA. .,Department of Pathology and Cell Biology, USF Health Heart Institute, University of South Florida, 560 Channelside Dr, Tampa, FL, 33602, USA.
| |
Collapse
|
26
|
Vignes H, Vagena-Pantoula C, Vermot J. Mechanical control of tissue shape: Cell-extrinsic and -intrinsic mechanisms join forces to regulate morphogenesis. Semin Cell Dev Biol 2022; 130:45-55. [PMID: 35367121 DOI: 10.1016/j.semcdb.2022.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 11/30/2022]
Abstract
During vertebrate development, cells must proliferate, move, and differentiate to form complex shapes. Elucidating the mechanisms underlying the molecular and cellular processes involved in tissue morphogenesis is essential to understanding developmental programmes. Mechanical stimuli act as a major contributor of morphogenetic processes and impact on cell behaviours to regulate tissue shape and size. Specifically, cell extrinsic physical forces are translated into biochemical signals within cells, through the process of mechanotransduction, activating multiple mechanosensitive pathways and defining cell behaviours. Physical forces generated by tissue mechanics and the extracellular matrix are crucial to orchestrate tissue patterning and cell fate specification. At the cell scale, the actomyosin network generates the cellular tension behind the tissue mechanics involved in building tissue. Thus, understanding the role of physical forces during morphogenetic processes requires the consideration of the contribution of cell intrinsic and cell extrinsic influences. The recent development of multidisciplinary approaches, as well as major advances in genetics, microscopy, and force-probing tools, have been key to push this field forward. With this review, we aim to discuss recent work on how tissue shape can be controlled by mechanical forces by focusing specifically on vertebrate organogenesis. We consider the influences of mechanical forces by discussing the cell-intrinsic forces (such as cell tension and proliferation) and cell-extrinsic forces (such as substrate stiffness and flow forces). We review recently described processes supporting the role of intratissue force generation and propagation in the context of shape emergence. Lastly, we discuss the emerging role of tissue-scale changes in tissue material properties, extrinsic forces, and shear stress on shape establishment.
Collapse
Affiliation(s)
- Hélène Vignes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France
| | | | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France; Department of Bioengineering, Imperial College London, London, United Kingdom.
| |
Collapse
|
27
|
Tsaryk R, Yucel N, Leonard EV, Diaz N, Bondareva O, Odenthal-Schnittler M, Arany Z, Vaquerizas JM, Schnittler H, Siekmann AF. Shear stress switches the association of endothelial enhancers from ETV/ETS to KLF transcription factor binding sites. Sci Rep 2022; 12:4795. [PMID: 35314737 PMCID: PMC8938417 DOI: 10.1038/s41598-022-08645-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Endothelial cells (ECs) lining blood vessels are exposed to mechanical forces, such as shear stress. These forces control many aspects of EC biology, including vascular tone, cell migration and proliferation. Despite a good understanding of the genes responding to shear stress, our insight into the transcriptional regulation of these genes is much more limited. Here, we set out to study alterations in the chromatin landscape of human umbilical vein endothelial cells (HUVEC) exposed to laminar shear stress. To do so, we performed ChIP-Seq for H3K27 acetylation, indicative of active enhancer elements and ATAC-Seq to mark regions of open chromatin in addition to RNA-Seq on HUVEC exposed to 6 h of laminar shear stress. Our results show a correlation of gained and lost enhancers with up and downregulated genes, respectively. DNA motif analysis revealed an over-representation of KLF transcription factor (TF) binding sites in gained enhancers, while lost enhancers contained more ETV/ETS motifs. We validated a subset of flow responsive enhancers using luciferase-based reporter constructs and CRISPR-Cas9 mediated genome editing. Lastly, we characterized the shear stress response in ECs of zebrafish embryos using RNA-Seq. Our results lay the groundwork for the exploration of shear stress responsive elements in controlling EC biology.
Collapse
Affiliation(s)
- Roman Tsaryk
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nora Yucel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Elvin V Leonard
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Noelia Diaz
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Olga Bondareva
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Philipp-Rosenthal-Str. 27, 04103, Leipzig, Germany
| | - Maria Odenthal-Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Juan M Vaquerizas
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Hans Schnittler
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
- Institute of Anatomy and Vascular Biology, Faculty of Medicine, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
- Institute of Neuropathology, Westfälische Wilhelms-Universität Münster, Pottkamp 2, 48149, Münster, Germany
| | - Arndt F Siekmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, 48149, Münster, Germany.
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany.
- Department of Cell and Developmental Biology and Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Endothelial MEKK3-KLF2/4 signaling integrates inflammatory and hemodynamic signals during definitive hematopoiesis. Blood 2022; 139:2942-2957. [PMID: 35245372 PMCID: PMC9101247 DOI: 10.1182/blood.2021013934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/11/2022] [Indexed: 11/20/2022] Open
Abstract
The hematopoietic stem cells (HSCs) that produce blood for the lifetime of an animal arise from RUNX1+ hemogenic endothelial cells (HECs) in the embryonic vasculature through a process of endothelial-to-hematopoietic transition (EHT). Studies have identified inflammatory mediators and fluid shear forces as critical environmental stimuli for EHT, raising the question of how such diverse inputs are integrated to drive HEC specification. Endothelial cell MEKK3-KLF2/4 signaling can be activated by both fluid shear forces and inflammatory mediators, and plays roles in cardiovascular development and disease that have been linked to both stimuli. Here we demonstrate that MEKK3 and KLF2/4 are required in endothelial cells for the specification of RUNX1+ HECs in both the yolk sac and dorsal aorta of the mouse embryo and for their transition to intra-aortic hematopoietic cluster cells (IAHCs). The inflammatory mediators lipopolysaccharide and interferon gamma increase RUNX1+ HECs in an MEKK3-dependent manner. Maternal administration of catecholamines that stimulate embryo cardiac function and accelerate yolk sac vascular remodeling increases EHT by wild-type but not MEKK3-deficient endothelium. These findings identify MEKK-KLF2/4 signaling as an essential pathway for EHT and provide a molecular basis for the integration of diverse environmental inputs, such as inflammatory mediators and hemodynamic forces, during definitive hematopoiesis.
Collapse
|
29
|
Vignes H, Vagena-Pantoula C, Prakash M, Fukui H, Norden C, Mochizuki N, Jug F, Vermot J. Extracellular mechanical forces drive endocardial cell volume decrease during zebrafish cardiac valve morphogenesis. Dev Cell 2022; 57:598-609.e5. [PMID: 35245444 DOI: 10.1016/j.devcel.2022.02.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/09/2021] [Accepted: 02/08/2022] [Indexed: 01/11/2023]
Abstract
Organ morphogenesis involves dynamic changes of tissue properties while cells adapt to their mechanical environment through mechanosensitive pathways. How mechanical cues influence cell behaviors during morphogenesis remains unclear. Here, we studied the formation of the zebrafish atrioventricular canal (AVC) where cardiac valves develop. We show that the AVC forms within a zone of tissue convergence associated with the increased activation of the actomyosin meshwork and cell-orientation changes. We demonstrate that tissue convergence occurs with a reduction of cell volume triggered by mechanical forces and the mechanosensitive channel TRPP2/TRPV4. Finally, we show that the extracellular matrix component hyaluronic acid controls cell volume changes. Together, our data suggest that multiple force-sensitive signaling pathways converge to modulate cell volume. We conclude that cell volume reduction is a key cellular feature activated by mechanotransduction during cardiovascular morphogenesis. This work further identifies how mechanical forces and extracellular matrix influence tissue remodeling in developing organs.
Collapse
Affiliation(s)
- Hélène Vignes
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Strasbourg, Illkirch, France
| | | | - Mangal Prakash
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Caren Norden
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Florian Jug
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany; Fondazione Human Technopole, Milan, Italy
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Strasbourg, Illkirch, France; Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
30
|
Cardiac forces regulate zebrafish heart valve delamination by modulating Nfat signaling. PLoS Biol 2022; 20:e3001505. [PMID: 35030171 PMCID: PMC8794269 DOI: 10.1371/journal.pbio.3001505] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/27/2022] [Accepted: 12/06/2021] [Indexed: 11/30/2022] Open
Abstract
In the clinic, most cases of congenital heart valve defects are thought to arise through errors that occur after the endothelial–mesenchymal transition (EndoMT) stage of valve development. Although mechanical forces caused by heartbeat are essential modulators of cardiovascular development, their role in these later developmental events is poorly understood. To address this question, we used the zebrafish superior atrioventricular valve (AV) as a model. We found that cellularized cushions of the superior atrioventricular canal (AVC) morph into valve leaflets via mesenchymal–endothelial transition (MEndoT) and tissue sheet delamination. Defects in delamination result in thickened, hyperplastic valves, and reduced heart function. Mechanical, chemical, and genetic perturbation of cardiac forces showed that mechanical stimuli are important regulators of valve delamination. Mechanistically, we show that forces modulate Nfatc activity to control delamination. Together, our results establish the cellular and molecular signature of cardiac valve delamination in vivo and demonstrate the continuous regulatory role of mechanical forces and blood flow during valve formation. Why do developing zebrafish atrioventricular heart valves become hyperplastic under certain hemodynamic conditions? This study suggests that part of the answer lies in how the mechanosensitive Nfat pathway regulates the valve mesenchymal-to-endothelial transition.
Collapse
|
31
|
Myocardial Afterload Is a Key Biomechanical Regulator of Atrioventricular Myocyte Differentiation in Zebrafish. J Cardiovasc Dev Dis 2022; 9:jcdd9010022. [PMID: 35050232 PMCID: PMC8779957 DOI: 10.3390/jcdd9010022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
Heart valve development is governed by both genetic and biomechanical inputs. Prior work has demonstrated that oscillating shear stress associated with blood flow is required for normal atrioventricular (AV) valve development. Cardiac afterload is defined as the pressure the ventricle must overcome in order to pump blood throughout the circulatory system. In human patients, conditions of high afterload can cause valve pathology. Whether high afterload adversely affects embryonic valve development remains poorly understood. Here we describe a zebrafish model exhibiting increased myocardial afterload, caused by vasopressin, a vasoconstrictive drug. We show that the application of vasopressin reliably produces an increase in afterload without directly acting on cardiac tissue in zebrafish embryos. We have found that increased afterload alters the rate of growth of the cardiac chambers and causes remodeling of cardiomyocytes. Consistent with pathology seen in patients with clinically high afterload, we see defects in both the form and the function of the valve leaflets. Our results suggest that valve defects are due to changes in atrioventricular myocyte signaling, rather than pressure directly acting on the endothelial valve leaflet cells. Cardiac afterload should therefore be considered a biomechanical factor that particularly impacts embryonic valve development.
Collapse
|
32
|
Deb N, Lacerda CMR. Valvular Endothelial Cell Response to the Mechanical Environment-A Review. Cell Biochem Biophys 2021; 79:695-709. [PMID: 34661855 DOI: 10.1007/s12013-021-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 10/02/2021] [Indexed: 01/08/2023]
Abstract
Heart valve leaflets are complex structures containing valve endothelial cells, interstitial cells, and extracellular matrix. Heart valve endothelial cells sense mechanical stimuli, and communicate amongst themselves and the surrounding cells and extracellular matrix to maintain tissue homeostasis. In the presence of abnormal mechanical stimuli, endothelial cell communication is triggered in defense and such processes may eventually lead to cardiac disease progression. This review focuses on the role of mechanical stimuli on heart valve endothelial surfaces-from heart valve development and maintenance of tissue integrity to disease progression with related signal pathways involved in this process.
Collapse
Affiliation(s)
- Nandini Deb
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US
| | - Carla M R Lacerda
- Jasper Department of Chemical Engineering, The University of Texas at Tyler, 3900 University Blvd, Tyler, 75799, TX, US.
| |
Collapse
|
33
|
Luo H, Zhang Y, Deng Y, Li L, Sheng Z, Yu Y, Lin Y, Chen X, Feng P. Nxhl Controls Angiogenesis by Targeting VE-PTP Through Interaction With Nucleolin. Front Cell Dev Biol 2021; 9:728821. [PMID: 34733844 PMCID: PMC8558974 DOI: 10.3389/fcell.2021.728821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Precise regulation of angiogenesis is required for organ development, wound repair, and tumor progression. Here, we identified a novel gene, nxhl (New XingHuo light), that is conserved in vertebrates and that plays a crucial role in vascular integrity and angiogenesis. Bioinformatic analysis uncovered its essential roles in development based on co-expression with several key developmental genes. Knockdown of nxhl in zebrafish causes global and pericardial edema, loss of blood circulation, and vascular defects characterized by both reduced vascularization in intersegmental vessels and decreased sprouting in the caudal vein plexus. The nxhl gene also affects human endothelial cell behavior in vitro. We found that nxhl functions in part by targeting VE-PTP through interaction with NCL (nucleolin). Loss of ptprb (a VE-PTP ortholo) in zebrafish resulted in defects similar to nxhl knockdown. Moreover, nxhl deficiency attenuates tumor invasion and proteins (including VE-PTP and NCL) associated with angiogenesis and EMT. These findings illustrate that nxhl can regulate angiogenesis via a novel nxhl-NCL-VE-PTP axis, providing a new therapeutic target for modulating vascular formation and function, especially for cancer treatment.
Collapse
Affiliation(s)
- Honglin Luo
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Yongde Zhang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yanfei Deng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoan Sheng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanling Yu
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yong Lin
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Xiaohan Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Pengfei Feng
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| |
Collapse
|
34
|
Shi WJ, Ma DD, Fang GZ, Zhang JG, Huang GY, Xie L, Chen HX, Hou LP, Ying GG. Levonorgestrel and dydrogesterone affect sex determination via different pathways in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105972. [PMID: 34571414 DOI: 10.1016/j.aquatox.2021.105972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
Levonorgestrel (LNG) and dydrogesterone (DDG) are two commonly used synthetic progestins that have been detected in aquatic environments. They could affect fish sex differentiation, but the underlying mechanisms remain unknown. Here we investigated the effects of LNG (5 ng L-1 and 50 ng L-1), DDG (100 ng L-1) and their mixtures on gonadal differentiation and sex determination in zebrafish at transcriptomic and histological levels from 2 hours post-fertilization (eleutheroembryos) to 144 days post-fertilization (sexual maturity). Germ cell development and oogenesis pathways were significantly enriched in LNG and the mixture of LNG and DDG treatments, while insulin and apoptosis pathways in the DDG treatment. LNG and the mixture of LNG and DDG strongly decreased transcripts of germ cell development and oogenesis related genes, while DDG increased the transcripts of insulin and apoptosis related genes at 28 days post fertilization (dpf) and 35 dpf. Furthermore, DDG caused ∼ 90% males, and LNG and the mixture of LNG and DDG resulted in 100% males on all sampling dates. Specifically, most males in LNG and the mixture of LNG and DDG treatments were "Type I" males without juvenile oocytes at 28 dpf and 35 dpf, while those in DDG treatment were "Type II" and "Type III" males with a few juvenile oocytes. These results indicated that LNG and DDG promoted testicular differentiation via different pathways to cause male bias. LNG and DDG mixtures have similar effect on testicular differentiation to LNG alone. The findings from this study could have significant ecological implications to fish populations.
Collapse
Affiliation(s)
- Wen-Jun Shi
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Dong-Dong Ma
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Gui-Zhen Fang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Jin-Ge Zhang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Guo-Yong Huang
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| | - Lingtian Xie
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Hong-Xing Chen
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China
| | - Li-Ping Hou
- School of Life Sciences, Guangzhou University, Guangzhou, 510655, China
| | - Guang-Guo Ying
- SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou 510006, China; School of Environment, South China Normal University, University Town, Guangzhou 510006, China.
| |
Collapse
|
35
|
Fukui H, Chow RWY, Xie J, Foo YY, Yap CH, Minc N, Mochizuki N, Vermot J. Bioelectric signaling and the control of cardiac cell identity in response to mechanical forces. Science 2021; 374:351-354. [PMID: 34648325 DOI: 10.1126/science.abc6229] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hajime Fukui
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France
| | - Jing Xie
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Yoke Yin Foo
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Choon Hwai Yap
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Department of Bioengineering, Imperial College London, London, UK
| | - Nicolas Minc
- Université de Paris, Centre National de la Recherche Scientifique UMR7592, Institut Jacques Monod, Paris, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U1258 and Université de Strasbourg, Illkirch, France.,Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
36
|
Derrick CJ, Pollitt EJG, Sanchez Sevilla Uruchurtu A, Hussein F, Grierson AJ, Noël ES. Lamb1a regulates atrial growth by limiting second heart field addition during zebrafish heart development. Development 2021; 148:272294. [PMID: 34568948 DOI: 10.1242/dev.199691] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
During early vertebrate heart development, the heart transitions from a linear tube to a complex asymmetric structure, a morphogenetic process that occurs simultaneously with growth of the heart. Cardiac growth during early heart morphogenesis is driven by deployment of cells from the second heart field (SHF) into both poles of the heart. Laminin is a core component of the extracellular matrix and, although mutations in laminin subunits are linked with cardiac abnormalities, no role for laminin has been identified in early vertebrate heart morphogenesis. We identified tissue-specific expression of laminin genes in the developing zebrafish heart, supporting a role for laminins in heart morphogenesis. Analysis of heart development in lamb1a zebrafish mutant embryos reveals mild morphogenetic defects and progressive cardiomegaly, and that Lamb1a functions to limit heart size during cardiac development by restricting SHF addition. lamb1a mutants exhibit hallmarks of altered haemodynamics, and blocking cardiac contractility in lamb1a mutants rescues heart size and atrial SHF addition. Together, these results suggest that laminin mediates interactions between SHF deployment and cardiac biomechanics during heart morphogenesis and growth in the developing embryo.
Collapse
Affiliation(s)
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
37
|
Paolini A, Fontana F, Pham VC, Rödel CJ, Abdelilah-Seyfried S. Mechanosensitive Notch-Dll4 and Klf2-Wnt9 signaling pathways intersect in guiding valvulogenesis in zebrafish. Cell Rep 2021; 37:109782. [PMID: 34610316 PMCID: PMC8511505 DOI: 10.1016/j.celrep.2021.109782] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 08/11/2021] [Accepted: 09/10/2021] [Indexed: 12/27/2022] Open
Abstract
In the zebrafish embryo, the onset of blood flow generates fluid shear stress on endocardial cells, which are specialized endothelial cells that line the interior of the heart. High levels of fluid shear stress activate both Notch and Klf2 signaling, which play crucial roles in atrioventricular valvulogenesis. However, it remains unclear why only individual endocardial cells ingress into the cardiac jelly and initiate valvulogenesis. Here, we show that lateral inhibition between endocardial cells, mediated by Notch, singles out Delta-like-4-positive endocardial cells. These cells ingress into the cardiac jelly, where they form an abluminal cell population. Delta-like-4-positive cells ingress in response to Wnt9a, which is produced in parallel through an Erk5-Klf2-Wnt9a signaling cascade also activated by blood flow. Hence, mechanical stimulation activates parallel mechanosensitive signaling pathways that produce binary effects by driving endocardial cells toward either luminal or abluminal fates. Ultimately, these cell fate decisions sculpt cardiac valve leaflets.
Collapse
Affiliation(s)
- Alessio Paolini
- Institute of Biochemistry and Biology, Potsdam University, 14476 Potsdam, Germany
| | - Federica Fontana
- Institute of Biochemistry and Biology, Potsdam University, 14476 Potsdam, Germany
| | - Van-Cuong Pham
- Institute of Biochemistry and Biology, Potsdam University, 14476 Potsdam, Germany
| | - Claudia Jasmin Rödel
- Institute of Biochemistry and Biology, Potsdam University, 14476 Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, 14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
38
|
Chakraborty S, Allmon E, Sepúlveda MS, Vlachos PP. Haemodynamic dependence of mechano-genetic evolution of the cardiovascular system in Japanese medaka. J R Soc Interface 2021; 18:20210752. [PMID: 34699728 PMCID: PMC8548083 DOI: 10.1098/rsif.2021.0752] [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: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 11/12/2022] Open
Abstract
The progression of cardiac gene expression-wall shear stress (WSS) interplay is critical to identifying developmental defects during cardiovascular morphogenesis. However, mechano-genetics from the embryonic to larval stages are poorly understood in vertebrates. We quantified peak WSS in the heart and tail vessels of Japanese medaka from 3 days post fertilization (dpf) to 14 dpf using in vivo micro-particle image velocimetry flow measurements, and in parallel analysed the expression of five cardiac genes (fgf8, hoxb6b, bmp4, nkx2.5, smyd1). Here, we report that WSS in the atrioventricular canal (AVC), ventricular outflow tract (OFT), and the caudal vessels in medaka peak with inflection points at 6 dpf and 10-11 dpf instead of a monotonic trend. Retrograde flows are captured at the AVC and OFT of the medaka heart for the first time. In addition, all genes were upregulated at 3 dpf and 7 dpf, indicating a possible correlation between the two, with the cardiac gene upregulation preceding WSS increase in order to facilitate cardiac wall remodelling.
Collapse
Affiliation(s)
- Sreyashi Chakraborty
- Department of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Elizabeth Allmon
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Maria S. Sepúlveda
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Pavlos P. Vlachos
- Department of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
39
|
Li J, Fang Y, Wu D. Mechanical forces and metabolic changes cooperate to drive cellular memory and endothelial phenotypes. CURRENT TOPICS IN MEMBRANES 2021; 87:199-253. [PMID: 34696886 PMCID: PMC8639155 DOI: 10.1016/bs.ctm.2021.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Endothelial cells line the innermost layer of arterial, venous, and lymphatic vascular tree and accordingly are subject to hemodynamic, stretch, and stiffness mechanical forces. Normally quiescent, endothelial cells have a hemodynamic set point and become "activated" in response to disturbed hemodynamics, which may signal impending nutrient or gas depletion. Endothelial cells in the majority of tissue beds are normally inactivated and maintain vessel barrier functions, are anti-inflammatory, anti-coagulant, and anti-thrombotic. However, under aberrant mechanical forces, endothelial signaling transforms in response, resulting cellular changes that herald pathological diseases. Endothelial cell metabolism is now recognized as the primary intermediate pathway that undergirds cellular transformation. In this review, we discuss the various mechanical forces endothelial cells sense in the large vessels, microvasculature, and lymphatics, and how changes in environmental mechanical forces result in changes in metabolism, which ultimately influence cell physiology, cellular memory, and ultimately disease initiation and progression.
Collapse
Affiliation(s)
- Jin Li
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - Yun Fang
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States
| | - David Wu
- Committee on Molecular Metabolism and Nutrition, Biological Sciences Division, University of Chicago, Chicago, IL, United States; Department of Medicine, Biological Sciences Division, University of Chicago, Chicago, IL, United States.
| |
Collapse
|
40
|
Hypoplastic left heart syndrome (HLHS): molecular pathogenesis and emerging drug targets for cardiac repair and regeneration. Expert Opin Ther Targets 2021; 25:621-632. [PMID: 34488532 DOI: 10.1080/14728222.2021.1978069] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Hypoplastic left heart syndrome (HLHS) is a severe developmental defect characterized by the underdevelopment of the left ventricle along with aortic and valvular defects. Multiple palliative surgeries are required for survival. Emerging studies have identified potential mechanisms for the disease onset, including genetic and hemodynamic causes. Genetic variants associated with HLHS include transcription factors, chromatin remodelers, structural proteins, and signaling proteins necessary for normal heart development. Nonetheless, current therapies are being tested clinically and have shown promising results at improving cardiac function in patients who have undergone palliative surgeries. AREAS COVERED We searched PubMed and clinicaltrials.gov to review most of the mechanistic research and clinical trials involving HLHS. This review discusses the anatomy and pathology of HLHS hearts. We highlight some of the identified genetic variants that underly the molecular pathogenesis of HLHS. Additionally, we discuss some of the emerging therapies and their limitations for HLHS. EXPERT OPINION While HLHS etiology is largely obscure, palliative therapies remain the most viable option for the patients. It is necessary to generate animal and stem cell models to understand the underlying genetic causes directly leading to HLHS and facilitate the use of gene-based therapies to improve cardiac development and regeneration.
Collapse
|
41
|
Bornhorst D, Abdelilah-Seyfried S. Strong as a Hippo's Heart: Biomechanical Hippo Signaling During Zebrafish Cardiac Development. Front Cell Dev Biol 2021; 9:731101. [PMID: 34422841 PMCID: PMC8375320 DOI: 10.3389/fcell.2021.731101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The heart is comprised of multiple tissues that contribute to its physiological functions. During development, the growth of myocardium and endocardium is coupled and morphogenetic processes within these separate tissue layers are integrated. Here, we discuss the roles of mechanosensitive Hippo signaling in growth and morphogenesis of the zebrafish heart. Hippo signaling is involved in defining numbers of cardiac progenitor cells derived from the secondary heart field, in restricting the growth of the epicardium, and in guiding trabeculation and outflow tract formation. Recent work also shows that myocardial chamber dimensions serve as a blueprint for Hippo signaling-dependent growth of the endocardium. Evidently, Hippo pathway components act at the crossroads of various signaling pathways involved in embryonic zebrafish heart development. Elucidating how biomechanical Hippo signaling guides heart morphogenesis has direct implications for our understanding of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Stem Cell Program, Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
42
|
Ling S, Jenkins MW, Watanabe M, Ford SM, Rollins AM. Prenatal ethanol exposure impairs the conduction delay at the atrioventricular junction in the looping heart. Am J Physiol Heart Circ Physiol 2021; 321:H294-H305. [PMID: 34142884 PMCID: PMC8526336 DOI: 10.1152/ajpheart.00107.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
The etiology of ethanol-related congenital heart defects has been the focus of much study, but most research has concentrated on cellular and molecular mechanisms. We have shown with optical coherence tomography (OCT) that ethanol exposure led to increased retrograde flow and smaller atrioventricular (AV) cushions compared with controls. Since AV cushions play a role in patterning the conduction delay at the atrioventricular junction (AVJ), this study aims to investigate whether ethanol exposure alters the AVJ conduction in early looping hearts and whether this alteration is related to the decreased cushion size. Quail embryos were exposed to a single dose of ethanol at gastrulation, and Hamburger-Hamilton stage 19-20 hearts were dissected for imaging. Cardiac conduction was measured using an optical mapping microscope and we imaged the endocardial cushions using OCT. Our results showed that, compared with controls, ethanol-exposed embryos exhibited abnormally fast AVJ conduction and reduced cushion size. However, this increased conduction velocity (CV) did not strictly correlate with decreased cushion volume and thickness. By matching the CV map to the cushion-size map along the inflow heart tube, we found that the slowest conduction location was consistently at the atrial side of the AVJ, which had the thinner cushions, not at the thickest cushion location at the ventricular side as expected. Our findings reveal regional differences in the AVJ myocardium even at this early stage in heart development. These findings reveal the early steps leading to the heterogeneity and complexity of conduction at the mature AVJ, a site where arrhythmias can be initiated.NEW & NOTEWORTHY To the best of our knowledge, this is the first study investigating the impact of ethanol exposure on the early cardiac conduction system. Our results showed that ethanol-exposed embryos exhibited abnormally fast atrioventricular conduction. In addition, our findings, in CV measurements and endocardial cushion thickness, reveal regional differences in the AVJ myocardium even at this early stage in heart development, suggesting that the differentiation and maturation at this site are complex and warrant further studies.
Collapse
Affiliation(s)
- Shan Ling
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Michael W Jenkins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Michiko Watanabe
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Stephanie M Ford
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Pediatric Cardiology, The Congenital Heart Collaborative, Rainbow Babies and Children's Hospital, Cleveland, Ohio
- Division of Neonatology, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - Andrew M Rollins
- Department of Biomedical Engineering, School of Engineering and School of Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
43
|
Wang Z, Ding Y, Satta S, Roustaei M, Fei P, Hsiai TK. A hybrid of light-field and light-sheet imaging to study myocardial function and intracardiac blood flow during zebrafish development. PLoS Comput Biol 2021; 17:e1009175. [PMID: 34228702 PMCID: PMC8284633 DOI: 10.1371/journal.pcbi.1009175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 07/16/2021] [Accepted: 06/11/2021] [Indexed: 01/07/2023] Open
Abstract
Biomechanical forces intimately contribute to cardiac morphogenesis. However, volumetric imaging to investigate the cardiac mechanics with high temporal and spatial resolution remains an imaging challenge. We hereby integrated light-field microscopy (LFM) with light-sheet fluorescence microscopy (LSFM), coupled with a retrospective gating method, to simultaneously access myocardial contraction and intracardiac blood flow at 200 volumes per second. While LSFM allows for the reconstruction of the myocardial function, LFM enables instantaneous acquisition of the intracardiac blood cells traversing across the valves. We further adopted deformable image registration to quantify the ventricular wall displacement and particle tracking velocimetry to monitor intracardiac blood flow. The integration of LFM and LSFM enabled the time-dependent tracking of the individual blood cells and the differential rates of segmental wall displacement during a cardiac cycle. Taken together, we demonstrated a hybrid system, coupled with our image analysis pipeline, to simultaneously capture the myocardial wall motion with intracardiac blood flow during cardiac development. During the conception of the heart, cardiac muscular contraction and blood flow generate biomechanical forces to influence the functional and structural development. To elucidate the underlying biomechanical mechanisms, we have embraced the zebrafish system for the ease of genetic and pharmacological manipulations and its rapidity for organ development. However, acquiring the dynamic processes (space + time domain) in the small beating zebrafish heart remains a challenge. In the presence of a rapid heartbeat, microscopy is confined by temporal resolution to image the cardiac contraction and blood flow. In this context, we demonstrated an integrated light-sheet and light-field imaging system to visualize cardiac contraction along with the flowing blood cells inside the cardiac chambers. Assuming the periodicity of the cardiac cycle, we synchronized the image data in post-processing for 3-D reconstruction. We further quantified the velocity of the various regions of cardiac muscular contraction, and tracked the individual blood cells during the cardiac cycles. The time-dependent velocity maps allow for uncovering differential segments of cardiac contraction and relaxation, and for revealing the patterns of blood flow. Thus, our integrated light-sheet and light-field imaging system provides an experimental basis to further investigate cardiac function and development.
Collapse
Affiliation(s)
- Zhaoqiang Wang
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Mehrdad Roustaei
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Peng Fei
- School of Optical and Electronic Information-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (PF); (TKH)
| | - Tzung K. Hsiai
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California, United States of America
- * E-mail: (PF); (TKH)
| |
Collapse
|
44
|
King O, Sunyovszki I, Terracciano CM. Vascularisation of pluripotent stem cell-derived myocardium: biomechanical insights for physiological relevance in cardiac tissue engineering. Pflugers Arch 2021; 473:1117-1136. [PMID: 33855631 PMCID: PMC8245389 DOI: 10.1007/s00424-021-02557-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/22/2022]
Abstract
The myocardium is a diverse environment, requiring coordination between a variety of specialised cell types. Biochemical crosstalk between cardiomyocytes (CM) and microvascular endothelial cells (MVEC) is essential to maintain contractility and healthy tissue homeostasis. Yet, as myocytes beat, heterocellular communication occurs also through constantly fluctuating biomechanical stimuli, namely (1) compressive and tensile forces generated directly by the beating myocardium, and (2) pulsatile shear stress caused by intra-microvascular flow. Despite endothelial cells (EC) being highly mechanosensitive, the role of biomechanical stimuli from beating CM as a regulatory mode of myocardial-microvascular crosstalk is relatively unexplored. Given that cardiac biomechanics are dramatically altered during disease, and disruption of myocardial-microvascular communication is a known driver of pathological remodelling, understanding the biomechanical context necessary for healthy myocardial-microvascular interaction is of high importance. The current gap in understanding can largely be attributed to technical limitations associated with reproducing dynamic physiological biomechanics in multicellular in vitro platforms, coupled with limited in vitro viability of primary cardiac tissue. However, differentiation of CM from human pluripotent stem cells (hPSC) has provided an unlimited source of human myocytes suitable for designing in vitro models. This technology is now converging with the diverse field of tissue engineering, which utilises in vitro techniques designed to enhance physiological relevance, such as biomimetic extracellular matrix (ECM) as 3D scaffolds, microfluidic perfusion of vascularised networks, and complex multicellular architectures generated via 3D bioprinting. These strategies are now allowing researchers to design in vitro platforms which emulate the cell composition, architectures, and biomechanics specific to the myocardial-microvascular microenvironment. Inclusion of physiological multicellularity and biomechanics may also induce a more mature phenotype in stem cell-derived CM, further enhancing their value. This review aims to highlight the importance of biomechanical stimuli as determinants of CM-EC crosstalk in cardiac health and disease, and to explore emerging tissue engineering and hPSC technologies which can recapitulate physiological dynamics to enhance the value of in vitro cardiac experimentation.
Collapse
Affiliation(s)
- Oisín King
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK.
| | - Ilona Sunyovszki
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| | - Cesare M Terracciano
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, ICTEM 4th floor, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
45
|
Francois M, Oszmiana A, Harvey NL. When form meets function: the cells and signals that shape the lymphatic vasculature during development. Development 2021; 148:268989. [PMID: 34080610 DOI: 10.1242/dev.167098] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The lymphatic vasculature is an integral component of the cardiovascular system. It is essential to maintain tissue fluid homeostasis, direct immune cell trafficking and absorb dietary lipids from the digestive tract. Major advances in our understanding of the genetic and cellular events important for constructing the lymphatic vasculature during development have recently been made. These include the identification of novel sources of lymphatic endothelial progenitor cells, the recognition of lymphatic endothelial cell specialisation and heterogeneity, and discovery of novel genes and signalling pathways underpinning developmental lymphangiogenesis. Here, we review these advances and discuss how they inform our understanding of lymphatic network formation, function and dysfunction.
Collapse
Affiliation(s)
- Mathias Francois
- The David Richmond Laboratory for Cardiovascular Development: Gene Regulation and Editing Program, The Centenary Institute, The University of Sydney, SOLES, 2006 Camperdown, Australia
| | - Anna Oszmiana
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5001, Australia
| | - Natasha L Harvey
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide 5001, Australia
| |
Collapse
|
46
|
Rödel CJ, Abdelilah-Seyfried S. A zebrafish toolbox for biomechanical signaling in cardiovascular development and disease. Curr Opin Hematol 2021; 28:198-207. [PMID: 33714969 DOI: 10.1097/moh.0000000000000648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The zebrafish embryo has emerged as a powerful model organism to investigate the mechanisms by which biophysical forces regulate vascular and cardiac cell biology during development and disease. A versatile arsenal of methods and tools is available to manipulate and analyze biomechanical signaling. This review aims to provide an overview of the experimental strategies and tools that have been utilized to study biomechanical signaling in cardiovascular developmental processes and different vascular disease models in the zebrafish embryo. Within the scope of this review, we focus on work published during the last two years. RECENT FINDINGS Genetic and pharmacological tools for the manipulation of cardiac function allow alterations of hemodynamic flow patterns in the zebrafish embryo and various types of transgenic lines are available to report endothelial cell responses to biophysical forces. These tools have not only revealed the impact of biophysical forces on cardiovascular development but also helped to establish more accurate models for cardiovascular diseases including cerebral cavernous malformations, hereditary hemorrhagic telangiectasias, arteriovenous malformations, and lymphangiopathies. SUMMARY The zebrafish embryo is a valuable vertebrate model in which in-vivo manipulations of biophysical forces due to cardiac contractility and blood flow can be performed. These analyses give important insights into biomechanical signaling pathways that control endothelial and endocardial cell behaviors. The technical advances using this vertebrate model will advance our understanding of the impact of biophysical forces in cardiovascular pathologies.
Collapse
Affiliation(s)
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
47
|
McGinn J, Hallou A, Han S, Krizic K, Ulyanchenko S, Iglesias-Bartolome R, England FJ, Verstreken C, Chalut KJ, Jensen KB, Simons BD, Alcolea MP. A biomechanical switch regulates the transition towards homeostasis in oesophageal epithelium. Nat Cell Biol 2021; 23:511-525. [PMID: 33972733 PMCID: PMC7611004 DOI: 10.1038/s41556-021-00679-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Epithelial cells rapidly adapt their behaviour in response to increasing tissue demands. However, the processes that finely control these cell decisions remain largely unknown. The postnatal period covering the transition between early tissue expansion and the establishment of adult homeostasis provides a convenient model with which to explore this question. Here, we demonstrate that the onset of homeostasis in the epithelium of the mouse oesophagus is guided by the progressive build-up of mechanical strain at the organ level. Single-cell RNA sequencing and whole-organ stretching experiments revealed that the mechanical stress experienced by the growing oesophagus triggers the emergence of a bright Krüppel-like factor 4 (KLF4) committed basal population, which balances cell proliferation and marks the transition towards homeostasis in a yes-associated protein (YAP)-dependent manner. Our results point to a simple mechanism whereby mechanical changes experienced at the whole-tissue level are integrated with those sensed at the cellular level to control epithelial cell fate.
Collapse
Affiliation(s)
- Jamie McGinn
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Oncology, University of Cambridge and Cancer Research UK Cambridge Centre, Cambridge, UK
| | - Adrien Hallou
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | - Seungmin Han
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Kata Krizic
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Svetlana Ulyanchenko
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ramiro Iglesias-Bartolome
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Frances J England
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Kevin J Chalut
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Kim B Jensen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Stem Cell Biology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Benjamin D Simons
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
| | - Maria P Alcolea
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Department of Oncology, University of Cambridge and Cancer Research UK Cambridge Centre, Cambridge, UK.
| |
Collapse
|
48
|
Fontana F, Haack T, Reichenbach M, Knaus P, Puceat M, Abdelilah-Seyfried S. Antagonistic Activities of Vegfr3/Flt4 and Notch1b Fine-tune Mechanosensitive Signaling during Zebrafish Cardiac Valvulogenesis. Cell Rep 2021; 32:107883. [PMID: 32668254 DOI: 10.1016/j.celrep.2020.107883] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 06/17/2020] [Indexed: 12/22/2022] Open
Abstract
The formation of cardiac valves depends on mechanical forces exerted by blood flow. Endocardial cells lining the interior of the heart are sensitive to these stimuli and respond by rearranging into luminal cells subjected to shear stress and abluminal cells not exposed to it. The mechanisms by which endocardial cells sense these dynamic biomechanical stimuli and how they evoke different cellular responses are largely unknown. Here, we show that blood flow activates two parallel mechanosensitive pathways, one mediated by Notch and the other by Klf2a. Both pathways negatively regulate the angiogenesis receptor Vegfr3/Flt4, which becomes restricted to abluminal endocardial cells. Its loss disrupts valve morphogenesis and results in the occurrence of Notch signaling within abluminal endocardial cells. Our work explains how antagonistic activities by Vegfr3/Flt4 on the abluminal side and by Notch on the luminal side shape cardiac valve leaflets by triggering unique differences in the fates of endocardial cells.
Collapse
Affiliation(s)
- Federica Fontana
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany
| | - Timm Haack
- Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany
| | - Maria Reichenbach
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Petra Knaus
- Institute of Biochemistry, Freie Universität Berlin, D-14195 Berlin, Germany
| | - Michel Puceat
- INSERM U-1251, MMG, Aix-Marseille University, 13885 Marseille, France
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, D-30625 Hannover, Germany.
| |
Collapse
|
49
|
Ford SM, Pedersen CJ, Ford MR, Kim JW, Karunamuni GH, McPheeters MT, Jawaid S, Jenkins MW, Rollins AM, Watanabe M. Folic acid prevents functional and structural heart defects induced by prenatal ethanol exposure. Am J Physiol Heart Circ Physiol 2021. [DOI: 10.1152/ajpheart.00817.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
State-of-the-art biophotonic tools captured blood flow and endocardial cushion volumes in tiny beating quail embryo hearts, an accessible model for studying four-chambered heart development. Both hemodynamic flow and endocardial cushion volumes were altered with ethanol exposure but normalized when folic acid was introduced with ethanol. Folic acid supplementation preserved hemodynamic function that is intimately involved in sculpting the heart from the earliest stages of heart development.
Collapse
Affiliation(s)
- Stephanie M. Ford
- Division of Neonatology, Department of Pediatrics, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Cameron J. Pedersen
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew R. Ford
- Department of Ophthalmology, Cole Eye Institute, Cleveland Clinic, Cleveland Ohio
| | - Jun W. Kim
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Ganga H. Karunamuni
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Matthew T. McPheeters
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Safdar Jawaid
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michael W. Jenkins
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Andrew M. Rollins
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Michiko Watanabe
- Division of Pediatric Cardiology, Department of Pediatrics, The Congenital Heart Collaborative, Rainbow Babies and Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, Ohio
| |
Collapse
|
50
|
Giardoglou P, Bournele D, Park M, Kanoni S, Dedoussis GV, Steinberg SF, Deloukas P, Beis D. A zebrafish forward genetic screen identifies an indispensable threonine residue in the kinase domain of PRKD2. Biol Open 2021; 10:bio.058542. [PMID: 33597201 PMCID: PMC7969590 DOI: 10.1242/bio.058542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Protein kinase D2 belongs to a family of evolutionarily conserved enzymes regulating several biological processes. In a forward genetic screen for zebrafish cardiovascular mutants, we identified a mutation in the prkd2 gene. Homozygous mutant embryos develop as wild type up to 36 h post-fertilization and initiate blood flow, but fail to maintain it, resulting in a complete outflow tract stenosis. We identified a mutation in the prkd2 gene that results in a T757A substitution at a conserved residue in the kinase domain activation loop (T714A in human PRKD2) that disrupts catalytic activity and drives this phenotype. Homozygous mutants survive without circulation for several days, allowing us to study the extreme phenotype of no intracardiac flow, in the background of a functional heart. We show dysregulation of atrioventricular and outflow tract markers in the mutants and higher sensitivity to the Calcineurin inhibitor, Cyclosporin A. Finally we identify TBX5 as a potential regulator of PRKD2. Our results implicate PRKD2 catalytic activity in outflow tract development in zebrafish. This article has an associated First Person interview with the first author of the paper. Summary: We identified, through a zebrafish forward screen, an evolutionarily conserved residue in the catalytic domain of protein kinase D2 and its homologues.
Collapse
Affiliation(s)
- Panagiota Giardoglou
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece.,Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Despina Bournele
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| | - Misun Park
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK
| | - George V Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens 176 71, Greece
| | - Susan F Steinberg
- Department of Pharmacology, Columbia University, New York 100 27, USA
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Clinical Pharmacology Centre, Queen Mary University of London, London, EC1M 6BQ, UK.,Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 222 52, Saudi Arabia
| | - Dimitris Beis
- Zebrafish Disease Model lab, Biomedical Research Foundation Academy of Athens, Athens 115 27, Greece
| |
Collapse
|