1
|
Li Y, Du J, Deng S, Liu B, Jing X, Yan Y, Liu Y, Wang J, Zhou X, She Q. The molecular mechanisms of cardiac development and related diseases. Signal Transduct Target Ther 2024; 9:368. [PMID: 39715759 DOI: 10.1038/s41392-024-02069-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiac development is a complex and intricate process involving numerous molecular signals and pathways. Researchers have explored cardiac development through a long journey, starting with early studies observing morphological changes and progressing to the exploration of molecular mechanisms using various molecular biology methods. Currently, advancements in stem cell technology and sequencing technology, such as the generation of human pluripotent stem cells and cardiac organoids, multi-omics sequencing, and artificial intelligence (AI) technology, have enabled researchers to understand the molecular mechanisms of cardiac development better. Many molecular signals regulate cardiac development, including various growth and transcription factors and signaling pathways, such as WNT signaling, retinoic acid signaling, and Notch signaling pathways. In addition, cilia, the extracellular matrix, epigenetic modifications, and hypoxia conditions also play important roles in cardiac development. These factors play crucial roles at one or even multiple stages of cardiac development. Recent studies have also identified roles for autophagy, metabolic transition, and macrophages in cardiac development. Deficiencies or abnormal expression of these factors can lead to various types of cardiac development abnormalities. Nowadays, congenital heart disease (CHD) management requires lifelong care, primarily involving surgical and pharmacological treatments. Advances in surgical techniques and the development of clinical genetic testing have enabled earlier diagnosis and treatment of CHD. However, these technologies still have significant limitations. The development of new technologies, such as sequencing and AI technologies, will help us better understand the molecular mechanisms of cardiac development and promote earlier prevention and treatment of CHD in the future.
Collapse
Affiliation(s)
- Yingrui Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Songbai Deng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Jing
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuling Yan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Germany; DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Mannheim, Germany
| | - Qiang She
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Jang J, Accornero F, Li D. Epigenetic determinants and non-myocardial signaling pathways contributing to heart growth and regeneration. Pharmacol Ther 2024; 257:108638. [PMID: 38548089 DOI: 10.1016/j.pharmthera.2024.108638] [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: 01/02/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Congenital heart disease is the most common birth defect worldwide. Defective cardiac myogenesis is either a major presentation or associated with many types of congenital heart disease. Non-myocardial tissues, including endocardium and epicardium, function as a supporting hub for myocardial growth and maturation during heart development. Recent research findings suggest an emerging role of epigenetics in nonmyocytes supporting myocardial development. Understanding how growth signaling pathways in non-myocardial tissues are regulated by epigenetic factors will likely identify new disease mechanisms for congenital heart diseases and shed lights for novel therapeutic strategies for heart regeneration.
Collapse
Affiliation(s)
- Jihyun Jang
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA.
| | - Federica Accornero
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
| | - Deqiang Li
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43215, USA.
| |
Collapse
|
3
|
Reimann MJ, Cremer S, Christiansen L, Ibragimov E, Gao F, Cirera S, Fredholm M, Olsen LH, Karlskov-Mortensen P. Mitral valve transcriptome analysis in thirty-four age-matched Cavalier King Charles Spaniels with or without congestive heart failure caused by myxomatous mitral valve disease. Mamm Genome 2024; 35:77-89. [PMID: 37938355 PMCID: PMC10884180 DOI: 10.1007/s00335-023-10024-1] [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: 01/09/2023] [Accepted: 10/08/2023] [Indexed: 11/09/2023]
Abstract
We here report the results of a mitral valve transcriptome study designed to identify genes and molecular pathways involved in development of congestive heart failure (CHF) following myxomatous mitral valve disease (MMVD) in dogs. The study is focused on a cohort of elderly age-matched dogs (n = 34, age ~ 10 years) from a single breed-Cavalier King Charles Spaniels (CKCS)-with a high incidence of MMVD. The cohort comprises 19 dogs (10♀, 9♂) without MMVD-associated CHF, and 15 dogs (6♀, 9♂) with CHF caused by MMVD; i.e., we compare gene expression in breed and age-matched groups of dogs, which only differ with respect to CHF status. We identify 56 genes, which are differentially expressed between the two groups. In this list of genes, we confirm an enrichment of genes related to the TNFβ-signaling pathway, extracellular matrix organization, vascular development, and endothelium damage, which also have been identified in previous studies. However, the genes with the greatest difference in expression between the two groups are CNTN3 and MYH1. Both genes encode proteins, which are predicted to have an effect on the contractile activity of myocardial cells, which in turn may have an effect on valvular performance and hemodynamics across the mitral valve. This may result in shear forces with impact on MMVD progression.
Collapse
Affiliation(s)
- Maria J Reimann
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Signe Cremer
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Liselotte Christiansen
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Emil Ibragimov
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Fei Gao
- Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Susanna Cirera
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Merete Fredholm
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Lisbeth H Olsen
- Preclinical Disease Biology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter Karlskov-Mortensen
- Animal Genetics and Breeding, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark.
| |
Collapse
|
4
|
Dark N, Cosson MV, Tsansizi LI, Owen TJ, Ferraro E, Francis AJ, Tsai S, Bouissou C, Weston A, Collinson L, Abi-Gerges N, Miller PE, MacLeod KT, Ehler E, Mitter R, Harding SE, Smith JC, Bernardo AS. Generation of left ventricle-like cardiomyocytes with improved structural, functional, and metabolic maturity from human pluripotent stem cells. CELL REPORTS METHODS 2023; 3:100456. [PMID: 37159667 PMCID: PMC10163040 DOI: 10.1016/j.crmeth.2023.100456] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/23/2023] [Accepted: 03/25/2023] [Indexed: 05/11/2023]
Abstract
Decreased left ventricle (LV) function caused by genetic mutations or injury often leads to debilitating and fatal cardiovascular disease. LV cardiomyocytes are, therefore, a potentially valuable therapeutical target. Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are neither homogeneous nor functionally mature, which reduces their utility. Here, we exploit cardiac development knowledge to instruct differentiation of hPSCs specifically toward LV cardiomyocytes. Correct mesoderm patterning and retinoic acid pathway blocking are essential to generate near-homogenous LV-specific hPSC-CMs (hPSC-LV-CMs). These cells transit via first heart field progenitors and display typical ventricular action potentials. Importantly, hPSC-LV-CMs exhibit increased metabolism, reduced proliferation, and improved cytoarchitecture and functional maturity compared with age-matched cardiomyocytes generated using the standard WNT-ON/WNT-OFF protocol. Similarly, engineered heart tissues made from hPSC-LV-CMs are better organized, produce higher force, and beat more slowly but can be paced to physiological levels. Together, we show that functionally matured hPSC-LV-CMs can be obtained rapidly without exposure to current maturation regimes.
Collapse
Affiliation(s)
| | | | - Lorenza I. Tsansizi
- The Francis Crick Institute, London, UK
- NHLI, Imperial College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andreia S. Bernardo
- The Francis Crick Institute, London, UK
- NHLI, Imperial College London, London, UK
| |
Collapse
|
5
|
Young WJ, Haessler J, Benjamins JW, Repetto L, Yao J, Isaacs A, Harper AR, Ramirez J, Garnier S, van Duijvenboden S, Baldassari AR, Concas MP, Duong T, Foco L, Isaksen JL, Mei H, Noordam R, Nursyifa C, Richmond A, Santolalla ML, Sitlani CM, Soroush N, Thériault S, Trompet S, Aeschbacher S, Ahmadizar F, Alonso A, Brody JA, Campbell A, Correa A, Darbar D, De Luca A, Deleuze JF, Ellervik C, Fuchsberger C, Goel A, Grace C, Guo X, Hansen T, Heckbert SR, Jackson RD, Kors JA, Lima-Costa MF, Linneberg A, Macfarlane PW, Morrison AC, Navarro P, Porteous DJ, Pramstaller PP, Reiner AP, Risch L, Schotten U, Shen X, Sinagra G, Soliman EZ, Stoll M, Tarazona-Santos E, Tinker A, Trajanoska K, Villard E, Warren HR, Whitsel EA, Wiggins KL, Arking DE, Avery CL, Conen D, Girotto G, Grarup N, Hayward C, Jukema JW, Mook-Kanamori DO, Olesen MS, Padmanabhan S, Psaty BM, Pattaro C, Ribeiro ALP, Rotter JI, Stricker BH, van der Harst P, van Duijn CM, Verweij N, Wilson JG, Orini M, Charron P, Watkins H, Kooperberg C, Lin HJ, Wilson JF, Kanters JK, Sotoodehnia N, Mifsud B, Lambiase PD, Tereshchenko LG, Munroe PB. Genetic architecture of spatial electrical biomarkers for cardiac arrhythmia and relationship with cardiovascular disease. Nat Commun 2023; 14:1411. [PMID: 36918541 PMCID: PMC10015012 DOI: 10.1038/s41467-023-36997-w] [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: 05/27/2022] [Accepted: 02/26/2023] [Indexed: 03/15/2023] Open
Abstract
The 3-dimensional spatial and 2-dimensional frontal QRS-T angles are measures derived from the vectorcardiogram. They are independent risk predictors for arrhythmia, but the underlying biology is unknown. Using multi-ancestry genome-wide association studies we identify 61 (58 previously unreported) loci for the spatial QRS-T angle (N = 118,780) and 11 for the frontal QRS-T angle (N = 159,715). Seven out of the 61 spatial QRS-T angle loci have not been reported for other electrocardiographic measures. Enrichments are observed in pathways related to cardiac and vascular development, muscle contraction, and hypertrophy. Pairwise genome-wide association studies with classical ECG traits identify shared genetic influences with PR interval and QRS duration. Phenome-wide scanning indicate associations with atrial fibrillation, atrioventricular block and arterial embolism and genetically determined QRS-T angle measures are associated with fascicular and bundle branch block (and also atrioventricular block for the frontal QRS-T angle). We identify potential biology involved in the QRS-T angle and their genetic relationships with cardiovascular traits and diseases, may inform future research and risk prediction.
Collapse
Affiliation(s)
- William J Young
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jan-Walter Benjamins
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - Linda Repetto
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Aaron Isaacs
- Dept. of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Andrew R Harper
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Julia Ramirez
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
- Aragon Institute of Engineering Research, University of Zaragoza, Zaragoza, Spain and Center of Biomedical Research Network, Bioengineering, Biomaterials and Nanomedicine, Zaragoza, Spain
| | - Sophie Garnier
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
| | - Stefan van Duijvenboden
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Antoine R Baldassari
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria Pina Concas
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luisa Foco
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
| | - Jonas L Isaksen
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Raymond Noordam
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Casia Nursyifa
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Meddly L Santolalla
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Emerge, Emerging Diseases and Climate Change Research Unit, School of Public Health and Administration, Universidad Peruana Cayetano Heredia, Lima, 15152, Peru
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Negin Soroush
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sébastien Thériault
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Quebec, QC, Canada
| | - Stella Trompet
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Stefanie Aeschbacher
- Cardiovascular Research Institute Basel, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
- Julius Global Health, University Utrecht Medical Center, Utrecht, the Netherlands
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Archie Campbell
- Usher Institute, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Health Data Research UK, University of Edinburgh, Nine, Edinburgh Bioquarter, 9 Little France Road, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Adolfo Correa
- Departments of Medicine, Pediatrics and Population Health Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Dawood Darbar
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Antonio De Luca
- Cardiothoracovascular Department, Division of Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina and University of Trieste, Trieste, Italy
| | - Jean-François Deleuze
- Université Paris-Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), 91057, Evry, France
- Laboratory of Excellence GENMED (Medical Genomics), Paris, France
- Centre d'Etude du Polymorphisme Humain, Fondation Jean Dausset, Paris, France
| | - Christina Ellervik
- Department of Data and Data Support, Region Zealand, 4180, Sorø, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Anuj Goel
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Christopher Grace
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Rebecca D Jackson
- Center for Clinical and Translational Science, Ohio State Medical Center, Columbus, OH, USA
| | - Jan A Kors
- Department of Medical Informatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, København, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter W Macfarlane
- Institute of Health and Wellbeing, School of Health and Wellbeing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Pau Navarro
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| | - Peter P Pramstaller
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Alexander P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Lorenz Risch
- Labormedizinisches zentrum Dr. Risch, Vaduz, Liechtenstein
- Faculty of Medical Sciences, Private University in the Principality of Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Ulrich Schotten
- Dept. of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Xia Shen
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Nansha District, Guangzhou, China
| | - Gianfranco Sinagra
- Cardiothoracovascular Department, Division of Cardiology, Azienda Sanitaria Universitaria Giuliano Isontina and University of Trieste, Trieste, Italy
| | - Elsayed Z Soliman
- Epidemiological Cardiology Research Center (EPICARE), Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Monika Stoll
- Maastricht Center for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
- Dept. of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
- Institute of Human Genetics, Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Eduardo Tarazona-Santos
- Department of Genetics, Ecology and Evolution, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andrew Tinker
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Katerina Trajanoska
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Eric Villard
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
| | - Helen R Warren
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christy L Avery
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Giorgia Girotto
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
- Durrer Center for Cardiovascular Research, Amsterdam, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands, Leiden, the Netherlands
| | | | - Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattte, WA, USA
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated with the University of Lübeck), Bolzano, Italy
| | - Antonio Luiz P Ribeiro
- Department of Internal Medicine, Faculdade de Medicina, Universidade Federal de Minas Gerais, Brazil, Belo Horizonte, Minas Gerais, Brazil
- Cardiology Service and Telehealth Center, Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil, Belo Horizonte, Minas Gerais, Brazil
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Departments of Pediatrics and Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Bruno H Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
- Department of Cardiology, Heart and Lung Division, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelia M van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, the Netherlands
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michele Orini
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Philippe Charron
- Sorbonne Universite, INSERM, UMR-S1166, Research Unit on Cardiovascular Disorders, Metabolism and Nutrition, Team Genomics & Pathophysiology of Cardiovascular Disease, Paris, 75013, France
- ICAN Institute for Cardiometabolism and Nutrition, Paris, 75013, France
- APHP, Cardiology Department, Pitié-Salpêtrière Hospital, Paris, 75013, France
- APHP, Département de Génétique, Centre de Référence Maladies Cardiaques Héréditaires, Pitié-Salpêtrière Hospital, Paris, 75013, France
| | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Division of Cardiovascular Medicine, John Radcliffe Hospital, Oxford, UK
- Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford, UK
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Henry J Lin
- Institute for Translational Genomics and Population Sciences/The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA, USA
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - James F Wilson
- Centre for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Jørgen K Kanters
- Laboratory of Experimental Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Borbala Mifsud
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Pier D Lambiase
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS trust, London, UK
- Institute of Cardiovascular Sciences, University of College London, London, UK
| | - Larisa G Tereshchenko
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Medicine, Cardiovascular Division, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| | - Patricia B Munroe
- William Harvey Research Institute, Clinical Pharmacology, Queen Mary University of London, London, UK.
- NIHR Barts Cardiovascular Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
6
|
Human multilineage pro-epicardium/foregut organoids support the development of an epicardium/myocardium organoid. Nat Commun 2022; 13:6981. [PMID: 36379937 PMCID: PMC9666429 DOI: 10.1038/s41467-022-34730-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 11/03/2022] [Indexed: 11/16/2022] Open
Abstract
The epicardium, the outer epithelial layer that covers the myocardium, derives from a transient organ known as pro-epicardium, crucial during heart organogenesis. The pro-epicardium develops from lateral plate mesoderm progenitors, next to septum transversum mesenchyme, a structure deeply involved in liver embryogenesis. Here we describe a self-organized human multilineage organoid that recreates the co-emergence of pro-epicardium, septum transversum mesenchyme and liver bud. Additionally, we study the impact of WNT, BMP and retinoic acid signaling modulation on multilineage organoid specification. By co-culturing these organoids with cardiomyocyte aggregates, we generated a self-organized heart organoid comprising an epicardium-like layer that fully surrounds a myocardium-like tissue. These heart organoids recapitulate the impact of epicardial cells on promoting cardiomyocyte proliferation and structural and functional maturation. Therefore, the human heart organoids described herein, open the path to advancing knowledge on how myocardium-epicardium interaction progresses during heart organogenesis in healthy or diseased settings.
Collapse
|
7
|
Abstract
An ensemble of in vitro cardiac tissue models has been developed over the past several decades to aid our understanding of complex cardiovascular disorders using a reductionist approach. These approaches often rely on recapitulating single or multiple clinically relevant end points in a dish indicative of the cardiac pathophysiology. The possibility to generate disease-relevant and patient-specific human induced pluripotent stem cells has further leveraged the utility of the cardiac models as screening tools at a large scale. To elucidate biological mechanisms in the cardiac models, it is critical to integrate physiological cues in form of biochemical, biophysical, and electromechanical stimuli to achieve desired tissue-like maturity for a robust phenotyping. Here, we review the latest advances in the directed stem cell differentiation approaches to derive a wide gamut of cardiovascular cell types, to allow customization in cardiac model systems, and to study diseased states in multiple cell types. We also highlight the recent progress in the development of several cardiovascular models, such as cardiac organoids, microtissues, engineered heart tissues, and microphysiological systems. We further expand our discussion on defining the context of use for the selection of currently available cardiac tissue models. Last, we discuss the limitations and challenges with the current state-of-the-art cardiac models and highlight future directions.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.).,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.)
| | - Suji Choi
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA (S.C., K.K.P.)
| | - Christina Alamana
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.).,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.)
| | - Kevin Kit Parker
- Disease Biophysics Group, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA (S.C., K.K.P.).,Harvard Stem Cell Institute, Harvard University, Cambridge, MA, Wyss Institute for Biologically Inspired Engineering, Boston, MA (K.K.P.)
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.).,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA (D.T., C.A., J.C.W.).,Greenstone Biosciences, Palo Alto, CA (J.C.W.)
| |
Collapse
|
8
|
Inhibition of RhoA and Cdc42 by miR-133a Modulates Retinoic Acid Signalling during Early Development of Posterior Cardiac Tube Segment. Int J Mol Sci 2022; 23:ijms23084179. [PMID: 35456995 PMCID: PMC9025022 DOI: 10.3390/ijms23084179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that multiple microRNAs play crucial roles in cardiovascular development, including miR-133a. Additionally, retinoic acid regulates atrial marker expression. In order to analyse the role of miR-133a as a modulator of retinoic acid signalling during the posterior segment of heart tube formation, we performed functional experiments with miR-133a and retinoic acid by means of microinjections into the posterior cardiac precursors of both primitive endocardial tubes in chick embryos. Subsequently, we subjected embryos to whole mount in situ hybridisation, immunohistochemistry and qPCR analysis. Our results demonstrate that miR-133a represses RhoA and Cdc42, as well as Raldh2/Aldh1a2, and the specific atrial markers Tbx5 and AMHC1, which play a key role during differentiation. Furthermore, we observed that miR-133a upregulates p21 and downregulates cyclin A by repressing RhoA and Cdc42, respectively, thus functioning as a cell proliferation inhibitor. Additionally, retinoic acid represses miR-133a, while it increases Raldh2, Tbx5 and AMHC1. Given that RhoA and Cdc42 are involved in Raldh2 expression and that they are modulated by miR-133a, which is influenced by retinoic acid signalling, our results suggest the presence of a negative feedback mechanism between miR-133a and retinoic acid during early development of the posterior cardiac tube segment. Despite additional unexplored factors being possible contributors to this negative feedback mechanism, miR-133a might also be considered as a potential therapeutic tool for the diagnosis, therapy and prognosis of cardiac diseases.
Collapse
|
9
|
Enhanced Loss of Retinoic Acid Network Genes in Xenopus laevis Achieves a Tighter Signal Regulation. Cells 2022; 11:cells11030327. [PMID: 35159137 PMCID: PMC8834563 DOI: 10.3390/cells11030327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 12/10/2022] Open
Abstract
Retinoic acid (RA) is a major regulatory signal during embryogenesis produced from vitamin A (retinol) by an extensive, autoregulating metabolic and signaling network to prevent fluctuations that result in developmental malformations. Xenopus laevis is an allotetraploid hybrid frog species whose genome includes L (long) and S (short) chromosomes from the originating species. Evolutionarily, the X. laevis subgenomes have been losing either L or S homoeologs in about 43% of genes to generate singletons. In the RA network, out of the 47 genes, about 47% have lost one of the homoeologs, like the genome average. Interestingly, RA metabolism genes from storage (retinyl esters) to retinaldehyde production exhibit enhanced gene loss with 75% singletons out of 28 genes. The effect of this gene loss on RA signaling autoregulation was studied. Employing transient RA manipulations, homoeolog gene pairs were identified in which one homoeolog exhibits enhanced responses or looser regulation than the other, while in other pairs both homoeologs exhibit similar RA responses. CRISPR/Cas9 targeting of individual homoeologs to reduce their activity supports the hypothesis where the RA metabolic network gene loss results in tighter network regulation and more efficient RA robustness responses to overcome complex regulation conditions.
Collapse
|
10
|
Wiesinger A, Boink GJJ, Christoffels VM, Devalla HD. Retinoic acid signaling in heart development: Application in the differentiation of cardiovascular lineages from human pluripotent stem cells. Stem Cell Reports 2021; 16:2589-2606. [PMID: 34653403 PMCID: PMC8581056 DOI: 10.1016/j.stemcr.2021.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/16/2021] [Accepted: 09/17/2021] [Indexed: 11/29/2022] Open
Abstract
Retinoic acid (RA) signaling plays an important role during heart development in establishing anteroposterior polarity, formation of inflow and outflow tract progenitors, and growth of the ventricular compact wall. RA is also utilized as a key ingredient in protocols designed for generating cardiac cell types from pluripotent stem cells (PSCs). This review discusses the role of RA in cardiogenesis, currently available protocols that employ RA for differentiation of various cardiovascular lineages, and plausible transcriptional mechanisms underlying this fate specification. These insights will inform further development of desired cardiac cell types from human PSCs and their application in preclinical and clinical research.
Collapse
Affiliation(s)
- Alexandra Wiesinger
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Department of Cardiology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Duong TB, Waxman JS. Patterning of vertebrate cardiac progenitor fields by retinoic acid signaling. Genesis 2021; 59:e23458. [PMID: 34665508 DOI: 10.1002/dvg.23458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
The influence of retinoic acid (RA) signaling on vertebrate development has a well-studied history. Cumulatively, we now understand that RA signaling has a conserved requirement early in development restricting cardiac progenitors within the anterior lateral plate mesoderm of vertebrate embryos. Moreover, genetic and pharmacological manipulations of RA signaling in vertebrate models have shown that proper heart development is achieved through the deployment of positive and negative feedback mechanisms, which maintain appropriate RA levels. In this brief review, we present a chronological overview of key work that has led to a current model of the critical role for early RA signaling in limiting the generation of cardiac progenitors within vertebrate embryos. Furthermore, we integrate the previous work in mice and our recent findings using zebrafish, which together show that RA signaling has remarkably conserved influences on the later-differentiating progenitor populations at the arterial and venous poles. We discuss how recognizing the significant conservation of RA signaling on the differentiation of these progenitor populations offers new perspectives and may impact future work dedicated to examining vertebrate heart development.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
12
|
Parihar M, Bendelac-Kapon L, Gur M, Abbou T, Belorkar A, Achanta S, Kinberg K, Vadigepalli R, Fainsod A. Retinoic Acid Fluctuation Activates an Uneven, Direction-Dependent Network-Wide Robustness Response in Early Embryogenesis. Front Cell Dev Biol 2021; 9:747969. [PMID: 34746144 PMCID: PMC8564372 DOI: 10.3389/fcell.2021.747969] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/23/2021] [Indexed: 01/15/2023] Open
Abstract
Robustness is a feature of regulatory pathways to ensure signal consistency in light of environmental changes or genetic polymorphisms. The retinoic acid (RA) pathway, is a central developmental and tissue homeostasis regulatory signal, strongly dependent on nutritional sources of retinoids and affected by environmental chemicals. This pathway is characterized by multiple proteins or enzymes capable of performing each step and their integration into a self-regulating network. We studied RA network robustness by transient physiological RA signaling disturbances followed by kinetic transcriptomic analysis of the recovery during embryogenesis. The RA metabolic network was identified as the main regulated module to achieve signaling robustness using an unbiased pattern analysis. We describe the network-wide responses to RA signal manipulation and found the feedback autoregulation to be sensitive to the direction of the RA perturbation: RA knockdown exhibited an upper response limit, whereas RA addition had a minimal feedback-activation threshold. Surprisingly, our robustness response analysis suggests that the RA metabolic network regulation exhibits a multi-objective optimization, known as Pareto optimization, characterized by trade-offs between competing functionalities. We observe that efficient robustness to increasing RA is accompanied by worsening robustness to reduced RA levels and vice versa. This direction-dependent trade-off in the network-wide feedback response, results in an uneven robustness capacity of the RA network during early embryogenesis, likely a significant contributor to the manifestation of developmental defects.
Collapse
Affiliation(s)
- Madhur Parihar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Liat Bendelac-Kapon
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Michal Gur
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Tali Abbou
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Abha Belorkar
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Sirisha Achanta
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Keren Kinberg
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abraham Fainsod
- Department of Developmental Biology and Cancer Research, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Jerusalem, Israel
| |
Collapse
|
13
|
Da Silva F, Jian Motamedi F, Weerasinghe Arachchige LC, Tison A, Bradford ST, Lefebvre J, Dolle P, Ghyselinck NB, Wagner KD, Schedl A. Retinoic acid signaling is directly activated in cardiomyocytes and protects mouse hearts from apoptosis after myocardial infarction. eLife 2021; 10:68280. [PMID: 34623260 PMCID: PMC8530512 DOI: 10.7554/elife.68280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 10/07/2021] [Indexed: 12/22/2022] Open
Abstract
Retinoic acid (RA) is an essential signaling molecule for cardiac development and plays a protective role in the heart after myocardial infarction (MI). In both cases, the effect of RA signaling on cardiomyocytes, the principle cell type of the heart, has been reported to be indirect. Here we have developed an inducible murine transgenic RA-reporter line using CreERT2 technology that permits lineage tracing of RA-responsive cells and faithfully recapitulates endogenous RA activity in multiple organs during embryonic development. Strikingly, we have observed a direct RA response in cardiomyocytes during mid-late gestation and after MI. Ablation of RA signaling through deletion of the Aldh1a1/a2/a3 genes encoding RA-synthesizing enzymes leads to increased cardiomyocyte apoptosis in adults subjected to MI. RNA sequencing analysis reveals Tgm2 and Ace1, two genes with well-established links to cardiac repair, as potential targets of RA signaling in primary cardiomyocytes, thereby providing novel links between the RA pathway and heart disease.
Collapse
Affiliation(s)
| | | | | | - Amelie Tison
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice, France
| | | | | | - Pascal Dolle
- IGBMC, Inserm U1258, UNISTRA CNRS, Illkirch, France
| | | | - Kay D Wagner
- Université Côte d'Azur, Inserm, CNRS, iBV, Nice, France
| | | |
Collapse
|
14
|
Approaches to Optimize Stem Cell-Derived Cardiomyocyte Maturation and Function. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00197-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
15
|
Honda Y, Li J, Hino A, Tsujimoto S, Lee JK. High-Throughput Drug Screening System Based on Human Induced Pluripotent Stem Cell-Derived Atrial Myocytes ∼ A Novel Platform to Detect Cardiac Toxicity for Atrial Arrhythmias. Front Pharmacol 2021; 12:680618. [PMID: 34413773 PMCID: PMC8369502 DOI: 10.3389/fphar.2021.680618] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
Evaluation of proarrhythmic properties is critical for drug discovery. In particular, QT prolongation in electrocardiograms has been utilized as a surrogate marker in many evaluation systems to assess the risk of torsade de pointes and lethal ventricular arrhythmia. Recently, new evaluation systems based on human iPS cell-derived cardiomyocytes have been established. On the other hand, in clinical situations, it has been reported that the incidence of atrial arrhythmias such as atrial fibrillation has been increasing every year, with the prediction of a persistent increase in the near future. As to the increased incidence of atrial arrhythmias, in addition to the increased population of geriatric patients, a wide variety of drug treatments may be related, as an experimental method to detect drug-induced atrial arrhythmia has not been established so far. In the present study, we characterized the atrial-like cardiomyocytes derived from human induced pluripotent stem cells and examined their potential for the evaluation of drug-induced atrial arrhythmia. Atrial-like cardiomyocytes were induced by adding retinoic acid (RA) during the process of myocardial differentiation, and their characteristics were compared to those of RA-free cardiomyocytes. Using gene expression and membrane potential analysis, it was confirmed that the cells with or without RA treatment have atrial or ventricular like cardiomyocytes, respectively. Using the ultra-rapid activating delayed rectifier potassium current (IKur) channel inhibitor, which is specific to atrial cardiomyocytes, Pulse width duration (PWD) 30cF prolongation was confirmed only in atrial-like cardiomyocytes. In addition, ventricular like cardiomyocytes exhibited an early after depolarization by treatment with rapidly activating delayed rectifier potassium current (IKr) channel inhibitor, which induces ventricular arrhythmia in clinical situations. Here, we have established a high-throughput drug evaluation system using human iPS cell-derived atrial-like cardiomyocytes. Based on the obtained data, the system might be a valuable platform to detect potential risks for drug-induced atrial arrhythmias.
Collapse
Affiliation(s)
- Yayoi Honda
- Sumitomo-Dainippon Pharma CO., Ltd., Osaka, Japan.,Bioanalysis Group, Osaka Laboratory, Technical Solution Headquarters, Sumika Chemical Analysis Service, Ltd., Osaka, Japan
| | - Jun Li
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Aya Hino
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
16
|
Ge Y, Smits AM, Liu J, Zhang J, van Brakel TJ, Goumans MJTH, Jongbloed MRM, de Vries AAF. Generation, Characterization, and Application of Inducible Proliferative Adult Human Epicardium-Derived Cells. Cells 2021; 10:2064. [PMID: 34440833 PMCID: PMC8391799 DOI: 10.3390/cells10082064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
RATIONALE In recent decades, the great potential of human epicardium-derived cells (EPDCs) as an endogenous cell source for cardiac regeneration has been recognized. The limited availability and low proliferation capacity of primary human EPDCs and phenotypic differences between EPDCs obtained from different individuals hampers their reproducible use for experimental studies. AIM To generate and characterize inducible proliferative adult human EPDCs for use in fundamental and applied research. METHODS AND RESULTS Inducible proliferation of human EPDCs was achieved by doxycycline-controlled expression of simian virus 40 large T antigen (LT) with a repressor-based lentiviral Tet-On system. In the presence of doxycycline, these inducible EPDCs (iEPDCs) displayed high and long-term proliferation capacity. After doxycycline removal, LT expression ceased and the iEPDCs regained their cuboidal epithelial morphology. Similar to primary EPDCs, iEPDCs underwent an epithelial-to-mesenchymal transition (EMT) after stimulation with transforming growth factor β3. This was confirmed by reverse transcription-quantitative polymerase chain reaction analysis of epithelial and mesenchymal marker gene expression and (immuno) cytochemical staining. Collagen gel-based cell invasion assays demonstrated that mesenchymal iEPDCs, like primary EPDCs, possess increased invasion and migration capacities as compared to their epithelial counterparts. Mesenchymal iEPDCs co-cultured with sympathetic ganglia stimulated neurite outgrowth similarly to primary EPDCs. CONCLUSION Using an inducible LT expression system, inducible proliferative adult human EPDCs were generated displaying high proliferative capacity in the presence of doxycycline. These iEPDCs maintain essential epicardial characteristics with respect to morphology, EMT ability, and paracrine signaling following doxycycline removal. This renders iEPDCs a highly useful new in vitro model for studying human epicardial properties.
Collapse
Affiliation(s)
- Yang Ge
- Department of Anatomy & Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (Y.G.); (M.R.M.J.)
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (J.L.); (J.Z.); (A.A.F.d.V.)
| | - Anke M. Smits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
| | - Jia Liu
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (J.L.); (J.Z.); (A.A.F.d.V.)
- Central Laboratory, Longgang District People’s Hospital of Shenzhen & The Third Affiliated Hospital of The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Juan Zhang
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (J.L.); (J.Z.); (A.A.F.d.V.)
| | - Thomas J. van Brakel
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZC Leiden, The Netherlands;
| | - Marie José T. H. Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands;
| | - Monique R. M. Jongbloed
- Department of Anatomy & Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands; (Y.G.); (M.R.M.J.)
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (J.L.); (J.Z.); (A.A.F.d.V.)
| | - Antoine A. F. de Vries
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; (J.L.); (J.Z.); (A.A.F.d.V.)
| |
Collapse
|
17
|
Every Beat You Take-The Wilms' Tumor Suppressor WT1 and the Heart. Int J Mol Sci 2021; 22:ijms22147675. [PMID: 34299295 PMCID: PMC8306835 DOI: 10.3390/ijms22147675] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
Nearly three decades ago, the Wilms’ tumor suppressor Wt1 was identified as a crucial regulator of heart development. Wt1 is a zinc finger transcription factor with multiple biological functions, implicated in the development of several organ systems, among them cardiovascular structures. This review summarizes the results from many research groups which allowed to establish a relevant function for Wt1 in cardiac development and disease. During development, Wt1 is involved in fundamental processes as the formation of the epicardium, epicardial epithelial-mesenchymal transition, coronary vessel development, valve formation, organization of the cardiac autonomous nervous system, and formation of the cardiac ventricles. Wt1 is further implicated in cardiac disease and repair in adult life. We summarize here the current knowledge about expression and function of Wt1 in heart development and disease and point out controversies to further stimulate additional research in the areas of cardiac development and pathophysiology. As re-activation of developmental programs is considered as paradigm for regeneration in response to injury, understanding of these processes and the molecules involved therein is essential for the development of therapeutic strategies, which we discuss on the example of WT1.
Collapse
|
18
|
Barisón MJ, Pereira IT, Waloski Robert A, Dallagiovanna B. Reorganization of Metabolism during Cardiomyogenesis Implies Time-Specific Signaling Pathway Regulation. Int J Mol Sci 2021; 22:1330. [PMID: 33572750 PMCID: PMC7869011 DOI: 10.3390/ijms22031330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding the cell differentiation process involves the characterization of signaling and regulatory pathways. The coordinated action involved in multilevel regulation determines the commitment of stem cells and their differentiation into a specific cell lineage. Cellular metabolism plays a relevant role in modulating the expression of genes, which act as sensors of the extra-and intracellular environment. In this work, we analyzed mRNAs associated with polysomes by focusing on the expression profile of metabolism-related genes during the cardiac differentiation of human embryonic stem cells (hESCs). We compared different time points during cardiac differentiation (pluripotency, embryoid body aggregation, cardiac mesoderm, cardiac progenitor and cardiomyocyte) and showed the immature cell profile of energy metabolism. Highly regulated canonical pathways are thoroughly discussed, such as those involved in metabolic signaling and lipid homeostasis. We reveal the critical relevance of retinoic X receptor (RXR) heterodimers in upstream retinoic acid metabolism and their relationship with thyroid hormone signaling. Additionally, we highlight the importance of lipid homeostasis and extracellular matrix component biosynthesis during cardiomyogenesis, providing new insights into how hESCs reorganize their metabolism during in vitro cardiac differentiation.
Collapse
Affiliation(s)
| | | | | | - Bruno Dallagiovanna
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Rua Professor Algacyr Munhoz Mader, 3775, Curitiba, PR 81350-010, Brazil; (M.J.B.); (I.T.P.); (A.W.R.)
| |
Collapse
|
19
|
Klyuyeva AV, Belyaeva OV, Goggans KR, Krezel W, Popov KM, Kedishvili NY. Changes in retinoid metabolism and signaling associated with metabolic remodeling during fasting and in type I diabetes. J Biol Chem 2021; 296:100323. [PMID: 33485967 PMCID: PMC7949101 DOI: 10.1016/j.jbc.2021.100323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/11/2021] [Accepted: 01/20/2021] [Indexed: 12/12/2022] Open
Abstract
Liver is the central metabolic hub that coordinates carbohydrate and lipid metabolism. The bioactive derivative of vitamin A, retinoic acid (RA), was shown to regulate major metabolic genes including phosphoenolpyruvate carboxykinase, fatty acid synthase, carnitine palmitoyltransferase 1, and glucokinase among others. Expression levels of these genes undergo profound changes during adaptation to fasting or in metabolic diseases such as type 1 diabetes (T1D). However, it is unknown whether the levels of hepatic RA change during metabolic remodeling. This study investigated the dynamics of hepatic retinoid metabolism and signaling in the fed state, in fasting, and in T1D. Our results show that fed-to-fasted transition is associated with significant decrease in hepatic retinol dehydrogenase (RDH) activity, the rate-limiting step in RA biosynthesis, and downregulation of RA signaling. The decrease in RDH activity correlates with the decreased abundance and altered subcellular distribution of RDH10 while Rdh10 transcript levels remain unchanged. In contrast to fasting, untreated T1D is associated with upregulation of RA signaling and an increase in hepatic RDH activity, which correlates with the increased abundance of RDH10 in microsomal membranes. The dynamic changes in RDH10 protein levels in the absence of changes in its transcript levels imply the existence of posttranscriptional regulation of RDH10 protein. Together, these data suggest that the downregulation of hepatic RA biosynthesis, in part via the decrease in RDH10, is an integral component of adaptation to fasting. In contrast, the upregulation of hepatic RA biosynthesis and signaling in T1D might contribute to metabolic inflexibility associated with this disease.
Collapse
Affiliation(s)
- Alla V Klyuyeva
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olga V Belyaeva
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kelli R Goggans
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Wojciech Krezel
- Institute of Genetics and Molecular and Cellular Biology (IGBMC) - INSERM, University of Strasbourg, Strasbourg, France
| | - Kirill M Popov
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Natalia Y Kedishvili
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
20
|
Fukushima H, Yoshioka M, Kawatou M, López-Dávila V, Takeda M, Kanda Y, Sekino Y, Yoshida Y, Yamashita JK. Specific induction and long-term maintenance of high purity ventricular cardiomyocytes from human induced pluripotent stem cells. PLoS One 2020; 15:e0241287. [PMID: 33137106 PMCID: PMC7605685 DOI: 10.1371/journal.pone.0241287] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022] Open
Abstract
Currently, cardiomyocyte (CM) differentiation methods require a purification step after CM induction to ensure the high purity of the cell population. Here we show an improved human CM differentiation protocol with which high-purity ventricular-type CMs can be obtained and maintained without any CM purification process. We induced and collected a mesodermal cell population (platelet-derived growth factor receptor-α (PDGFRα)-positive cells) that can respond to CM differentiation cues, and then stimulated CM differentiation by means of Wnt inhibition. This method reproducibly generated CMs with purities above 95% in several human pluripotent stem cell lines. Furthermore, these CM populations were maintained in culture at such high purity without any further CM purification step for over 200 days. The majority of these CMs (>95%) exhibited a ventricular-like phenotype with a tendency to structural and electrophysiological maturation, including T-tubule-like structure formation and the ability to respond to QT prolongation drugs. This is a simple and valuable method to stably generate CM populations suitable for cardiac toxicology testing, disease modeling and regenerative medicine.
Collapse
Affiliation(s)
- Hiroyuki Fukushima
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Miki Yoshioka
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masahide Kawatou
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Cardiovascular Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Víctor López-Dávila
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Masafumi Takeda
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yuko Sekino
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yoshinori Yoshida
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Jun K. Yamashita
- Department of Cell Growth and Differentiation, Laboratory of Stem Cell Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
21
|
Bernheim S, Meilhac SM. Mesoderm patterning by a dynamic gradient of retinoic acid signalling. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190556. [PMID: 32829679 PMCID: PMC7482219 DOI: 10.1098/rstb.2019.0556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/15/2022] Open
Abstract
Retinoic acid (RA), derived from vitamin A, is a major teratogen, clinically recognized in 1983. Identification of its natural presence in the embryo and dissection of its molecular mechanism of action became possible in the animal model with the advent of molecular biology, starting with the cloning of its nuclear receptor. In normal development, the dose of RA is tightly controlled to regulate organ formation. Its production depends on enzymes, which have a dynamic expression profile during embryonic development. As a small molecule, it diffuses rapidly and acts as a morphogen. Here, we review advances in deciphering how endogenously produced RA provides positional information to cells. We compare three mesodermal tissues, the limb, the somites and the heart, and discuss how RA signalling regulates antero-posterior and left-right patterning. A common principle is the establishment of its spatio-temporal dynamics by positive and negative feedback mechanisms and by antagonistic signalling by FGF. However, the response is cell-specific, pointing to the existence of cofactors and effectors, which are as yet incompletely characterized. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Ségolène Bernheim
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France
- INSERM UMR1163, 75015 Paris, France
- Université de Paris, Paris, France
| | - Sigolène M. Meilhac
- Imagine-Institut Pasteur, Laboratory of Heart Morphogenesis, 75015 Paris, France
- INSERM UMR1163, 75015 Paris, France
- Université de Paris, Paris, France
| |
Collapse
|
22
|
Protze SI, Lee JH, Keller GM. Human Pluripotent Stem Cell-Derived Cardiovascular Cells: From Developmental Biology to Therapeutic Applications. Cell Stem Cell 2020; 25:311-327. [PMID: 31491395 DOI: 10.1016/j.stem.2019.07.010] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Advances in our understanding of cardiovascular development have provided a roadmap for the directed differentiation of human pluripotent stem cells (hPSCs) to the major cell types found in the heart. In this Perspective, we review the state of the field in generating and maturing cardiovascular cells from hPSCs based on our fundamental understanding of heart development. We then highlight their applications for studying human heart development, modeling disease-performing drug screening, and cell replacement therapy. With the advancements highlighted here, the promise that hPSCs will deliver new treatments for degenerative and debilitating diseases may soon be fulfilled.
Collapse
Affiliation(s)
- Stephanie I Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Jee Hoon Lee
- BlueRock Therapeutics ULC, Toronto, ON M5G 1L7, Canada
| | - Gordon M Keller
- McEwen Stem Cell Institute, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada; Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
23
|
Gunawan MG, Sangha SS, Shafaattalab S, Lin E, Heims-Waldron DA, Bezzerides VJ, Laksman Z, Tibbits GF. Drug screening platform using human induced pluripotent stem cell-derived atrial cardiomyocytes and optical mapping. Stem Cells Transl Med 2020; 10:68-82. [PMID: 32927497 PMCID: PMC7780813 DOI: 10.1002/sctm.19-0440] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022] Open
Abstract
Current drug development efforts for the treatment of atrial fibrillation are hampered by the fact that many preclinical models have been unsuccessful in reproducing human cardiac physiology and its response to medications. In this study, we demonstrated an approach using human induced pluripotent stem cell-derived atrial and ventricular cardiomyocytes (hiPSC-aCMs and hiPSC-vCMs, respectively) coupled with a sophisticated optical mapping system for drug screening of atrial-selective compounds in vitro. We optimized differentiation of hiPSC-aCMs by modulating the WNT and retinoid signaling pathways. Characterization of the transcriptome and proteome revealed that retinoic acid pushes the differentiation process into the atrial lineage and generated hiPSC-aCMs. Functional characterization using optical mapping showed that hiPSC-aCMs have shorter action potential durations and faster Ca2+ handling dynamics compared with hiPSC-vCMs. Furthermore, pharmacological investigation of hiPSC-aCMs captured atrial-selective effects by displaying greater sensitivity to atrial-selective compounds 4-aminopyridine, AVE0118, UCL1684, and vernakalant when compared with hiPSC-vCMs. These results established that a model system incorporating hiPSC-aCMs combined with optical mapping is well-suited for preclinical drug screening of novel and targeted atrial selective compounds.
Collapse
Affiliation(s)
- Marvin G Gunawan
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Sarabjit S Sangha
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Cardiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Eric Lin
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | | | - Zachary Laksman
- Division of Cardiology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.,Tibbits Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
24
|
Asp M, Giacomello S, Larsson L, Wu C, Fürth D, Qian X, Wärdell E, Custodio J, Reimegård J, Salmén F, Österholm C, Ståhl PL, Sundström E, Åkesson E, Bergmann O, Bienko M, Månsson-Broberg A, Nilsson M, Sylvén C, Lundeberg J. A Spatiotemporal Organ-Wide Gene Expression and Cell Atlas of the Developing Human Heart. Cell 2020; 179:1647-1660.e19. [PMID: 31835037 DOI: 10.1016/j.cell.2019.11.025] [Citation(s) in RCA: 397] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/06/2019] [Accepted: 11/14/2019] [Indexed: 10/25/2022]
Abstract
The process of cardiac morphogenesis in humans is incompletely understood. Its full characterization requires a deep exploration of the organ-wide orchestration of gene expression with a single-cell spatial resolution. Here, we present a molecular approach that reveals the comprehensive transcriptional landscape of cell types populating the embryonic heart at three developmental stages and that maps cell-type-specific gene expression to specific anatomical domains. Spatial transcriptomics identified unique gene profiles that correspond to distinct anatomical regions in each developmental stage. Human embryonic cardiac cell types identified by single-cell RNA sequencing confirmed and enriched the spatial annotation of embryonic cardiac gene expression. In situ sequencing was then used to refine these results and create a spatial subcellular map for the three developmental phases. Finally, we generated a publicly available web resource of the human developing heart to facilitate future studies on human cardiogenesis.
Collapse
Affiliation(s)
- Michaela Asp
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Ludvig Larsson
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Chenglin Wu
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Daniel Fürth
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Xiaoyan Qian
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Eva Wärdell
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Joaquin Custodio
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Johan Reimegård
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fredrik Salmén
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, Cancer Genomics Netherlands, Utrecht, the Netherlands
| | - Cecilia Österholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Patrik L Ståhl
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, R&D Unit, Stockholms Sjukhem, Stockholm, Sweden
| | - Elisabet Åkesson
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, R&D Unit, Stockholms Sjukhem, Stockholm, Sweden
| | - Olaf Bergmann
- Center for Regenerative Therapies Dresden, TU-Dresden, Dresden, Germany; Karolinska Institutet, Cell and Molecular Biology, Stockholm, Sweden
| | - Magda Bienko
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Christer Sylvén
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden.
| |
Collapse
|
25
|
How Dietary Deficiency Studies Have Illuminated the Many Roles of Vitamin A During Development and Postnatal Life. Subcell Biochem 2020; 95:1-26. [PMID: 32297294 DOI: 10.1007/978-3-030-42282-0_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Vitamin A deficiency studies have been carried out since the early 1900s. Initially, these studies led to the identification of fat soluble A as a unique and essential component of the diet of rodents, birds, and humans. Continuing work established that vitamin A deficiency produces biochemical and physiological dysfunction in almost every vertebrate organ system from conception to death. This chapter begins with a review of representative historical and current studies that used the nutritional vitamin A deficiency research model to gain an understanding of the many roles vitamin A plays in prenatal and postnatal development and well-being. This is followed by a discussion of recent studies that show specific effects of vitamin A deficiency on prenatal development and postnatal maintenance of the olfactory epithelium, brain, and heart. Vitamin A deficiency studies have helped define the necessity of vitamin A for the health of all vertebrates, including farm animals, but the breadth of deficient states and their individual effects on health have not been fully determined. Future work is needed to develop tools to assess the complete vitamin A status of an organism and to define the levels of vitamin A that optimally support molecular and systems level processes during all ages and stages of life.
Collapse
|
26
|
Abstract
As the first organ to form and function in all vertebrates, the heart is crucial to development. Tightly-regulated levels of retinoic acid (RA) are critical for the establishment of the regulatory networks that drive normal cardiac development. Thus, the heart is an ideal organ to investigate RA signaling, with much work remaining to be done in this area. Herein, we highlight the role of RA signaling in vertebrate heart development and provide an overview of the field's inception, its current state, and in what directions it might progress so that it may yield fruitful insight for therapeutic applications within the domain of regenerative medicine.
Collapse
|
27
|
Abstract
The heart is lined by a single layer of mesothelial cells called the epicardium that provides important cellular contributions for embryonic heart formation. The epicardium harbors a population of progenitor cells that undergo epithelial-to-mesenchymal transition displaying characteristic conversion of planar epithelial cells into multipolar and invasive mesenchymal cells before differentiating into nonmyocyte cardiac lineages, such as vascular smooth muscle cells, pericytes, and fibroblasts. The epicardium is also a source of paracrine cues that are essential for fetal cardiac growth, coronary vessel patterning, and regenerative heart repair. Although the epicardium becomes dormant after birth, cardiac injury reactivates developmental gene programs that stimulate epithelial-to-mesenchymal transition; however, it is not clear how the epicardium contributes to disease progression or repair in the adult. In this review, we will summarize the molecular mechanisms that control epicardium-derived progenitor cell migration, and the functional contributions of the epicardium to heart formation and cardiomyopathy. Future perspectives will be presented to highlight emerging therapeutic strategies aimed at harnessing the regenerative potential of the fetal epicardium for cardiac repair.
Collapse
Affiliation(s)
- Pearl Quijada
- From the Aab Cardiovascular Research Institute (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY.,Department of Medicine (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY
| | | | - Eric M Small
- From the Aab Cardiovascular Research Institute (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY.,Department of Medicine (P.Q., E.M.S.), University of Rochester, School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
28
|
Sirbu IO, Chiş AR, Moise AR. Role of carotenoids and retinoids during heart development. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158636. [PMID: 31978553 DOI: 10.1016/j.bbalip.2020.158636] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 02/08/2023]
Abstract
The nutritional requirements of the developing embryo are complex. In the case of dietary vitamin A (retinol, retinyl esters and provitamin A carotenoids), maternal derived nutrients serve as precursors to signaling molecules such as retinoic acid, which is required for embryonic patterning and organogenesis. Despite variations in the composition and levels of maternal vitamin A, embryonic tissues need to generate a precise amount of retinoic acid to avoid congenital malformations. Here, we summarize recent findings regarding the role and metabolism of vitamin A during heart development and we survey the association of genes known to affect retinoid metabolism or signaling with various inherited disorders. A better understanding of the roles of vitamin A in the heart and of the factors that affect retinoid metabolism and signaling can help design strategies to meet nutritional needs and to prevent birth defects and disorders associated with altered retinoid metabolism. This article is part of a Special Issue entitled Carotenoids recent advances in cell and molecular biology edited by Johannes von Lintig and Loredana Quadro.
Collapse
Affiliation(s)
- Ioan Ovidiu Sirbu
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, 300041 Timisoara, Romania; Timisoara Institute of Complex Systems, V. Lucaciu 18, 300044 Timisoara, Romania.
| | - Aimée Rodica Chiş
- Biochemistry Department, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Nr. 2, 300041 Timisoara, Romania
| | - Alexander Radu Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; Department of Chemistry and Biochemistry, Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON P3E 2C6, Canada.
| |
Collapse
|
29
|
Halver J, Wenzel K, Sendker J, Carrillo García C, Erdelmeier CAJ, Willems E, Mercola M, Symma N, Könemann S, Koch E, Hensel A, Schade D. Crataegus Extract WS®1442 Stimulates Cardiomyogenesis and Angiogenesis From Stem Cells: A Possible New Pharmacology for Hawthorn? Front Pharmacol 2019; 10:1357. [PMID: 31849643 PMCID: PMC6902660 DOI: 10.3389/fphar.2019.01357] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022] Open
Abstract
Extracts from the leaves and flowers of Crataegus spp. (i.e., hawthorn species) have been traditionally used with documented preclinical and clinical activities in cardiovascular medicine. Based on reported positive effects on heart muscle after ischemic injury and the overall cardioprotective profile, the present study addressed potential contributions of Crataegus extracts to cardiopoietic differentiation from stem cells. The quantified Crataegus extract WS®1442 stimulated cardiomyogenesis from murine and human embryonic stem cells (ESCs). Mechanistically, this effect was found to be induced by promoting differentiation of cardiovascular progenitor cell populations but not by proliferation. Bioassay-guided fractionation, phytochemical and analytical profiling suggested high-molecular weight ingredients as the active principle with at least part of the activity due to oligomeric procyanidines (OPCs) with a degree of polymerization between 3 and 6 (DP3–6). Transcriptome profiling in mESCs suggested two main, plausible mechanisms: These were early, stress-associated cellular events along with the modulation of distinct developmental pathways, including the upregulation of brain-derived neurotrophic factor (BDNF) and retinoic acid as well as the inhibition of transforming growth factor β/bone morphogenetic protein (TGFβ/BMP) and fibroblast growth factor (FGF) signaling. In addition, WS®1442 stimulated angiogenesis ex vivo in Sca-1+ progenitor cells from adult mice hearts. These in vitro data provide evidence for a differentiation promoting activity of WS®1442 on distinct cardiovascular stem/progenitor cells that could be valuable for therapeutic heart regeneration after myocardial infarction. However, the in vivo relevance of this new pharmacological activity of Crataegus spp. remains to be investigated and active ingredients from bioactive fractions will have to be further characterized.
Collapse
Affiliation(s)
- Jonas Halver
- Department of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany
| | - Kristin Wenzel
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Partner site Greifswald, DZHK, German Centre for Cardiovascular Research, Greifswald, Germany
| | - Jandirk Sendker
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Carmen Carrillo García
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Erik Willems
- Muscle Development and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute (SBP), La Jolla, United States
| | - Mark Mercola
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, Stanford, United States
| | - Nico Symma
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Stephanie Könemann
- Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany.,Partner site Greifswald, DZHK, German Centre for Cardiovascular Research, Greifswald, Germany
| | - Egon Koch
- Preclinical Research, Dr. Willmar Schwabe GmbH & Co. KG, Karlsruhe, Germany
| | - Andreas Hensel
- Institute of Pharmaceutical Biology and Phytochemistry, University of Münster, Münster, Germany
| | - Dennis Schade
- Department of Chemistry and Chemical Biology, Technical University of Dortmund, Dortmund, Germany.,Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Christian-Albrechts-University of Kiel, Kiel, Germany.,Partner site Kiel, DZHK, German Centre for Cardiovascular Research, Kiel, Germany
| |
Collapse
|
30
|
Wang S, Yu J, Kane MA, Moise AR. Modulation of retinoid signaling: therapeutic opportunities in organ fibrosis and repair. Pharmacol Ther 2019; 205:107415. [PMID: 31629008 DOI: 10.1016/j.pharmthera.2019.107415] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/17/2019] [Indexed: 02/08/2023]
Abstract
The vitamin A metabolite, retinoic acid, is an important signaling molecule during embryonic development serving critical roles in morphogenesis, organ patterning and skeletal and neural development. Retinoic acid is also important in postnatal life in the maintenance of tissue homeostasis, while retinoid-based therapies have long been used in the treatment of a variety of cancers and skin disorders. As the number of people living with chronic disorders continues to increase, there is great interest in extending the use of retinoid therapies in promoting the maintenance and repair of adult tissues. However, there are still many conflicting results as we struggle to understand the role of retinoic acid in the multitude of processes that contribute to tissue injury and repair. This review will assess our current knowledge of the role retinoic acid signaling in the development of fibroblasts, and their transformation to myofibroblasts, and of the potential use of retinoid therapies in the treatment of organ fibrosis.
Collapse
Affiliation(s)
- Suya Wang
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, 21201, USA.
| | - Alexander R Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada; Departments of Chemistry and Biochemistry, and Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| |
Collapse
|
31
|
Wang S, Moise AR. Recent insights on the role and regulation of retinoic acid signaling during epicardial development. Genesis 2019; 57:e23303. [PMID: 31066193 PMCID: PMC6682438 DOI: 10.1002/dvg.23303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/18/2022]
Abstract
The vitamin A metabolite, retinoic acid, carries out essential and conserved roles in vertebrate heart development. Retinoic acid signals via retinoic acid receptors (RAR)/retinoid X receptors (RXRs) heterodimers to induce the expression of genes that control cell fate specification, proliferation, and differentiation. Alterations in retinoic acid levels are often associated with congenital heart defects. Therefore, embryonic levels of retinoic acid need to be carefully regulated through the activity of enzymes, binding proteins and transporters involved in vitamin A metabolism. Here, we review evidence of the complex mechanisms that control the fetal uptake and synthesis of retinoic acid from vitamin A precursors. Next, we highlight recent evidence of the role of retinoic acid in orchestrating myocardial compact zone growth and coronary vascular development.
Collapse
Affiliation(s)
- Suya Wang
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Alexander R. Moise
- Medical Sciences Division, Northern Ontario School of Medicine, Sudbury, ON P3E 2C6, Canada
- Departments of Chemistry and Biochemistry, and Biology and Biomolecular Sciences Program, Laurentian University, Sudbury, ON, P3E 2C6 Canada
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, 66045, USA
| |
Collapse
|
32
|
Perl E, Waxman JS. Reiterative Mechanisms of Retinoic Acid Signaling during Vertebrate Heart Development. J Dev Biol 2019; 7:jdb7020011. [PMID: 31151214 PMCID: PMC6631158 DOI: 10.3390/jdb7020011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 01/07/2023] Open
Abstract
Tightly-regulated levels of retinoic acid (RA) are critical for promoting normal vertebrate development. The extensive history of research on RA has shown that its proper regulation is essential for cardiac progenitor specification and organogenesis. Here, we discuss the roles of RA signaling and its establishment of networks that drive both early and later steps of normal vertebrate heart development. We focus on studies that highlight the drastic effects alternative levels of RA have on early cardiomyocyte (CM) specification and cardiac chamber morphogenesis, consequences of improper RA synthesis and degradation, and known effectors downstream of RA. We conclude with the implications of these findings to our understanding of cardiac regeneration and the etiologies of congenital heart defects.
Collapse
Affiliation(s)
- Eliyahu Perl
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Joshua S Waxman
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
33
|
Nakajima Y. Retinoic acid signaling in heart development. Genesis 2019; 57:e23300. [PMID: 31021052 DOI: 10.1002/dvg.23300] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 12/30/2022]
Abstract
Retinoic acid (RA) is a vitamin A metabolite that acts as a morphogen and teratogen. Excess or defective RA signaling causes developmental defects including in the heart. The heart develops from the anterior lateral plate mesoderm. Cardiogenesis involves successive steps, including formation of the primitive heart tube, cardiac looping, septation, chamber development, coronary vascularization, and completion of the four-chambered heart. RA is dispensable for primitive heart tube formation. Before looping, RA is required to define the anterior/posterior boundaries of the heart-forming mesoderm as well as to form the atrium and sinus venosus. In outflow tract elongation and septation, RA signaling is required to maintain/differentiate cardiogenic progenitors in the second heart field at the posterior pharyngeal arches level. Epicardium-secreted insulin-like growth factor, the expression of which is regulated by hepatic mesoderm-derived erythropoietin under the control of RA, promotes myocardial proliferation of the ventricular wall. Epicardium-derived RA induces the expression of angiogenic factors in the myocardium to form the coronary vasculature. In cardiogenic events at different stages, properly controlled RA signaling is required to establish the functional heart.
Collapse
Affiliation(s)
- Yuji Nakajima
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Osaka City University, Osaka, Japan
| |
Collapse
|
34
|
De Bono C, Thellier C, Bertrand N, Sturny R, Jullian E, Cortes C, Stefanovic S, Zaffran S, Théveniau-Ruissy M, Kelly RG. T-box genes and retinoic acid signaling regulate the segregation of arterial and venous pole progenitor cells in the murine second heart field. Hum Mol Genet 2019; 27:3747-3760. [PMID: 30016433 DOI: 10.1093/hmg/ddy266] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/11/2018] [Indexed: 01/10/2023] Open
Abstract
The arterial and venous poles of the mammalian heart are hotspots of congenital heart defects (CHD) such as those observed in 22q11.2 deletion (or DiGeorge) and Holt-Oram syndromes. These regions of the heart are derived from late differentiating cardiac progenitor cells of the Second Heart Field (SHF) located in pharyngeal mesoderm contiguous with the elongating heart tube. The T-box transcription factor Tbx1, encoded by the major 22q11.2 deletion syndrome gene, regulates SHF addition to both cardiac poles from a common progenitor population. Despite the significance of this cellular addition the mechanisms regulating the deployment of common progenitor cells to alternate cardiac poles remain poorly understood. Here we demonstrate that Tbx5, mutated in Holt-Oram syndrome and essential for venous pole development, is activated in Tbx1 expressing cells in the posterior region of the SHF at early stages of heart tube elongation. A subset of the SHF transcriptional program, including Tbx1 expression, is subsequently downregulated in Tbx5 expressing cells, generating a transcriptional boundary between Tbx1-positive arterial pole and Tbx5-positive venous pole progenitor cell populations. We show that normal downregulation of the definitive arterial pole progenitor cell program in the posterior SHF is dependent on both Tbx1 and Tbx5. Furthermore, retinoic acid (RA) signaling is required for Tbx5 activation in Tbx1-positive cells and blocking RA signaling at the time of Tbx5 activation results in atrioventricular septal defects at fetal stages. Our results reveal sequential steps of cardiac progenitor cell patterning and provide mechanistic insights into the origin of common forms of CHD.
Collapse
Affiliation(s)
| | | | | | - Rachel Sturny
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | - Claudio Cortes
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| | | | | | | | - Robert G Kelly
- Aix-Marseille Univ, CNRS UMR 7288, IBDM, Marseille, France
| |
Collapse
|
35
|
Frank D, Sela-Donenfeld D. Hindbrain induction and patterning during early vertebrate development. Cell Mol Life Sci 2019; 76:941-960. [PMID: 30519881 PMCID: PMC11105337 DOI: 10.1007/s00018-018-2974-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/28/2022]
Abstract
The hindbrain is a key relay hub of the central nervous system (CNS), linking the bilaterally symmetric half-sides of lower and upper CNS centers via an extensive network of neural pathways. Dedicated neural assemblies within the hindbrain control many physiological processes, including respiration, blood pressure, motor coordination and different sensations. During early development, the hindbrain forms metameric segmented units known as rhombomeres along the antero-posterior (AP) axis of the nervous system. These compartmentalized units are highly conserved during vertebrate evolution and act as the template for adult brainstem structure and function. TALE and HOX homeodomain family transcription factors play a key role in the initial induction of the hindbrain and its specification into rhombomeric cell fate identities along the AP axis. Signaling pathways, such as canonical-Wnt, FGF and retinoic acid, play multiple roles to initially induce the hindbrain and regulate Hox gene-family expression to control rhombomeric identity. Additional transcription factors including Krox20, Kreisler and others act both upstream and downstream to Hox genes, modulating their expression and protein activity. In this review, we will examine the earliest embryonic signaling pathways that induce the hindbrain and subsequent rhombomeric segmentation via Hox and other gene expression. We will examine how these signaling pathways and transcription factors interact to activate downstream targets that organize the segmented AP pattern of the embryonic vertebrate hindbrain.
Collapse
Affiliation(s)
- Dale Frank
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, 31096, Haifa, Israel.
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel.
| |
Collapse
|
36
|
Wang S, Huang W, Castillo HA, Kane MA, Xavier-Neto J, Trainor PA, Moise AR. Alterations in retinoic acid signaling affect the development of the mouse coronary vasculature. Dev Dyn 2018; 247:976-991. [PMID: 29806219 DOI: 10.1002/dvdy.24639] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND During the final stages of heart development the myocardium grows and becomes vascularized by means of paracrine factors and cell progenitors derived from the epicardium. There is evidence to suggest that retinoic acid (RA), a metabolite of vitamin A, plays an important role in epicardial-based developmental programming. However, the consequences of altered RA-signaling in coronary development have not been systematically investigated. RESULTS We explored the developmental consequences of altered RA-signaling in late cardiogenic events that involve the epicardium. For this, we used a model of embryonic RA excess based on mouse embryos deficient in the retinaldehyde reductase DHRS3, and a complementary model of embryonic RA deficiency based on pharmacological inhibition of RA synthesis. We found that alterations in embryonic RA signaling led to a thin myocardium and aberrant coronary vessel formation and remodeling. Both excess, and deficient RA-signaling are associated with reductions in ventricular coverage and density of coronary vessels, altered vessel morphology, and impaired recruitment of epicardial-derived mural cells. Using a combined transcriptome and proteome profiling approach, we found that RA treatment of epicardial cells influenced key signaling pathways relevant for cardiac development. CONCLUSIONS Epicardial RA-signaling plays critical roles in the development of the coronary vasculature needed to support myocardial growth. Developmental Dynamics 247:976-991, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Suya Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - Hozana A Castillo
- Brazilian Biosciences National Laboratory, LNBio, Rua Giuseppe Máximo Scolfaro, Polo II de Alta Tecnologia de Campinas, Campinas, SP, Brazil
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland
| | - José Xavier-Neto
- Conselho Nacional do Desenvolvimnto Científico e Tecnológico (Cnpq) CEP 01414000 Cerqueira Cesar Sao Paulo, Sao Paulo, Brazil
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas
| | - Alexander R Moise
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas.,Northern Ontario School of Medicine, Biomolecular Sciences Program and Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
37
|
Wu P, Wang K, Yang Q, Zhou J, Chen D, Ma J, Tang Q, Jin L, Xiao W, Jiang A, Jiang Y, Zhu L, Li M, Li X, Tang G. Identifying SNPs and candidate genes for three litter traits using single-step GWAS across six parities in Landrace and Large White pigs. Physiol Genomics 2018; 50:1026-1035. [PMID: 30289746 DOI: 10.1152/physiolgenomics.00071.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Total number born (TNB), number born alive (NBA), and litter weight born alive (LWB) are critically important traits in pig production. The sow's parity is one of the major factors influencing litter traits. Because of monogenic or polygenic contributions and the presence of temporal gene effects in different sows' parities, it is difficult to clarify the biological and genetic background. To systematically explore the genetic mechanism of litter traits, we conducted 18 GWASs using single-step GWAS (ssGWAS) based on two breeds (908 Landrace and 1,130 Large White sow litter records) for each litter trait in different parities. A total of 300 Landrace and 300 Large White sows were genotyped by sequencing (GBS). ssGWAS was performed separately for each breed and each parity due to population stratification and temporal gene effect. In summary, we identified 80 (15 for Landrace and 65 for Large White), 227 (52 for Landrace, 175 for Large White), and 187 (34 for Landrace, 153 for Large White) single nucleotide polymorphisms (SNPs) affecting TNB, NBA, and LWB, respectively. Of them, we suggest that a total of 22 loci (SSC1: 125098202, SSC1: 117560058, SSC14: 147794697, SSC8: 84823302, SSC9: 143554876, and SSC9: 138766097 for Landrace; SSC1: 4023577, SSC1: 3859573, SSC1: 4891063, SSC16: 5197665, SSC10: 32050819, SSC13: 13552924, SSC13: 92819, SSC17: 3579607, SSC13: 196698221, SSC7: 30918403, SSC16: 46221484, SSC16: 46169204, SSC2: 41988642, SSC2: 44475457, SSC2: 42521875, and SSC7: 58411951 for Large White) are shared by TNB, NBA, and LWB. These results indicate the existence of gene temporal effect in each parity. Furthermore, our findings suggest four interesting candidate genes (FBXL7, ALDH1A2, LEPR, and DDX1) associated with litter traits in different parities that have a major effect on embryonic development progression. In conclusion, 22 crucial SNPs and four interesting candidate genes were identified for three litter traits across six parities. These findings advance our understanding of the genetic architecture of litter traits and confirm the presence of temporal gene effects in different parities. Importantly, functional validation studies for findings of particular interest are recommended in litter traits.
Collapse
Affiliation(s)
- Pingxian Wu
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Kai Wang
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Qiang Yang
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Jie Zhou
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Dejuan Chen
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Jideng Ma
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Qianzi Tang
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Long Jin
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Weihang Xiao
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Anan Jiang
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Yanzhi Jiang
- College of Life Science, Sichuan Agricultural University, Yaan, Sichuan , China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| | - Guoqing Tang
- College of Animal Science and Technology, Sichuan Agricultural University , Chengdu, Sichuan , China
| |
Collapse
|
38
|
Lee JH, Protze SI, Laksman Z, Backx PH, Keller GM. Human Pluripotent Stem Cell-Derived Atrial and Ventricular Cardiomyocytes Develop from Distinct Mesoderm Populations. Cell Stem Cell 2018; 21:179-194.e4. [PMID: 28777944 DOI: 10.1016/j.stem.2017.07.003] [Citation(s) in RCA: 261] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Revised: 05/08/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023]
Abstract
The ability to direct the differentiation of human pluripotent stem cells (hPSCs) to the different cardiomyocyte subtypes is a prerequisite for modeling specific forms of cardiovascular disease in vitro and for developing novel therapies to treat them. Here we have investigated the development of the human atrial and ventricular lineages from hPSCs, and we show that retinoic acid signaling at the mesoderm stage of development is required for atrial specification. Analyses of early developmental stages revealed that ventricular and atrial cardiomyocytes derive from different mesoderm populations that can be distinguished based on CD235a and RALDH2 expression, respectively. Molecular and electrophysiological characterization of the derivative cardiomyocytes revealed that optimal specification of ventricular and atrial cells is dependent on induction of the appropriate mesoderm. Together these findings provide new insights into the development of the human atrial and ventricular lineages that enable the generation of highly enriched, functional cardiomyocyte populations for therapeutic applications.
Collapse
Affiliation(s)
- Jee Hoon Lee
- McEwen Centre for Regenerative Medicine and Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7 Canada
| | - Stephanie I Protze
- McEwen Centre for Regenerative Medicine and Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC V6E 1M7, Canada
| | - Peter H Backx
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; Division of Cardiology and the Peter Munk Cardiac Centre, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Gordon M Keller
- McEwen Centre for Regenerative Medicine and Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7 Canada.
| |
Collapse
|
39
|
Wang S, Yu J, Jones JW, Pierzchalski K, Kane MA, Trainor PA, Xavier-Neto J, Moise AR. Retinoic acid signaling promotes the cytoskeletal rearrangement of embryonic epicardial cells. FASEB J 2018; 32:3765-3781. [PMID: 29447006 DOI: 10.1096/fj.201701038r] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
All- trans-retinoic acid (RA), a vitamin A metabolite, is an important signaling molecule required for the proper development of the heart. The epicardium is the main source of RA in the embryonic heart, yet the cardiogenic functions of epicardial-produced RA are not fully understood. Here, we investigated the roles of RA signaling in the embryonic epicardium using in vivo and in vitro models of excess or deficiency of RA. Our results suggested that RA signaling facilitates the cytoskeletal rearrangement required for the epicardial-to-mesenchymal transition of epicardial cells. In vivo treatment with an inhibitor of RA synthesis delayed the migration of epicardial-derived precursor cells (EPDCs) into the myocardium; the opposite was seen in the case of dehydrogenase/reductase superfamily (DHRS)3-deficient embryos, a mouse model of RA excess. Analysis of the behavior of epicardial cells exposed to RA receptor agonists or inhibitors of RA synthesis in vitro revealed that appropriate levels of RA are important in orchestrating the platelet-derived growth factor-induced loss of epithelial character, cytoskeletal remodeling, and migration, necessary for the infiltration of the myocardium by EPDCs. To understand the molecular mechanisms by which RA regulates epicardial cytoskeletal rearrangement, we used a whole transcriptome profiling approach, which in combination with pull-down and inhibition assays, demonstrated that the Ras homolog gene family, member A (RhoA) pathway is required for the morphologic changes induced by RA in epicardial cells. Collectively, these data demonstrate that RA regulates the cytoskeletal rearrangement of epicardial cells via a signaling cascade that involves the RhoA pathway.-Wang, S., Yu, J., Jones, J. W., Pierzchalski, K., Kane, M. A., Trainor, P. A., Xavier-Neto, J., Moise, A. R. Retinoic acid signaling promotes the cytoskeletal rearrangement of embryonic epicardial cells.
Collapse
Affiliation(s)
- Suya Wang
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas, USA
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jace W Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Keely Pierzchalski
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, USA.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - José Xavier-Neto
- Conselho Nacional do Desenvolvimento Científico e Tecnológico (CNPq) Sao Paulo, Brazil; and
| | - Alexander R Moise
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas, USA.,Northern Ontario School of Medicine, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
40
|
Li J, Miao L, Zhao C, Shaikh Qureshi WM, Shieh D, Guo H, Lu Y, Hu S, Huang A, Zhang L, Cai CL, Wan LQ, Xin H, Vincent P, Singer HA, Zheng Y, Cleaver O, Fan ZC, Wu M. CDC42 is required for epicardial and pro-epicardial development by mediating FGF receptor trafficking to the plasma membrane. Development 2017; 144:1635-1647. [PMID: 28465335 PMCID: PMC5450847 DOI: 10.1242/dev.147173] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/16/2017] [Indexed: 01/26/2023]
Abstract
The epicardium contributes to multiple cardiac lineages and is essential for cardiac development and regeneration. However, the mechanism of epicardium formation is unclear. This study aimed to establish the cellular and molecular mechanisms underlying the dissociation of pro-epicardial cells (PECs) from the pro-epicardium (PE) and their subsequent translocation to the heart to form the epicardium. We used lineage tracing, conditional deletion, mosaic analysis and ligand stimulation in mice to determine that both villous protrusions and floating cysts contribute to PEC translocation to myocardium in a CDC42-dependent manner. We resolved a controversy by demonstrating that physical contact of the PE with the myocardium constitutes a third mechanism for PEC translocation to myocardium, and observed a fourth mechanism in which PECs migrate along the surface of the inflow tract to reach the ventricles. Epicardial-specific Cdc42 deletion disrupted epicardium formation, and Cdc42 null PECs proliferated less, lost polarity and failed to form villous protrusions and floating cysts. FGF signaling promotes epicardium formation in vivo, and biochemical studies demonstrated that CDC42 is involved in the trafficking of FGF receptors to the cell membrane to regulate epicardium formation. Highlighted article: During epicardial formation in mice, four different mechanisms of pro-epicardial cell translocation to the myocardium can be identified, with CDC42 playing a key role.
Collapse
Affiliation(s)
- Jingjing Li
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Lianjie Miao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.,Institute of Translational Medicine, Nanchang University, Nanchang 330031, China.,School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Chen Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | | | - David Shieh
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Hua Guo
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Yangyang Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Saiyang Hu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alice Huang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Lu Zhang
- Developmental and Regenerative Biology, Mount Sinai Hospital, New York, NY 10029, USA
| | - Chen-Leng Cai
- Developmental and Regenerative Biology, Mount Sinai Hospital, New York, NY 10029, USA
| | - Leo Q Wan
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th street, Biotech 2147, Troy, NY 12180, USA
| | - Hongbo Xin
- Institute of Translational Medicine, Nanchang University, Nanchang 330031, China.,School of Life Sciences, Nanchang University, Nanchang 330031, China
| | - Peter Vincent
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ondine Cleaver
- Molecular Biology, UT Southwestern, Dallas, TX 75390, USA
| | - Zhen-Chuan Fan
- International Collaborative Research Center for Health Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Mingfu Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| |
Collapse
|
41
|
Chu M, Zhu X, Wang C, Rong J, Wang Y, Wang S, Xing B, Tao Y, Zhuang X, Jiang L. The rs4238326 polymorphism in ALDH1A2 gene potentially associated with non-post traumatic knee osteoarthritis susceptibility: a two-stage population-based study. Osteoarthritis Cartilage 2017; 25:1062-1067. [PMID: 28089900 DOI: 10.1016/j.joca.2017.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 12/27/2016] [Accepted: 01/06/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE A recent genome-wide association study reported significant associations of genetic variants within the ALDH1A2 gene with osteoarthritis (OA) of the hand in European populations. However, these findings have not been well generalized to other joints, or to other populations. METHODS We performed a two-stage population-based case-control study including 196 non-post traumatic knee OA cases and 442 controls in the first stage and independent 143 non-post traumatic knee OA cases and 238 controls in the second stage in a Chinese population by genotyping eight tagging polymorphisms in ALDH1A2. RESULTS In the first stage, the single nucleotide polymorphism (SNP) rs4238326 was found to be potentially associated with knee OA risk (additive model: odds ratio [OR] = 0.70; 95% confidence interval [95% CI] = 0.49-1.01; P = 0.055), which was further confirmed in the second stage with similar effect (additive model: OR = 0.60; 95% CI = 0.38-0.95; P = 0.029). After combining the two stages, we found that the variant C allele of rs4238326 was probably associated with decreased risk of knee OA (additive model: OR = 0.65; 95% CI = 0.49-0.86; P = 0.003). Furthermore, interaction analyses showed that rs4238326 interacted multiplicatively with age to contribute to knee OA risk (interaction P = 0.041). CONCLUSIONS These findings indicate that the SNP rs4238326 in ALDH1A2 gene may potentially modify individual susceptibility to knee OA in the Chinese population. Beyond that, further studies are warranted to validate and extend our findings, and future functional studies are required to clarify the possible mechanisms.
Collapse
Affiliation(s)
- M Chu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu Province, PR China
| | - X Zhu
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu Province, PR China
| | - C Wang
- College of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu Province, PR China
| | - J Rong
- Second Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - Y Wang
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu Province, PR China
| | - S Wang
- Second Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - B Xing
- Hongqi Community Health Service Center, Xiangfang District, Harbin, Heilongjiang Province, PR China
| | - Y Tao
- Department of Health Education, Public Health College, Harbin Medical University, Harbin, Heilongjiang Province, PR China
| | - X Zhuang
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu Province, PR China
| | - L Jiang
- Department of Epidemiology, School of Public Health, Nantong University, Nantong, Jiangsu Province, PR China.
| |
Collapse
|
42
|
Stefanovic S, Zaffran S. Mechanisms of retinoic acid signaling during cardiogenesis. Mech Dev 2016; 143:9-19. [PMID: 28007475 DOI: 10.1016/j.mod.2016.12.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
Abstract
Substantial experimental and epidemiological data have highlighted the interplay between nutritional and genetic factors in the development of congenital heart defects. Retinoic acid (RA), a derivative of vitamin A, plays a key role during vertebrate development including the formation of the heart. Retinoids bind to RA and retinoid X receptors (RARs and RXRs) which then regulate tissue-specific genes. Here, we will focus on the roles of RA signaling and receptors in gene regulation during cardiogenesis, and the consequence of deregulated retinoid signaling on heart formation and congenital heart defects.
Collapse
|
43
|
Metzler MA, Sandell LL. Enzymatic Metabolism of Vitamin A in Developing Vertebrate Embryos. Nutrients 2016; 8:E812. [PMID: 27983671 PMCID: PMC5188467 DOI: 10.3390/nu8120812] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022] Open
Abstract
Embryonic development is orchestrated by a small number of signaling pathways, one of which is the retinoic acid (RA) signaling pathway. Vitamin A is essential for vertebrate embryonic development because it is the molecular precursor of the essential signaling molecule RA. The level and distribution of RA signaling within a developing embryo must be tightly regulated; too much, or too little, or abnormal distribution, all disrupt embryonic development. Precise regulation of RA signaling during embryogenesis is achieved by proteins involved in vitamin A metabolism, retinoid transport, nuclear signaling, and RA catabolism. The reversible first step in conversion of the precursor vitamin A to the active retinoid RA is mediated by retinol dehydrogenase 10 (RDH10) and dehydrogenase/reductase (SDR family) member 3 (DHRS3), two related membrane-bound proteins that functionally activate each other to mediate the interconversion of retinol and retinal. Alcohol dehydrogenase (ADH) enzymes do not contribute to RA production under normal conditions during embryogenesis. Genes involved in vitamin A metabolism and RA catabolism are expressed in tissue-specific patterns and are subject to feedback regulation. Mutations in genes encoding these proteins disrupt morphogenesis of many systems in a developing embryo. Together these observations demonstrate the importance of vitamin A metabolism in regulating RA signaling during embryonic development in vertebrates.
Collapse
Affiliation(s)
- Melissa A Metzler
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| | - Lisa L Sandell
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| |
Collapse
|
44
|
Calderon D, Bardot E, Dubois N. Probing early heart development to instruct stem cell differentiation strategies. Dev Dyn 2016; 245:1130-1144. [PMID: 27580352 DOI: 10.1002/dvdy.24441] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/20/2016] [Accepted: 08/20/2016] [Indexed: 12/19/2022] Open
Abstract
Scientists have studied organs and their development for centuries and, along that path, described models and mechanisms explaining the developmental principles of organogenesis. In particular, with respect to the heart, new fundamental discoveries are reported continuously that keep changing the way we think about early cardiac development. These discoveries are driven by the need to answer long-standing questions regarding the origin of the earliest cells specified to the cardiac lineage, the differentiation potential of distinct cardiac progenitor cells, and, very importantly, the molecular mechanisms underlying these specification events. As evidenced by numerous examples, the wealth of developmental knowledge collected over the years has had an invaluable impact on establishing efficient strategies to generate cardiovascular cell types ex vivo, from either pluripotent stem cells or via direct reprogramming approaches. The ability to generate functional cardiovascular cells in an efficient and reliable manner will contribute to therapeutic strategies aimed at alleviating the increasing burden of cardiovascular disease and morbidity. Here we will discuss the recent discoveries in the field of cardiac progenitor biology and their translation to the pluripotent stem cell model to illustrate how developmental concepts have instructed regenerative model systems in the past and promise to do so in the future. Developmental Dynamics 245:1130-1144, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Damelys Calderon
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Evan Bardot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| | - Nicole Dubois
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, NY, USA.,Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, NY, USA.,Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, NY, USA
| |
Collapse
|
45
|
Cano E, Carmona R, Velecela V, Martínez-Estrada O, Muñoz-Chápuli R. The proepicardium keeps a potential for glomerular marker expression which supports its evolutionary origin from the pronephros. Evol Dev 2016; 17:224-30. [PMID: 26174098 DOI: 10.1111/ede.12130] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The proepicardium is the embryonic primordium of the epicardium. This transient structure is essential for cardiac development giving rise to the epicardium and supplying the heart with vascular and cardiac connective tissue progenitors. However, their nature and evolutionary origin are poorly-known. We have suggested elsewhere (Pombal et al. Evol. Dev. 10: 210-216, 2008; Cano et al., J. Dev. Biol. 1: 3-19, 2013) that the proepicardium is an evolutionary derivative of the primordium of an ancient external pronephric glomerulus, devoid of its original excretory function. In this study, we describe for the first time expression of two podocyte markers in the chick proepicardium (glepp1 and synaptopodin) and we have shown how these podocyte markers as well as the intermediate mesoderm marker Pax2 are strongly upregulated when the proepicardium is cultured with nephrogenic inducers. Retinoic acid treatment also induced in the proepicardium expression of Hoxb4, a gene which confers to intermediate mesoderm competence to respond to nephrogenic signals. Thus, a latent nephrogenic potential persists in the proepicardium and also that its original glomerular fate can be partially rescued. The transcription factor Wt1, essential for kidney and epicardial development, plays opposite roles in both tissues, inducing epithelial-mesenchymal transition in the proepicardium and promoting epithelialization in the kidneys (Essafi et al., Dev. Cell 21: 559-574, 2011). Consistently with this antithetical function of Wt1, we have observed an upregulation of podocalyxin in the epicardium of mouse embryos with conditional deletion of the Wt1 gene, while this protein is transcriptionally activated by Wt1 in podocytes.
Collapse
Affiliation(s)
- Elena Cano
- Department of Animal Biology, University of Málaga, Faculty of Science, Málaga 29071, Spain.,Andalusian Center for Nanomedicine and Biotechnology (BIONAND), Severo Ochoa n°25, Campanillas 29590, Spain.,Department of Vertebrate Genomics and Cardiovascular Genetics, Group of Cardiovascular Genetics, Experimental and Clinical Research Center. Charité-Universitätsmedizin Berlin and Max Delbrück Center (MDC) for Molecular Medicine. Lindenberger Weg 80, Berlin 13125, Germany
| | - Rita Carmona
- Department of Animal Biology, University of Málaga, Faculty of Science, Málaga 29071, Spain.,Andalusian Center for Nanomedicine and Biotechnology (BIONAND), Severo Ochoa n°25, Campanillas 29590, Spain
| | - Víctor Velecela
- Department of Cell Biology, Faculty of Biology, University of Barcelona. Av. Diagonal, 643, Barcelona 08028, Spain
| | - Ofelia Martínez-Estrada
- Department of Cell Biology, Faculty of Biology, University of Barcelona. Av. Diagonal, 643, Barcelona 08028, Spain
| | - Ramón Muñoz-Chápuli
- Department of Animal Biology, University of Málaga, Faculty of Science, Málaga 29071, Spain.,Andalusian Center for Nanomedicine and Biotechnology (BIONAND), Severo Ochoa n°25, Campanillas 29590, Spain
| |
Collapse
|
46
|
Lighthouse JK, Small EM. Transcriptional control of cardiac fibroblast plasticity. J Mol Cell Cardiol 2016; 91:52-60. [PMID: 26721596 PMCID: PMC4764462 DOI: 10.1016/j.yjmcc.2015.12.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/15/2015] [Accepted: 12/20/2015] [Indexed: 12/11/2022]
Abstract
Cardiac fibroblasts help maintain the normal architecture of the healthy heart and are responsible for scar formation and the healing response to pathological insults. Various genetic, biomechanical, or humoral factors stimulate fibroblasts to become contractile smooth muscle-like cells called myofibroblasts that secrete large amounts of extracellular matrix. Unfortunately, unchecked myofibroblast activation in heart disease leads to pathological fibrosis, which is a major risk factor for the development of cardiac arrhythmias and heart failure. A better understanding of the molecular mechanisms that control fibroblast plasticity and myofibroblast activation is essential to develop novel strategies to specifically target pathological cardiac fibrosis without disrupting the adaptive healing response. This review highlights the major transcriptional mediators of fibroblast origin and function in development and disease. The contribution of the fetal epicardial gene program will be discussed in the context of fibroblast origin in development and following injury, primarily focusing on Tcf21 and C/EBP. We will also highlight the major transcriptional regulatory axes that control fibroblast plasticity in the adult heart, including transforming growth factor β (TGFβ)/Smad signaling, the Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF) axis, and Calcineurin/transient receptor potential channel (TRP)/nuclear factor of activated T-Cell (NFAT) signaling. Finally, we will discuss recent strategies to divert the fibroblast transcriptional program in an effort to promote cardiomyocyte regeneration. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling".
Collapse
Affiliation(s)
- Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Eric M Small
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA.
| |
Collapse
|
47
|
El Robrini N, Etchevers HC, Ryckebüsch L, Faure E, Eudes N, Niederreither K, Zaffran S, Bertrand N. Cardiac outflow morphogenesis depends on effects of retinoic acid signaling on multiple cell lineages. Dev Dyn 2015; 245:388-401. [PMID: 26442704 DOI: 10.1002/dvdy.24357] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 08/20/2015] [Accepted: 09/27/2015] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Retinoic acid (RA), the bioactive derivative of vitamin A, is essential for vertebrate heart development. Both excess and reduced RA signaling lead to cardiovascular malformations affecting the outflow tract (OFT). To address the cellular mechanisms underlying the effects of RA signaling during OFT morphogenesis, we used transient maternal RA supplementation to rescue the early lethality resulting from inactivation of the murine retinaldehyde dehydrogenase 2 (Raldh2) gene. RESULTS By embryonic day 13.5, all rescued Raldh2(-/-) hearts exhibit severe, reproducible OFT septation defects, although wild-type and Raldh2(+/-) littermates have normal hearts. Cardiac neural crest cells (cNCC) were present in OFT cushions of Raldh2(-/-) mutant embryos but ectopically located in the periphery of the endocardial cushions, rather than immediately underlying the endocardium. Excess mesenchyme was generated by Raldh2(-/-) mutant endocardium, which displaced cNCC derivatives from their subendocardial, medial position. CONCLUSIONS RA signaling affects not only cNCC numbers but also their position relative to endocardial mesenchyme during the septation process. Our study shows that inappropriate coordination between the different cell types of the OFT perturbs its morphogenesis and leads to a severe congenital heart defect, persistent truncus arteriosus.
Collapse
Affiliation(s)
- Nicolas El Robrini
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Heather C Etchevers
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Lucile Ryckebüsch
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Emilie Faure
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Nathalie Eudes
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Karen Niederreither
- CNRS UMR 7104, INSERM U964, IGBMC, University of Strasbourg, Illkirch, France
| | - Stéphane Zaffran
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| | - Nicolas Bertrand
- Aix-Marseille University, GMGF, UMR_S910, Faculté de Médecine, Marseille, France.,INSERM U910, Faculté de Médecine, Marseille, France
| |
Collapse
|
48
|
Li ASW, Marikawa Y. An in vitro gastrulation model recapitulates the morphogenetic impact of pharmacological inhibitors of developmental signaling pathways. Mol Reprod Dev 2015; 82:1015-36. [PMID: 26387793 DOI: 10.1002/mrd.22585] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/17/2015] [Indexed: 12/21/2022]
Abstract
Certain chemical agents act as teratogens, causing birth defects and fetal deaths when pregnant women are exposed to them. The establishment of in vitro models that recapitulate crucial embryonic events is therefore vital to facilitate screening of potential teratogens. Previously, we created a three-dimensional culture method for mouse P19C5 embryonal carcinoma stem cells that, when cultured as embryoid bodies, display elongation morphogenesis resembling gastrulation, which is the critical event resulting in the germ layers and major body axes. Determination of how well this in vitro morphogenesis represents in vivo gastrulation is essential to assess its applicability as well as to identify limitations of the model for detecting teratogenic agents. Here, we investigated the morphological and molecular characteristics of P19C5 morphogenesis using pharmacological agents that are known to cause abnormal patterning in the embryo in vivo by inhibiting major developmental signaling--e.g., involving Wnt, Nodal, Bone morphogenic protein (Bmp), Fibroblast growth factor (Fgf), Retinoic acid, Notch, and Hedgehog pathways. Inhibitors of Wnt, Nodal, Bmp, Fgf, and Retinoic acid signaling caused distinct changes in P19C5 morphogenesis that were quantifiable using morphometric parameters. These five inhibitors, plus the Notch inhibitor, also altered temporal expression profiles of developmental regulator genes in a manner consistent with the in vivo roles of the corresponding signaling pathways. In contrast, the Hedgehog inhibitor did not have any impact on the process, suggesting an absence of active Hedgehog signaling in these embryoid bodies. These results indicate that the P19C5 in vitro gastrulation model is a promising tool to screen for teratogenic agents that interfere with many of the key developmental signals.
Collapse
Affiliation(s)
- Aileen S W Li
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Hawaii
| | - Yusuke Marikawa
- Developmental and Reproductive Biology Graduate Program, Institute for Biogenesis Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Hawaii
| |
Collapse
|
49
|
Rao KS, Aronshtam A, McElory-Yaggy KL, Bakondi B, VanBuren P, Sobel BE, Spees JL. Human epicardial cell-conditioned medium contains HGF/IgG complexes that phosphorylate RYK and protect against vascular injury. Cardiovasc Res 2015; 107:277-86. [PMID: 26025956 DOI: 10.1093/cvr/cvv168] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 05/21/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS The aim of this study was to evaluate the paracrine activity of human epicardial-derived cells (hEPDCs) to screen for secreted vasoprotective factors and develop therapeutics to treat vascular reperfusion injury. METHODS AND RESULTS Epicardial cells support cardiac development, repair, and remodelling after injury in part, through paracrine activity. We hypothesized that secreted ligands from hEPDCs would protect vascular integrity after myocardial infarction (MI) with reperfusion. During simulated ischaemia in culture (24-48 h), concentrated hEPDC-conditioned medium (EPI CdM) increased survival of primary cardiac endothelial cells. In a rat MI model, EPI CdM treatment reduced vascular injury in vivo after reperfusion. By phospho-receptor tyrosine kinase (RTK) arrays, ELISA, and neutralizing antibody screens, we identified hepatocyte growth factor (HGF) as a key vasoprotective factor in EPI CdM. Unexpectedly, we observed that some of the HGF in EPI CdM formed complexes with polyclonal IgG. Following reperfusion, preparations of HGF/IgG complexes provided greater vascular protection than free HGF with IgG. HGF/IgG complexes localized to blood vessels in vivo and increased HGF retention time after administration. In subsequent screens, we found that 'related to tyrosine kinase' (RYK) receptor was phosphorylated after exposure of cardiac endothelial cells to HGF/IgG complexes, but not to free HGF with IgG. The enhanced protection conferred by HGF/IgG complexes was lost after antibody blockade of RYK. Notably, the HGF/IgG complex is the first 'ligand' shown to promote phosphorylation of RYK. CONCLUSION Early treatment with HGF/IgG complexes after myocardial ischaemia with reperfusion may rescue tissue through vasoprotection conferred by c-Met and RYK signalling.
Collapse
Affiliation(s)
- Krithika S Rao
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Colchester, VT, USA Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Alexander Aronshtam
- Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Keara L McElory-Yaggy
- Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Benjamin Bakondi
- Cellular, Molecular and Biomedical Sciences Graduate Program, University of Vermont, Colchester, VT, USA Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Peter VanBuren
- Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Burton E Sobel
- Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA
| | - Jeffrey L Spees
- Department of Medicine and Cardiovascular Research Institute, University of Vermont, 208 South Park Drive, Ste 2, Colchester, VT 05446, USA Stem Cell Core, University of Vermont, Colchester, VT 05446, USA
| |
Collapse
|
50
|
Moazzen H, Lu X, Liu M, Feng Q. Pregestational diabetes induces fetal coronary artery malformation via reactive oxygen species signaling. Diabetes 2015; 64:1431-43. [PMID: 25422104 DOI: 10.2337/db14-0190] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Hypoplastic coronary artery disease is a congenital coronary artery malformation associated with a high risk of sudden cardiac death. However, the etiology and pathogenesis of hypoplastic coronary artery disease remain undefined. Pregestational diabetes increases reactive oxygen species (ROS) levels and the risk of congenital heart defects. We show that pregestational diabetes in mice induced by streptozotocin significantly increased 4-hydroxynonenal production and decreased coronary artery volume in fetal hearts. Pregestational diabetes also impaired epicardial epithelial-to-mesenchymal transition (EMT) as shown by analyses of the epicardium, epicardial-derived cells, and fate mapping. Additionally, the expression of hypoxia-inducible factor 1α (Hif-1α), Snail1, Slug, basic fibroblast growth factor (bFgf), and retinaldehyde dehydrogenase (Aldh1a2) was decreased and E-cadherin expression was increased in the hearts of fetuses of diabetic mothers. Of note, these abnormalities were all rescued by treatment with N-acetylcysteine (NAC) in diabetic females during gestation. Ex vivo analysis showed that high glucose levels inhibited epicardial EMT, which was reversed by NAC treatment. We conclude that pregestational diabetes in mice can cause coronary artery malformation through ROS signaling. This study may provide a rationale for further clinical studies to investigate whether pregestational diabetes could cause hypoplastic coronary artery disease in humans.
Collapse
Affiliation(s)
- Hoda Moazzen
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Xiangru Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Murong Liu
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Qingping Feng
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada Department of Medicine, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|