1
|
Nakamura M, Huang GN. Why some hearts heal and others don't: The phylogenetic landscape of cardiac regenerative capacity. Semin Cell Dev Biol 2025; 170:103609. [PMID: 40220599 DOI: 10.1016/j.semcdb.2025.103609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
The limited ability of adult humans to replenish lost heart muscle cells after a heart attack has attracted scientists to explore natural heart regeneration capabilities in the animal kingdom. In particular, research has accelerated since the landmark discovery more than twenty years ago that zebrafish can completely regrow myocardial tissue. In this review, we survey heart regeneration studies in diverse model and non-model animals, aiming to gain insights into both the evolutionary trends in cardiac regenerative potential and the variations among closely related species. Differences in cardiomyogenesis, vasculature formation, and the communication between cardiovascular cells and other players have been investigated to understand the cellular basis, although the precise molecular and genetic causes underlying the stark differences in cardiac regenerative potential among certain close cousins remain largely unknown. By studying cardiovascular regeneration and repair in diverse organisms, we may uncover distinct mechanisms, offering new perspectives for advancing regenerative medicine.
Collapse
Affiliation(s)
- Makoto Nakamura
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Chen C, Liu W, Yuan F, Wang X, Xu X, Ling CC, Ge X, Shen X, Li B, Shen Y, Liu D. G protein-coupled receptor GPR182 negatively regulates sprouting angiogenesis via modulating CXCL12-CXCR4 axis signaling. Angiogenesis 2025; 28:25. [PMID: 40314798 PMCID: PMC12048421 DOI: 10.1007/s10456-025-09977-5] [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: 10/28/2024] [Accepted: 03/25/2025] [Indexed: 05/03/2025]
Abstract
Angiogenesis is a critical process for tumor progression, regulated by various signaling pathways. Although antiangiogenic therapies targeting the VEGF pathway have shown potential, their effectiveness is inconsistent across different tumor types. GPR182, an endothelial cell-specific G protein-coupled receptor, is frequently downregulated in hypervascular tumors, but its specific role in angiogenesis has not been well defined. Our study reveals that GPR182 expression is markedly reduced in hepatocellular carcinoma (HCC) and inversely correlates with CD31, a pan-endothelial marker. In zebrafish embryos, Gpr182 deficiency resulted in enhanced angiogenic sprouting and hypervascularization, and GPR182-deficient human umbilical vein endothelial cells (HUVECs) showed increased migration and proliferation. At the molecular level, GPR182 acts as a decoy receptor, binding CXCL12 and regulating its gradient, which in turn suppresses CXCR4-mediated angiogenesis. The pharmacological blockade of CXCR4 with AMD3100 corrected the abnormal angiogenic phenotype in Gpr182-deficient zebrafish embryos and in the livers of a zebrafish HCC model. This work uncovers GPR182 as a negative regulator of angiogenesis, a key process in tumor growth and metastasis, and proposes that targeting GPR182 may offer a novel therapeutic approach for antiangiogenic strategies in cancer treatment.
Collapse
MESH Headings
- Receptors, CXCR4/metabolism
- Receptors, CXCR4/genetics
- Animals
- Zebrafish/embryology
- Humans
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Chemokine CXCL12/metabolism
- Chemokine CXCL12/genetics
- Signal Transduction
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/genetics
- Zebrafish Proteins/metabolism
- Zebrafish Proteins/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Human Umbilical Vein Endothelial Cells/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/blood supply
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Neovascularization, Physiologic
- Cell Movement
- Cell Proliferation
- Angiogenesis
Collapse
Affiliation(s)
- Changsheng Chen
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China.
| | - Wei Liu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Medical Research Institute, Wuhan University, Wuhan, Hubei Province, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, Hubei Province, China
| | - Fang Yuan
- Medical College of Nantong University, Nantong, Jiangsu Province, China
- Huai'an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai'an, Jiangsu Province, China
| | - Xiaoning Wang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xi Xu
- Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Chang Chun Ling
- Department of Intervention and Vascular Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiaojuan Ge
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China
| | - Xiaozhong Shen
- Medical College of Nantong University, Nantong, Jiangsu Province, China
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Li
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China
| | - Yuqian Shen
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China
- Department of Translational Medicine, IGBMC, INSERM U964, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Dong Liu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Seyuan Road 9, Nantong, Jiangsu Province, 226019, China.
- Medical College of Nantong University, Nantong, Jiangsu Province, China.
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
| |
Collapse
|
3
|
Ghosh S, Bishnoi B, Das S. Artery regeneration: Molecules, mechanisms and impact on organ function. Semin Cell Dev Biol 2025; 171:103611. [PMID: 40318557 DOI: 10.1016/j.semcdb.2025.103611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 03/25/2025] [Indexed: 05/07/2025]
Abstract
Replenishment of artery cells to repair or create new arteries is a promising strategy to re-vascularize ischemic tissue. However, limited understanding of cellular and molecular programs associated with artery (re-)growth impedes our efforts towards designing optimal therapeutic approaches. In this review, we summarize different cellular mechanisms that drive injury-induced artery regeneration in distinct organs and organisms. Artery formation during embryogenesis includes migration, self-amplification, and changes in cell fates. These processes are coordinated by multiple signaling pathways, like Vegf, Wnt, Notch, Cxcr4; many of which, also involved in injury-induced vascular responses. We also highlight how physiological and environmental factors determine the extent of arterial re-vascularization. Finally, we discuss different in vitro cellular reprogramming and tissue engineering approaches to promote artery regeneration, in vivo. This review provides the current understanding of endothelial cell fate reprogramming and explores avenues for regenerating arteries to restore organ function through efficient revascularization.
Collapse
Affiliation(s)
- Swarnadip Ghosh
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Bhavnesh Bishnoi
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India
| | - Soumyashree Das
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru, KA 560065, India.
| |
Collapse
|
4
|
Li L, Lu M, Guo L, Zhang X, Liu Q, Zhang M, Gao J, Xu M, Lu Y, Zhang F, Li Y, Zhang R, Liu X, Pan S, Zhang X, Li Z, Chen Y, Su X, Zhang N, Guo W, Yang T, Chen J, Qin Y, Zhang Z, Cui W, Yu L, Gu Y, Yang H, Xu X, Wang J, Burns CE, Burns CG, Han K, Zhao L, Fan G, Su Y. An organ-wide spatiotemporal transcriptomic and cellular atlas of the regenerating zebrafish heart. Nat Commun 2025; 16:3716. [PMID: 40253397 PMCID: PMC12009352 DOI: 10.1038/s41467-025-59070-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Adult zebrafish robustly regenerate injured hearts through a complex orchestration of molecular and cellular activities. However, this remarkable process, which is largely non-existent in humans, remains incompletely understood. Here, we utilize integrated spatial transcriptomics (Stereo-seq) and single-cell RNA-sequencing (scRNA-seq) to generate a spatially-resolved molecular and cellular atlas of regenerating zebrafish heart across eight stages. We characterize the cascade of cardiomyocyte cell states responsible for producing regenerated myocardium and explore a potential role for tpm4a in cardiomyocyte re-differentiation. Moreover, we uncover the activation of ifrd1 and atp6ap2 genes as a unique feature of regenerative hearts. Lastly, we reconstruct a 4D "virtual regenerating heart" comprising 569,896 cells/spots derived from 36 scRNA-seq libraries and 224 Stereo-seq slices. Our comprehensive atlas serves as a valuable resource to the cardiovascular and regeneration scientific communities and their ongoing efforts to understand the molecular and cellular mechanisms underlying vertebrate heart regeneration.
Collapse
Affiliation(s)
- Lei Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Meina Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Lidong Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qun Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Meiling Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Junying Gao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Mengyang Xu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yijian Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yao Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Ruihua Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiawei Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Shanshan Pan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xianghui Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Zhen Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Yadong Chen
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiaoshan Su
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Nannan Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Wenjie Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Yating Qin
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Wei Cui
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Lindong Yu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Ying Gu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Han
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark.
| | - Long Zhao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Fisheries, Ocean University of China, Qingdao, 266003, China.
| | - Guangyi Fan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
- BGI Research, Hangzhou, 310030, China.
| | - Ying Su
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
5
|
Rios Coronado PE, Zhou J, Fan X, Zanetti D, Naftaly JA, Prabala P, Martínez Jaimes AM, Farah EN, Kundu S, Deshpande SS, Evergreen I, Kho PF, Ma Q, Hilliard AT, Abramowitz S, Pyarajan S, Dochtermann D, Damrauer SM, Chang KM, Levin MG, Winn VD, Paşca AM, Plomondon ME, Waldo SW, Tsao PS, Kundaje A, Chi NC, Clarke SL, Red-Horse K, Assimes TL. CXCL12 drives natural variation in coronary artery anatomy across diverse populations. Cell 2025; 188:1784-1806.e22. [PMID: 40049164 PMCID: PMC12029448 DOI: 10.1016/j.cell.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 03/12/2025]
Abstract
Coronary arteries have a specific branching pattern crucial for oxygenating heart muscle. Among humans, there is natural variation in coronary anatomy with respect to perfusion of the inferior/posterior left heart, which can branch from either the right arterial tree, the left, or both-a phenotype known as coronary dominance. Using angiographic data for >60,000 US veterans of diverse ancestry, we conducted a genome-wide association study of coronary dominance, revealing moderate heritability and identifying ten significant loci. The strongest association occurred near CXCL12 in both European- and African-ancestry cohorts, with downstream analyses implicating effects on CXCL12 expression. We show that CXCL12 is expressed in human fetal hearts at the time dominance is established. Reducing Cxcl12 in mice altered coronary dominance and caused septal arteries to develop away from Cxcl12 expression domains. These findings indicate that CXCL12 patterns human coronary arteries, paving the way for "medical revascularization" through targeting developmental pathways.
Collapse
Affiliation(s)
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Xiaochen Fan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Daniela Zanetti
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Institute of Genetic and Biomedical Research, National Research Council, Cagliari, Sardinia, Italy
| | | | - Pratima Prabala
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Azalia M Martínez Jaimes
- Department of Biology, Stanford University, Stanford, CA, USA; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elie N Farah
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Salil S Deshpande
- Institute for Computational and Mathematical Engineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivy Evergreen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qixuan Ma
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | | | - Sarah Abramowitz
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Sarnoff Cardiovascular Research Foundation, McLean, VA, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Daniel Dochtermann
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA, USA
| | - Scott M Damrauer
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA; Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michael G Levin
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anca M Paşca
- Department of Pediatrics, Neonatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mary E Plomondon
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA
| | - Stephen W Waldo
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA; CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA; Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Neil C Chi
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Shoa L Clarke
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Themistocles L Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
6
|
Chiba A, Yamamoto T, Fukui H, Fukumoto M, Shirai M, Nakajima H, Mochizuki N. Zonated Wnt/β-catenin signal-activated cardiomyocytes at the atrioventricular canal promote coronary vessel formation in zebrafish. Dev Cell 2025; 60:21-29.e8. [PMID: 39395410 DOI: 10.1016/j.devcel.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/18/2024] [Accepted: 09/11/2024] [Indexed: 10/14/2024]
Abstract
Cells functioning at a specific zone by clustering according to gene expression are recognized as zonated cells. Here, we demonstrate anatomical and functional zones in the zebrafish heart. The cardiomyocytes (CMs) at the atrioventricular canal between the atrium and ventricle could be grouped into three zones according to the localization of signal-activated CMs: Wnt/β-catenin signal+, Bmp signal+, and Tbx2b+ zones. Endocardial endothelial cells (ECs) changed their characteristics, penetrated the Wnt/β-catenin signal+ CM zone, and became coronary ECs covering the heart. Coronary vessel length was reduced when the Wnt/β-catenin signal+ CMs were depleted. Collectively, we demonstrate the importance of anatomical and functional zonation of CMs in the zebrafish heart.
Collapse
Affiliation(s)
- Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan; Department of Pharmacology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan; Medical-Risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan; Division of Biomechanics and Signaling, Institute of Advanced Medical Sciences, Tokushima University, Tokushima 770-8503, Japan
| | - Moe Fukumoto
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Manabu Shirai
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 564-8565, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan.
| |
Collapse
|
7
|
Wang ZY, Mehra A, Wang QC, Gupta S, Ribeiro da Silva A, Juan T, Günther S, Looso M, Detleffsen J, Stainier DYR, Marín-Juez R. flt1 inactivation promotes zebrafish cardiac regeneration by enhancing endothelial activity and limiting the fibrotic response. Development 2024; 151:dev203028. [PMID: 39612288 PMCID: PMC11634031 DOI: 10.1242/dev.203028] [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: 05/08/2024] [Accepted: 10/22/2024] [Indexed: 12/01/2024]
Abstract
VEGFA administration has been explored as a pro-angiogenic therapy for cardiovascular diseases including heart failure for several years, but with little success. Here, we investigate a different approach to augment VEGFA bioavailability: by deleting the VEGFA decoy receptor VEGFR1 (also known as FLT1), one can achieve more physiological VEGFA concentrations. We find that after cryoinjury, zebrafish flt1 mutant hearts display enhanced coronary revascularization and endocardial expansion, increased cardiomyocyte dedifferentiation and proliferation, and decreased scarring. Suppressing Vegfa signaling in flt1 mutants abrogates these beneficial effects of flt1 deletion. Transcriptomic analyses of cryoinjured flt1 mutant hearts reveal enhanced endothelial MAPK/ERK signaling and downregulation of the transcription factor gene egr3. Using newly generated genetic tools, we observe egr3 upregulation in the regenerating endocardium, and find that Egr3 promotes myofibroblast differentiation. These data indicate that with enhanced Vegfa bioavailability, the endocardium limits myofibroblast differentiation via egr3 downregulation, thereby providing a more permissive microenvironment for cardiomyocyte replenishment after injury.
Collapse
Affiliation(s)
- Zhen-Yu Wang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Armaan Mehra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Qian-Chen Wang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Savita Gupta
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Agatha Ribeiro da Silva
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Stefan Günther
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Mario Looso
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Jan Detleffsen
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK) Partner Site Rhine-Main, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Center, 3175 Chemin de la Côte-Sainte-Catherine, H3T 1C5 Montréal, QC, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, H3T 1J4 Montréal, QC, Canada
| |
Collapse
|
8
|
Lee J, Goeckel ME, Levitas A, Colijn S, Shin J, Hindes A, Mun G, Burton Z, Chintalapati B, Yin Y, Abello J, Solnica-Krezel L, Stratman AN. CXCR3-CXCL11 Signaling Restricts Angiogenesis and Promotes Pericyte Recruitment. Arterioscler Thromb Vasc Biol 2024; 44:2577-2595. [PMID: 39360413 PMCID: PMC11594002 DOI: 10.1161/atvbaha.124.321434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Endothelial cell (EC)-pericyte interactions are known to remodel in response to hemodynamic forces; yet there is a lack of mechanistic understanding of the signaling pathways that underlie these events. Here, we have identified a novel signaling network regulated by blood flow in ECs-the chemokine receptor CXCR3 (CXC motif chemokine receptor 3) and one of its ligands, CXCL11 (CXC motif chemokine ligand 11)-that delimits EC angiogenic potential and promotes pericyte recruitment to ECs during development. METHODS We investigated the role of CXCR3 on vascular development using both 2- and 3-dimensional in vitro assays, to study EC-pericyte interactions and EC behavioral responses to blood flow. Additionally, genetic mutants and pharmacological modulators were used in zebrafish in vivo to study the impacts of CXCR3 loss and gain of function on vascular development. RESULTS In vitro modeling of EC-pericyte interactions demonstrates that suppression of EC-specific CXCR3 signaling leads to loss of pericyte association with EC tubes. In vivo, phenotypic defects are particularly noted in the cranial vasculature, where we see a loss of pericyte association with ECs and expansion of the vasculature in zebrafish treated with the Cxcr3 inhibitor AMG487 or in homozygous cxcr3.1/3.2/3.3 triple mutants. We also demonstrate that CXCR3-deficient ECs are more elongated, move more slowly, and have impaired EC-EC junctions compared with their control counterparts. CONCLUSIONS Our results suggest that CXCR3 signaling in ECs helps promote vascular stabilization events during development by preventing EC overgrowth and promoting pericyte recruitment.
Collapse
Affiliation(s)
- Jihui Lee
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Megan E. Goeckel
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Allison Levitas
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Sarah Colijn
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Jimann Shin
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Anna Hindes
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Geonyoung Mun
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Zarek Burton
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Bharadwaj Chintalapati
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Ying Yin
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Javier Abello
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology (J.S., A.H., L.S.-K.), Washington University School of Medicine, St. Louis, MO
- Center of Regenerative Medicine (L.S.-K.), Washington University School of Medicine, St. Louis, MO
| | - Amber N. Stratman
- Department of Cell Biology and Physiology (J.L., M.E.G., A.L., S.C., G.M., Z.B., B.C., Y.Y., J.A., A.N.S.), Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
9
|
Duca S, Xia Y, Abd Elmagid L, Bakis I, Qiu M, Cao Y, Guo Y, Eichenbaum JV, McCain ML, Kang J, Harrison MRM, Cao J. Differential vegfc expression dictates lymphatic response during zebrafish heart development and regeneration. Development 2024; 151:dev202947. [PMID: 39514676 PMCID: PMC11607685 DOI: 10.1242/dev.202947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Vascular endothelial growth factor C (Vegfc) is crucial for lymphatic and blood vessel development, yet its cellular sources and specific functions in heart development remain unclear. To address this, we created a vegfc reporter and an inducible overexpression line in zebrafish. We found vegfc expression in large coronary arteries, circulating thrombocytes, cardiac adipocytes, and outflow tract smooth muscle cells. Notably, although coronary lymphangiogenesis aligns with Vegfc-expressing arteries in juveniles, it occurs only after coronary artery formation. Vegfc overexpression induced ectopic lymphatics on the ventricular surface prior to arterial formation, indicating that Vegfc abundance, rather than arterial presence, drives lymphatic development. However, this overexpression did not affect coronary artery coverage, suggesting a specific role for Vegfc in lymphatic, rather than arterial, development. Thrombocytes emerged as the initial Vegfc source during inflammation following heart injuries, transitioning to endocardial and myocardial expression during regeneration. Lower Vegfc levels in an amputation model corresponded with a lack of lymphatic expansion. Importantly, Vegfc overexpression enhanced lymphatic expansion and promoted scar resolution without affecting cardiomyocyte proliferation, highlighting its role in regulating lymphangiogenesis and promoting heart regeneration.
Collapse
Affiliation(s)
- Sierra Duca
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yu Xia
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Laila Abd Elmagid
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Isaac Bakis
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Miaoyan Qiu
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Yingxi Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Ylan Guo
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - James V. Eichenbaum
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin – Madison, Madison, WI 53705, USA
| | - Michael R. M. Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| |
Collapse
|
10
|
Shin K, Rodriguez-Parks A, Kim C, Silaban IM, Xia Y, Sun J, Dong C, Keles S, Wang J, Cao J, Kang J. Harnessing the regenerative potential of interleukin11 to enhance heart repair. Nat Commun 2024; 15:9666. [PMID: 39516197 PMCID: PMC11549343 DOI: 10.1038/s41467-024-54060-0] [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: 11/02/2023] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Balancing between regenerative processes and fibrosis is crucial for heart repair, yet strategies regulating this balance remain a barrier to developing therapies. The role of Interleukin 11 (IL11) in heart regeneration remains controversial, as both regenerative and fibrotic functions have been reported. We uncovered that il11a, an Il11 homolog in zebrafish, can trigger robust regenerative programs in zebrafish hearts, including cardiomyocytes proliferation and coronary expansion, even in the absence of injury. Notably, il11a induction in uninjured hearts also activates the quiescent epicardium to produce epicardial progenitor cells, which later differentiate into cardiac fibroblasts. Consequently, prolonged il11a induction indirectly leads to persistent fibroblast emergence, resulting in cardiac fibrosis. While deciphering the regenerative and fibrotic effects of il11a, we found that il11-dependent fibrosis, but not regeneration, is mediated through ERK activity, suggesting to potentially uncouple il11a dual effects on regeneration and fibrosis. To harness the il11a's regenerative ability, we devised a combinatorial treatment through il11a induction with ERK inhibition. This approach enhances cardiomyocyte proliferation with mitigated fibrosis, achieving a balance between regenerative processes and fibrosis. Thus, we unveil the mechanistic insights into regenerative il11 roles, offering therapeutic avenues to foster cardiac repair without exacerbating fibrosis.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Anjelica Rodriguez-Parks
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Chanul Kim
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Isabella M Silaban
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA
| | - Yu Xia
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chenyang Dong
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Sunduz Keles
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA.
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Sun B, Li Q, Xiao X, Zhang J, Zhou Y, Huang Y, Gao J, Cao X. The loach haplotype-resolved genome and the identification of Mex3a involved in fish air breathing. CELL GENOMICS 2024; 4:100670. [PMID: 39389021 PMCID: PMC11602589 DOI: 10.1016/j.xgen.2024.100670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/30/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Fish air breathing is crucial for the transition of vertebrates from water to land. So far, the genes involved in fish air breathing have not been well identified. Here, we performed gene enrichment analysis of positively selected genes (PSGs) in loach (Misgurnus anguillicaudatus, an air-breathing fish) in comparison to Triplophysa tibetana (a non-air-breathing fish), haplotype-resolved genome assembly of the loach, and gene evolutionary analysis of air-breathing and non-air-breathing fishes and found that the PSG mex3a originated from ancient air-breathing fish species. Deletion of Mex3a impaired loach air-breathing capacity by inhibiting angiogenesis through its interaction with T-box transcription factor 20. Mex3a overexpression significantly promoted angiogenesis. Structural analysis and point mutation revealed the critical role of the 201st amino acid in loach Mex3a for angiogenesis. Our findings innovatively indicate that the ancient mex3a is a fish air-breathing gene, which holds significance for understanding fish air breathing and provides a valuable resource for cultivating hypoxia-tolerant fish varieties.
Collapse
Affiliation(s)
- Bing Sun
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Qingshan Li
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinxin Xiao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianwei Zhang
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan 430070, China
| | - Ying Zhou
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuwei Huang
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Gao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xiaojuan Cao
- College of Fisheries, Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education/Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education/Key Lab of Freshwater Animal Breeding, Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
12
|
Gillespie W, Zhang Y, Ruiz OE, Cerda J, Ortiz-Guzman J, Turner WD, Largoza G, Sherman M, Mosser LE, Fujimoto E, Chien CB, Kwan KM, Arenkiel BR, Devine WP, Wythe JD. Multisite Assembly of Gateway Induced Clones (MAGIC): a flexible cloning toolbox with diverse applications in vertebrate model systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.13.603267. [PMID: 39026881 PMCID: PMC11257631 DOI: 10.1101/2024.07.13.603267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Here we present the Multisite Assembly of Gateway Induced Clones (MAGIC) system, which harnesses site-specific recombination-based cloning via Gateway technology for rapid, modular assembly of between 1 and 3 "Entry" vector components, all into a fourth, standard high copy "Destination" plasmid backbone. The MAGIC toolkit spans a range of in vitro and in vivo uses, from directing tunable gene expression, to driving simultaneous expression of microRNAs and fluorescent reporters, to enabling site-specific recombinase-dependent gene expression. All MAGIC system components are directly compatible with existing multisite gateway Tol2 systems currently used in zebrafish, as well as existing eukaryotic cell culture expression Destination plasmids, and available mammalian lentiviral and adenoviral Destination vectors, allowing rapid cross-species experimentation. Moreover, herein we describe novel vectors with flanking piggyBac transposon elements for stable genomic integration in vitro or in vivo when used with piggyBac transposase. Collectively, the MAGIC system facilitates transgenesis in cultured mammalian cells, electroporated mouse and chick embryos, as well as in injected zebrafish embryos, enabling the rapid generation of innovative DNA constructs for biological research due to a shared, common plasmid platform.
Collapse
|
13
|
Wong D, Martinez J, Quijada P. Exploring the Function of Epicardial Cells Beyond the Surface. Circ Res 2024; 135:353-371. [PMID: 38963865 PMCID: PMC11225799 DOI: 10.1161/circresaha.124.321567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
The epicardium, previously viewed as a passive outer layer around the heart, is now recognized as an essential component in development, regeneration, and repair. In this review, we explore the cellular and molecular makeup of the epicardium, highlighting its roles in heart regeneration and repair in zebrafish and salamanders, as well as its activation in young and adult postnatal mammals. We also examine the latest technologies used to study the function of epicardial cells for therapeutic interventions. Analysis of highly regenerative animal models shows that the epicardium is essential in regulating cardiomyocyte proliferation, transient fibrosis, and neovascularization. However, despite the epicardium's unique cellular programs to resolve cardiac damage, it remains unclear how to replicate these processes in nonregenerative mammalian organisms. During myocardial infarction, epicardial cells secrete signaling factors that modulate fibrotic, vascular, and inflammatory remodeling, which differentially enhance or inhibit cardiac repair. Recent transcriptomic studies have validated the cellular and molecular heterogeneity of the epicardium across various species and developmental stages, shedding further light on its function under pathological conditions. These studies have also provided insights into the function of regulatory epicardial-derived signaling molecules in various diseases, which could lead to new therapies and advances in reparative cardiovascular medicine. Moreover, insights gained from investigating epicardial cell function have initiated the development of novel techniques, including using human pluripotent stem cells and cardiac organoids to model reparative processes within the cardiovascular system. This growing understanding of epicardial function holds the potential for developing innovative therapeutic strategies aimed at addressing developmental heart disorders, enhancing regenerative therapies, and mitigating cardiovascular disease progression.
Collapse
Affiliation(s)
- David Wong
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, CA 90029
| | - Julie Martinez
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Molecular, Cellular and Integrative Physiology Graduate Program, University of California, Los Angeles, CA 90029
| | - Pearl Quijada
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90029
- Eli and Edythe Broad Stem Research Center, University of California, Los Angeles, CA 90029
- Molecular Biology Institute, University of California, Los Angeles, CA 90029
| |
Collapse
|
14
|
Rios Coronado PE, Zanetti D, Zhou J, Naftaly JA, Prabala P, Martínez Jaimes AM, Farah EN, Fan X, Kundu S, Deshpande SS, Evergreen I, Kho PF, Hilliard AT, Abramowitz S, Pyarajan S, Dochtermann D, Damrauer SM, Chang KM, Levin MG, Winn VD, Paşca AM, Plomondon ME, Waldo SW, Tsao PS, Kundaje A, Chi NC, Clarke SL, Red-Horse K, Assimes TL. CXCL12 drives natural variation in coronary artery anatomy across diverse populations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.27.23297507. [PMID: 37961706 PMCID: PMC10635223 DOI: 10.1101/2023.10.27.23297507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
To efficiently distribute blood flow to cardiac muscle, the coronary artery tree must follow a specific branching pattern over the heart. How this pattern arises in humans is unknown due to the limitations of studying human heart development. Here, we leveraged a natural variation of coronary artery anatomy, known as coronary dominance, in genetic association studies to identify the first known driver of human coronary developmental patterning. Coronary dominance refers to whether the right, left, or both coronary arteries branch over the posterior left ventricle, but whether this variability is heritable and how it would be genetically regulated was completely unknown. By conducting the first large-scale, multi-ancestry genome-wide association study (GWAS) of coronary dominance in 61,043 participants of the VA Million Veteran Program, we observed moderate heritability (27.7%) with ten loci reaching genome wide significance. An exceptionally strong association mapped DNA variants to a non-coding region near the chemokine CXCL12 in both European and African ancestries, which overlapped with variants associated with coronary artery disease. Genomic analyses predicted these variants to impact CXCL12 levels, and imaging revealed dominance to develop during fetal life coincident with CXCL12 expression. Reducing Cxcl12 in mice to model the human genetics altered septal artery dominance patterns and caused coronary branches to develop away from Cxcl12 expression domains. Cxcl12 heterozygosity did not compromise overall artery coverage as seen with full deletion, but instead changed artery patterning, reminiscent of the human scenario. Together, our data support CXCL12 as a critical determinant of human coronary artery growth and patterning and lay a foundation for the utilization of developmental pathways to guide future precision 'medical revascularization' therapeutics.
Collapse
Affiliation(s)
| | - Daniela Zanetti
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Stanford, CA, USA
- VA Palo Alto Health Care System; Palo Alto, CA, USA
- Institute of Genetic and Biomedical Research, National Research Council; Cagliari, Sardinia, Italy
| | - Jiayan Zhou
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Stanford, CA, USA
- VA Palo Alto Health Care System; Palo Alto, CA, USA
| | | | - Pratima Prabala
- Department of Biology, Stanford University; Stanford, CA, USA
| | - Azalia M. Martínez Jaimes
- Department of Biology, Stanford University; Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine; Stanford, CA, USA
| | - Elie N. Farah
- Department of Medicine, Division of Cardiology, University of California San Diego; La Jolla, CA, USA
| | - Xiaochen Fan
- Department of Biology, Stanford University; Stanford, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
- Department of Computer Science, Stanford University; Stanford, CA, USA
| | - Salil S. Deshpande
- Institute for Computational and Mathematical Engineering, Stanford University School of Medicine; Stanford, CA, USA
| | - Ivy Evergreen
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
| | - Pik Fang Kho
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Stanford, CA, USA
- VA Palo Alto Health Care System; Palo Alto, CA, USA
| | | | - Sarah Abramowitz
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Sarnoff Cardiovascular Research Foundation; McLean, VA, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell; Hempstead, NY, USA
| | - Saiju Pyarajan
- Center for Data and Computational Sciences, VA Boston Healthcare System; Boston, MA, USA
| | - Daniel Dochtermann
- Center for Data and Computational Sciences, VA Boston Healthcare System; Boston, MA, USA
| | | | - Scott M. Damrauer
- Corporal Michael J. Crescenz VA Medical Center; Philadelphia, PA, USA
- Department of Surgery, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center; Philadelphia, PA, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
| | - Michael G. Levin
- Department of Medicine, Division of Cardiovascular Medicine, University of Pennsylvania Perelman School of Medicine; Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center; Philadelphia, PA, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine; Stanford, CA, USA
| | - Anca M. Paşca
- Department of Pediatrics, Neonatology, Stanford University School of Medicine; Stanford, CA, USA
| | - Mary E. Plomondon
- Department of Medicine, Rocky Mountain Regional VA Medical Center; Aurora, CO, USA
- CART Program, VHA Office of Quality and Patient Safety; Washington, DC, USA
| | - Stephen W. Waldo
- Department of Medicine, Rocky Mountain Regional VA Medical Center; Aurora, CO, USA
- CART Program, VHA Office of Quality and Patient Safety; Washington, DC, USA
- Division of Cardiology, University of Colorado School of Medicine; Aurora, CO, USA
| | - Philip S. Tsao
- VA Palo Alto Health Care System; Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine; Stanford, CA, USA
- Cardiovascular Institute, Stanford University School of Medicine; Stanford, CA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine; Stanford, CA, USA
- Department of Computer Science, Stanford University; Stanford, CA, USA
| | - Neil C. Chi
- Department of Medicine, Division of Cardiology, University of California San Diego; La Jolla, CA, USA
| | - Shoa L. Clarke
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Stanford, CA, USA
- VA Palo Alto Health Care System; Palo Alto, CA, USA
- Department of Medicine, Stanford Prevention Research Center, Stanford University School of Medicine; Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University; Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine; Stanford, CA, USA
- Howard Hughes Medical Institute; Chevy Chase, MD, USA
| | - Themistocles L. Assimes
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine; Stanford, CA, USA
- VA Palo Alto Health Care System; Palo Alto, CA, USA
- Cardiovascular Institute, Stanford University School of Medicine; Stanford, CA, USA
- Department of Epidemiology and Population Health, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
15
|
Abu Nahia K, Sulej A, Migdał M, Ochocka N, Ho R, Kamińska B, Zagorski M, Winata CL. scRNA-seq reveals the diversity of the developing cardiac cell lineage and molecular players in heart rhythm regulation. iScience 2024; 27:110083. [PMID: 38872974 PMCID: PMC11170199 DOI: 10.1016/j.isci.2024.110083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/26/2024] [Accepted: 05/20/2024] [Indexed: 06/15/2024] Open
Abstract
We utilized scRNA-seq to delineate the diversity of cell types in the zebrafish heart. Transcriptome profiling of over 50,000 cells at 48 and 72 hpf defined at least 18 discrete cell lineages of the developing heart. Utilizing well-established gene signatures, we identified a population of cells likely to be the primary pacemaker and characterized the transcriptome profile defining this critical cell type. Two previously uncharacterized genes, atp1b3b and colec10, were found to be enriched in the sinoatrial cardiomyocytes. CRISPR/Cas9-mediated knockout of these two genes significantly reduced heart rate, implicating their role in cardiac development and conduction. Additionally, we describe other cardiac cell lineages, including the endothelial and neural cells, providing their expression profiles as a resource. Our results established a detailed atlas of the developing heart, providing valuable insights into cellular and molecular mechanisms, and pinpointed potential new players in heart rhythm regulation.
Collapse
Affiliation(s)
- Karim Abu Nahia
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agata Sulej
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Migdał
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Natalia Ochocka
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Richard Ho
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
- The Njord Centre, Department of Physics, University of Oslo, Oslo, Norway
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Marcin Zagorski
- Institute of Theoretical Physics and Mark Kac Center for Complex Systems Research, Jagiellonian University, Cracow, Poland
| | | |
Collapse
|
16
|
Shin K, Rodriguez-Parks A, Kim C, Silaban IM, Xia Y, Sun J, Dong C, Keles S, Wang J, Cao J, Kang J. Harnessing the regenerative potential of interleukin11 to enhance heart repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.577788. [PMID: 38352555 PMCID: PMC10862709 DOI: 10.1101/2024.01.29.577788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Balancing between regenerative processes and fibrosis is crucial for heart repair, yet strategies regulating this balance remain a barrier to developing therapies. While Interleukin11 (IL11) is known as a fibrotic factor, its contribution to heart regeneration is poorly understood. We uncovered that il11a, an Il11 homolog in zebrafish, can trigger robust regenerative programs in zebrafish hearts, including cardiomyocytes proliferation and coronary expansion, even in the absence of injury. However, prolonged il11a induction in uninjured hearts causes persistent fibroblast emergence, resulting in fibrosis. While deciphering the regenerative and fibrotic effects of il11a, we found that il11-dependent fibrosis, but not regeneration, is mediated through ERK activity, suggesting to potentially uncouple il11a dual effects on regeneration and fibrosis. To harness the il11a's regenerative ability, we devised a combinatorial treatment through il11a induction with ERK inhibition. This approach enhances cardiomyocyte proliferation with mitigated fibrosis, achieving a balance between regenerative processes and fibrosis. Thus, we unveil the mechanistic insights into regenerative il11 roles, offering therapeutic avenues to foster cardiac repair without exacerbating fibrosis.
Collapse
Affiliation(s)
- Kwangdeok Shin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Anjelica Rodriguez-Parks
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Chanul Kim
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Isabella M Silaban
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Yu Xia
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Chenyang Dong
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Sunduz Keles
- Departments of Statistics and of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI 53706, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Junsu Kang
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin - Madison, Madison, WI, 53705, USA
| |
Collapse
|
17
|
Yang D, Jian Z, Tang C, Chen Z, Zhou Z, Zheng L, Peng X. Zebrafish Congenital Heart Disease Models: Opportunities and Challenges. Int J Mol Sci 2024; 25:5943. [PMID: 38892128 PMCID: PMC11172925 DOI: 10.3390/ijms25115943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Congenital heart defects (CHDs) are common human birth defects. Genetic mutations potentially cause the exhibition of various pathological phenotypes associated with CHDs, occurring alone or as part of certain syndromes. Zebrafish, a model organism with a strong molecular conservation similar to humans, is commonly used in studies on cardiovascular diseases owing to its advantageous features, such as a similarity to human electrophysiology, transparent embryos and larvae for observation, and suitability for forward and reverse genetics technology, to create various economical and easily controlled zebrafish CHD models. In this review, we outline the pros and cons of zebrafish CHD models created by genetic mutations associated with single defects and syndromes and the underlying pathogenic mechanism of CHDs discovered in these models. The challenges of zebrafish CHD models generated through gene editing are also discussed, since the cardiac phenotypes resulting from a single-candidate pathological gene mutation in zebrafish might not mirror the corresponding human phenotypes. The comprehensive review of these zebrafish CHD models will facilitate the understanding of the pathogenic mechanisms of CHDs and offer new opportunities for their treatments and intervention strategies.
Collapse
|
18
|
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Ruiz-Villalba A, Guadix JA, Pérez-Pomares JM. Epicardium and Coronary Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:155-166. [PMID: 38884710 DOI: 10.1007/978-3-031-44087-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Congenital anomalies and acquired diseases of the coronary blood vessels are of great clinical relevance. The early diagnosis of these conditions remains, however, challenging. In order to improve our knowledge of these ailments, progress has to be achieved in the research of the molecular and cellular mechanisms that control development of the coronary vascular bed. The aim of this chapter is to provide a succint account of the key elements of coronary blood vessel development, especially in the context of the role played by the epicardium and epicardial cellular derivatives. We will discuss the importance of the epicardium in coronary blood vessel morphogenesis, from the contribution of the epicardially derived mesenchyme to these blood vessels to its role as an instructive signaling center, attempting to relate these concepts to the origin of coronary disease.
Collapse
Affiliation(s)
- Adrián Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain
| | - Juan Antonio Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain
| | - José M Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain.
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain.
| |
Collapse
|
20
|
Bakis I, Sun Y, Abd Elmagid L, Feng X, Garibyan M, Yip JK, Yu FZ, Chowdhary S, Fernandez GE, Cao J, McCain ML, Lien CL, Harrison MR. Methods for dynamic and whole volume imaging of the zebrafish heart. Dev Biol 2023; 504:75-85. [PMID: 37708968 PMCID: PMC10841891 DOI: 10.1016/j.ydbio.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/02/2023] [Accepted: 09/06/2023] [Indexed: 09/16/2023]
Abstract
Tissue development and regeneration are dynamic processes involving complex cell migration and cell-cell interactions. We have developed a protocol for complementary time-lapse and three-dimensional (3D) imaging of tissue for developmental and regeneration studies which we apply here to the zebrafish cardiac vasculature. 3D imaging of fixed specimens is used to first define the subject at high resolution then live imaging captures how it changes dynamically. Hearts from adult and juvenile zebrafish are extracted and cleaned in preparation for the different imaging modalities. For whole-mount 3D confocal imaging, single or multiple hearts with native fluorescence or immuno-labeling are prepared for stabilization or clearing, and then imaged. For live imaging, hearts are placed in a prefabricated fluidic device and set on a temperature-controlled microscope for culture and imaging over several days. This protocol allows complete visualization of morphogenic processes in a 3D context and provides the ability to follow cell behaviors to complement in vivo and fixed tissue studies. This culture and imaging protocol can be applied to different cell and tissue types. Here, we have used it to observe zebrafish coronary vasculature and the migration of coronary endothelial cells during heart regeneration.
Collapse
Affiliation(s)
- Isaac Bakis
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10021, USA
| | - Yuhan Sun
- Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Laila Abd Elmagid
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10021, USA
| | - Xidi Feng
- Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Mher Garibyan
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Joycelyn K Yip
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA
| | - Fang Zhou Yu
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Emergency Medicine, Nuvance Health, Poughkeepsie, NY, 12601, USA
| | - Sayali Chowdhary
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10021, USA
| | - Gerardo Esteban Fernandez
- Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA
| | - Jingli Cao
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10021, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, 90089, USA; Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Ching-Ling Lien
- Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA, 90027, USA; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Michael Rm Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA; Department of Cell and Developmental Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10021, USA.
| |
Collapse
|
21
|
Zlatanova I, Sun F, Wu RS, Chen X, Lau BH, Colombier P, Sinha T, Celona B, Xu SM, Materna SC, Huang GN, Black BL. An injury-responsive mmp14b enhancer is required for heart regeneration. SCIENCE ADVANCES 2023; 9:eadh5313. [PMID: 38019918 PMCID: PMC10686572 DOI: 10.1126/sciadv.adh5313] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Mammals have limited capacity for heart regeneration, whereas zebrafish have extraordinary regeneration abilities. During zebrafish heart regeneration, endothelial cells promote cardiomyocyte cell cycle reentry and myocardial repair, but the mechanisms responsible for promoting an injury microenvironment conducive to regeneration remain incompletely defined. Here, we identify the matrix metalloproteinase Mmp14b as an essential regulator of heart regeneration. We identify a TEAD-dependent mmp14b endothelial enhancer induced by heart injury in zebrafish and mice, and we show that the enhancer is required for regeneration, supporting a role for Hippo signaling upstream of mmp14b. Last, we show that MMP-14 function in mice is important for the accumulation of Agrin, an essential regulator of neonatal mouse heart regeneration. These findings reveal mechanisms for extracellular matrix remodeling that promote heart regeneration.
Collapse
Affiliation(s)
- Ivana Zlatanova
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Fei Sun
- Duke Regeneration Center, Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Roland S. Wu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xiaoxin Chen
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bryan H. Lau
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Pauline Colombier
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tanvi Sinha
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barbara Celona
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Shan-Mei Xu
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stefan C. Materna
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Guo N. Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brian L. Black
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
22
|
Meier AB, Zawada D, De Angelis MT, Martens LD, Santamaria G, Zengerle S, Nowak-Imialek M, Kornherr J, Zhang F, Tian Q, Wolf CM, Kupatt C, Sahara M, Lipp P, Theis FJ, Gagneur J, Goedel A, Laugwitz KL, Dorn T, Moretti A. Epicardioid single-cell genomics uncovers principles of human epicardium biology in heart development and disease. Nat Biotechnol 2023; 41:1787-1800. [PMID: 37012447 PMCID: PMC10713454 DOI: 10.1038/s41587-023-01718-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 02/22/2023] [Indexed: 04/05/2023]
Abstract
The epicardium, the mesothelial envelope of the vertebrate heart, is the source of multiple cardiac cell lineages during embryonic development and provides signals that are essential to myocardial growth and repair. Here we generate self-organizing human pluripotent stem cell-derived epicardioids that display retinoic acid-dependent morphological, molecular and functional patterning of the epicardium and myocardium typical of the left ventricular wall. By combining lineage tracing, single-cell transcriptomics and chromatin accessibility profiling, we describe the specification and differentiation process of different cell lineages in epicardioids and draw comparisons to human fetal development at the transcriptional and morphological levels. We then use epicardioids to investigate the functional cross-talk between cardiac cell types, gaining new insights into the role of IGF2/IGF1R and NRP2 signaling in human cardiogenesis. Finally, we show that epicardioids mimic the multicellular pathogenesis of congenital or stress-induced hypertrophy and fibrotic remodeling. As such, epicardioids offer a unique testing ground of epicardial activity in heart development, disease and regeneration.
Collapse
Affiliation(s)
- Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Dorota Zawada
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Maria Teresa De Angelis
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Experimental and Clinical Medicine, University 'Magna Graecia', Catanzaro, Italy
| | - Laura D Martens
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
- Helmholtz Association-Munich School for Data Science (MUDS), Munich, Germany
| | - Gianluca Santamaria
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Experimental and Clinical Medicine, University 'Magna Graecia', Catanzaro, Italy
| | - Sophie Zengerle
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Monika Nowak-Imialek
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Jessica Kornherr
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Fangfang Zhang
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Qinghai Tian
- Center for Molecular Signaling (PZMS), Institute for Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, Homburg, Germany
| | - Cordula M Wolf
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
- Department of Congenital Heart Defects and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Christian Kupatt
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Peter Lipp
- Center for Molecular Signaling (PZMS), Institute for Molecular Cell Biology, Research Center for Molecular Imaging and Screening, Medical Faculty, Saarland University, Homburg, Germany
| | - Fabian J Theis
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Julien Gagneur
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
- Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Goedel
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Karl-Ludwig Laugwitz
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Tatjana Dorn
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- Regenerative Medicine in Cardiovascular Diseases, First Department of Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany.
- German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany.
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
23
|
Jiang H, Zhao Z, Yu H, Lin Q, Liu Y. Evolutionary traits and functional roles of chemokines and their receptors in the male pregnancy of the Syngnathidae. MARINE LIFE SCIENCE & TECHNOLOGY 2023; 5:500-510. [PMID: 38045539 PMCID: PMC10689615 DOI: 10.1007/s42995-023-00205-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/27/2023] [Indexed: 12/05/2023]
Abstract
Vertebrates have developed various modes of reproduction, some of which are found in Teleosts. Over 300 species of the Syngnathidae (seahorses, pipefishes and seadragons) exhibit male pregnancies; the males have specialized brood pouches that provide immune protection, nourishment, and oxygen regulation. Chemokines play a vital role at the mammalian maternal-fetal interface; however, their functions in fish reproduction are unclear. This study revealed the evolutionary traits and potential functions of chemokine genes in 22 oviparous, ovoviviparous, and viviparous fish species through comparative genomic analyses. Our results showed that chemokine gene copy numbers and evolutionary rates vary among species with different modes of reproduction. Syngnathidae lost cxcl13 and cxcr5, which are involved in key receptor-ligand pairs for lymphoid organ development. Notably, Syngnathidae have site-specific mutations in cxcl12b and ccl44, suggesting immune function during gestation. Moreover, transcriptome analysis revealed that chemokine gene expression varies among Syngnathidae species with different types of brood pouches, suggesting adaptive variations in chemokine functions among seahorses and their relatives. Furthermore, challenge experiments on seahorse brood pouches revealed a joint immune function of chemokine genes during male pregnancy. This study provides insights into the evolutionary diversity of chemokine genes associated with different reproductive modes in fish. Supplementary Information The online version contains supplementary material available at 10.1007/s42995-023-00205-x.
Collapse
Affiliation(s)
- Han Jiang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- University of Chinese Academy of Sciences, Beijing, 101400 China
| | - Zhanwei Zhao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- University of Chinese Academy of Sciences, Beijing, 101400 China
| | - Haiyan Yu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
| | - Qiang Lin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- University of Chinese Academy of Sciences, Beijing, 101400 China
| | - Yali Liu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, 510301 China
- University of Chinese Academy of Sciences, Beijing, 101400 China
| |
Collapse
|
24
|
Goeckel ME, Lee J, Levitas A, Colijn S, Mun G, Burton Z, Chintalapati B, Yin Y, Abello J, Stratman A. CXCR3-CXCL11 signaling restricts angiogenesis and promotes pericyte recruitment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.16.557842. [PMID: 37745440 PMCID: PMC10516035 DOI: 10.1101/2023.09.16.557842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Endothelial cell (EC)-pericyte interactions are known to remodel in response to hemodynamic forces, yet there is a lack of mechanistic understanding of the signaling pathways that underlie these events. Here, we have identified a novel signaling network regulated by blood flow in ECs-the chemokine receptor, CXCR3, and one of its ligands, CXCL11-that delimits EC angiogenic potential and suppresses pericyte recruitment during development through regulation of pdgfb expression in ECs. In vitro modeling of EC-pericyte interactions demonstrates that suppression of EC-specific CXCR3 signaling leads to loss of pericyte association with EC tubes. In vivo, phenotypic defects are particularly noted in the cranial vasculature, where we see a loss of pericyte association with and expansion of the vasculature in zebrafish treated with the Cxcr3 inhibitor AMG487. We also demonstrate using flow modeling platforms that CXCR3-deficient ECs are more elongated, move more slowly, and have impaired EC-EC junctions compared to their control counterparts. Together these data suggest that CXCR3 signaling in ECs drives vascular stabilization events during development.
Collapse
Affiliation(s)
- Megan E. Goeckel
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
- University of Nebraska Medical Center, Graduate Studies, Nebraska Medical Center, Omaha, NE 68198
| | - Jihui Lee
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Allison Levitas
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Sarah Colijn
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Geonyoung Mun
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Zarek Burton
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Bharadwaj Chintalapati
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Ying Yin
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Javier Abello
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| | - Amber Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine St. Louis, MO, 63110
| |
Collapse
|
25
|
Mizukami K, Higashiyama H, Arima Y, Ando K, Okada N, Kose K, Yamada S, Takeuchi JK, Koshiba-Takeuchi K, Fukuhara S, Miyagawa-Tomita S, Kurihara H. Coronary artery established through amniote evolution. eLife 2023; 12:e83005. [PMID: 37605519 PMCID: PMC10444023 DOI: 10.7554/elife.83005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 07/17/2023] [Indexed: 08/23/2023] Open
Abstract
Coronary arteries are a critical part of the vascular system and provide nourishment to the heart. In humans, even minor defects in coronary arteries can be lethal, emphasizing their importance for survival. However, some teleosts survive without coronary arteries, suggesting that there may have been some evolutionary changes in the morphology and function of coronary arteries in the tetrapod lineage. Here, we propose that the true ventricular coronary arteries were newly established during amniote evolution through remodeling of the ancestral coronary vasculature. In mouse (Mus musculus) and Japanese quail (Coturnix japonica) embryos, the coronary arteries unique to amniotes are established by the reconstitution of transient vascular plexuses: aortic subepicardial vessels (ASVs) in the outflow tract and the primitive coronary plexus on the ventricle. In contrast, amphibians (Hyla japonica, Lithobates catesbeianus, Xenopus laevis, and Cynops pyrrhogaster) retain the ASV-like vasculature as truncal coronary arteries throughout their lives and have no primitive coronary plexus. The anatomy and development of zebrafish (Danio rerio) and chondrichthyans suggest that their hypobranchial arteries are ASV-like structures serving as the root of the coronary vasculature throughout their lives. Thus, the ventricular coronary artery of adult amniotes is a novel structure that has acquired a new remodeling process, while the ASVs, which occur transiently during embryonic development, are remnants of the ancestral coronary vessels. This evolutionary change may be related to the modification of branchial arteries, indicating considerable morphological changes underlying the physiological transition during amniote evolution.
Collapse
Affiliation(s)
- Kaoru Mizukami
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Hiroki Higashiyama
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| | - Yuichiro Arima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
- Developmental Cardiology Laboratory, International Research Center for Medical Science, Kumamoto UniversityKumamotoJapan
| | - Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical SchoolTokyoJapan
| | | | - Katsumi Kose
- Institute of Applied Physics, University of TsukubaTsukubaJapan
| | - Shigehito Yamada
- Congenital Anomaly Research Center, Kyoto University Graduate School of MedicineKyotoJapan
| | - Jun K Takeuchi
- Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental UniversityTokyoJapan
| | | | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical SchoolTokyoJapan
| | - Sachiko Miyagawa-Tomita
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
- Heart Center, Department of Pediatric Cardiology, Tokyo Women’s Medical UniversityTokyoJapan
- Department of Animal Nursing Science, Yamazaki University of Animal Health TechnologyTokyoJapan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of TokyoTokyoJapan
| |
Collapse
|
26
|
Zhuo D, Lei I, Li W, Liu L, Li L, Ni J, Liu Z, Fan G. The origin, progress, and application of cell-based cardiac regeneration therapy. J Cell Physiol 2023; 238:1732-1755. [PMID: 37334836 DOI: 10.1002/jcp.31060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/21/2023]
Abstract
Cardiovascular disease (CVD) has become a severe threat to human health, with morbidity and mortality increasing yearly and gradually becoming younger. When the disease progresses to the middle and late stages, the loss of a large number of cardiomyocytes is irreparable to the body itself, and clinical drug therapy and mechanical support therapy cannot reverse the development of the disease. To explore the source of regenerated myocardium in model animals with the ability of heart regeneration through lineage tracing and other methods, and develop a new alternative therapy for CVDs, namely cell therapy. It directly compensates for cardiomyocyte proliferation through adult stem cell differentiation or cell reprogramming, which indirectly promotes cardiomyocyte proliferation through non-cardiomyocyte paracrine, to play a role in heart repair and regeneration. This review comprehensively summarizes the origin of newly generated cardiomyocytes, the research progress of cardiac regeneration based on cell therapy, the opportunity and development of cardiac regeneration in the context of bioengineering, and the clinical application of cell therapy in ischemic diseases.
Collapse
Affiliation(s)
- Danping Zhuo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjun Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Liu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihao Liu
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
27
|
Sun J, Peterson EA, Chen X, Wang J. hapln1a + cells guide coronary growth during heart morphogenesis and regeneration. Nat Commun 2023; 14:3505. [PMID: 37311876 PMCID: PMC10264374 DOI: 10.1038/s41467-023-39323-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/07/2023] [Indexed: 06/15/2023] Open
Abstract
Although several tissues and chemokines orchestrate coronary formation, the guidance cues for coronary growth remain unclear. Here, we profile the juvenile zebrafish epicardium during coronary vascularization and identify hapln1a+ cells enriched with vascular-regulating genes. hapln1a+ cells not only envelop vessels but also form linear structures ahead of coronary sprouts. Live-imaging demonstrates that coronary growth occurs along these pre-formed structures, with depletion of hapln1a+ cells blocking this growth. hapln1a+ cells also pre-lead coronary sprouts during regeneration and hapln1a+ cell loss inhibits revascularization. Further, we identify serpine1 expression in hapln1a+ cells adjacent to coronary sprouts, and serpine1 inhibition blocks vascularization and revascularization. Moreover, we observe the hapln1a substrate, hyaluronan, forming linear structures along and preceding coronary vessels. Depletion of hapln1a+ cells or serpine1 activity inhibition disrupts hyaluronan structure. Our studies reveal that hapln1a+ cells and serpine1 are required for coronary production by establishing a microenvironment to facilitate guided coronary growth.
Collapse
Affiliation(s)
- Jisheng Sun
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Cardiology Division, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
28
|
Liu J, Zhang M, Dong H, Liu J, Mao A, Ning G, Cao Y, Zhang Y, Wang Q. Chemokine signaling synchronizes angioblast proliferation and differentiation during pharyngeal arch artery vasculogenesis. Development 2022; 149:285824. [PMID: 36468454 PMCID: PMC10114070 DOI: 10.1242/dev.200754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 11/14/2022] [Indexed: 12/09/2022]
Abstract
Developmentally, the great vessels of the heart originate from the pharyngeal arch arteries (PAAs). During PAA vasculogenesis, PAA precursors undergo sequential cell fate decisions that are accompanied by proliferative expansion. However, how these two processes are synchronized remains poorly understood. Here, we find that the zebrafish chemokine receptor Cxcr4a is expressed in PAA precursors, and genetic ablation of either cxcr4a or the ligand gene cxcl12b causes PAA stenosis. Cxcr4a is required for the activation of the downstream PI3K/AKT cascade, which promotes not only PAA angioblast proliferation, but also differentiation. AKT has a well-known role in accelerating cell-cycle progression through the activation of cyclin-dependent kinases. Despite this, we demonstrate that AKT phosphorylates Etv2 and Scl, the key regulators of angioblast commitment, on conserved serine residues, thereby protecting them from ubiquitin-mediated proteasomal degradation. Altogether, our study reveals a central role for chemokine signaling in PAA vasculogenesis through orchestrating angioblast proliferation and differentiation.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Mingming Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Haojian Dong
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Jingwen Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Aihua Mao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Guozhu Ning
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Cao
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Yiyue Zhang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Qiang Wang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
29
|
D'Amato G, Phansalkar R, Naftaly JA, Fan X, Amir ZA, Rios Coronado PE, Cowley DO, Quinn KE, Sharma B, Caron KM, Vigilante A, Red-Horse K. Endocardium-to-coronary artery differentiation during heart development and regeneration involves sequential roles of Bmp2 and Cxcl12/Cxcr4. Dev Cell 2022; 57:2517-2532.e6. [PMID: 36347256 PMCID: PMC9833645 DOI: 10.1016/j.devcel.2022.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/28/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Endocardial cells lining the heart lumen are coronary vessel progenitors during embryogenesis. Re-igniting this developmental process in adults could regenerate blood vessels lost during cardiac injury, but this requires additional knowledge of molecular mechanisms. Here, we use mouse genetics and scRNA-seq to identify regulators of endocardial angiogenesis and precisely assess the role of CXCL12/CXCR4 signaling. Time-specific lineage tracing demonstrated that endocardial cells differentiated into coronary endothelial cells primarily at mid-gestation. A new mouse line reporting CXCR4 activity-along with cell-specific gene deletions-demonstrated it was specifically required for artery morphogenesis rather than angiogenesis. Integrating scRNA-seq data of endocardial-derived coronary vessels from mid- and late-gestation identified a Bmp2-expressing transitioning population specific to mid-gestation. Bmp2 stimulated endocardial angiogenesis in vitro and in injured neonatal mouse hearts. Our data demonstrate how understanding the molecular mechanisms underlying endocardial angiogenesis can identify new potential therapeutic targets promoting revascularization of the injured heart.
Collapse
Affiliation(s)
- Gaetano D'Amato
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ragini Phansalkar
- Department of Biology, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Xiaochen Fan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Zhainib A Amir
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Dale O Cowley
- Animal Models Core, University of North Carolina, Chapel Hill, NC, USA
| | - Kelsey E Quinn
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alessandra Vigilante
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London, UK
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford, CA, USA.
| |
Collapse
|
30
|
Diago-Domingo L, Cuervo H, Benedito R. BMPing up endocardial angiogenesis to generate coronary vessels. Dev Cell 2022; 57:2515-2516. [DOI: 10.1016/j.devcel.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Abstract
The efficacy of implanted biomaterials is largely dependent on the response of the host's immune and stromal cells. Severe foreign body response (FBR) can impede the integration of the implant into the host tissue and compromise the intended mechanical and biochemical function. Many features of FBR, including late-stage fibrotic encapsulation of implants, parallel the formation of fibrotic scar tissue after tissue injury. Regenerative organisms like zebrafish and salamanders can avoid fibrosis after injury entirely, but FBR in these research organisms is rarely investigated because their immune competence is much lower than humans. The recent characterization of a regenerative mammal, the spiny mouse (Acomys), has inspired us to take a closer look at cellular regulation in regenerative organisms across the animal kingdom for insights into avoiding FBR in humans. Here, we highlight how major features of regeneration, such as blastema formation, macrophage polarization, and matrix composition, can be modulated across a range of regenerative research organisms to elucidate common features that may be harnessed to minimize FBR. Leveraging a deeper understanding of regenerative biology for biomaterial design may help to reduce FBR and improve device integration and performance.
Collapse
Affiliation(s)
- Sunaina Sapru
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Michele N Dill
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Chelsey S Simmons
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, United States.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
32
|
Abd Elmagid L, Mittal N, Bakis I, Lien CL, Harrison MR. Intramyocardial Injection for the Study of Cardiac Lymphatic Function in Zebrafish. J Vis Exp 2022:10.3791/64504. [PMID: 36282694 PMCID: PMC11376687 DOI: 10.3791/64504] [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] [Indexed: 09/01/2023] Open
Abstract
Zebrafish have proved to be an important model for studying cardiovascular formation and function during postembryonic development and regeneration. The present protocol describes a method for injecting fluorescent tracers into the zebrafish myocardium to study interstitial fluid and debris uptake into cardiac lymphatic vessels. To do so, microspheres (200 nm diameter) and quantum dots (<10 nm diameter) are introduced into the myocardium of live zebrafish, which can be tracked using ex vivo confocal microscopy. These tracers are then tracked intermittently over several hours to follow clearance from the myocardium into cardiac lymphatic vessels. Quantum dots are transported through cardiac lymphatic vessels away from the heart, while larger microspheres remain at the injection site for over three weeks. This method of intramyocardial injection can be extended to other uses, including the injection of encapsulated MS or hydrogels to locally release cells, proteins, or compounds of interest to a targeted region of the heart.
Collapse
Affiliation(s)
- Laila Abd Elmagid
- Cardiovascular Research Institute, Weill Cornell Medicine; Department of Cell and Developmental Biology, Weill Cornell Medicine
| | - Nishant Mittal
- Cardiovascular Research Institute, Weill Cornell Medicine; Department of Cell and Developmental Biology, Weill Cornell Medicine
| | - Isaac Bakis
- Cardiovascular Research Institute, Weill Cornell Medicine; Department of Cell and Developmental Biology, Weill Cornell Medicine
| | - Ching-Ling Lien
- The Saban Research Institute and Heart Institute, Children's Hospital Los Angeles
| | - Michael R Harrison
- Cardiovascular Research Institute, Weill Cornell Medicine; Department of Cell and Developmental Biology, Weill Cornell Medicine;
| |
Collapse
|
33
|
Sun J, Peterson EA, Wang AZ, Ou J, Smith KE, Poss KD, Wang J. hapln1 Defines an Epicardial Cell Subpopulation Required for Cardiomyocyte Expansion During Heart Morphogenesis and Regeneration. Circulation 2022; 146:48-63. [PMID: 35652354 PMCID: PMC9308751 DOI: 10.1161/circulationaha.121.055468] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Certain nonmammalian species such as zebrafish have an elevated capacity for innate heart regeneration. Understanding how heart regeneration occurs in these contexts can help illuminate cellular and molecular events that can be targets for heart failure prevention or treatment. The epicardium, a mesothelial tissue layer that encompasses the heart, is a dynamic structure that is essential for cardiac regeneration in zebrafish. The extent to which different cell subpopulations or states facilitate heart regeneration requires research attention. METHODS To dissect epicardial cell states and associated proregenerative functions, we performed single-cell RNA sequencing and identified 7 epicardial cell clusters in adult zebrafish, 3 of which displayed enhanced cell numbers during regeneration. We identified paralogs of hapln1 as factors associated with the extracellular matrix and largely expressed in cluster 1. We assessed HAPLN1 expression in published single-cell RNA sequencing data sets from different stages and injury states of murine and human hearts, and we performed molecular genetics to determine the requirements for hapln1-expressing cells and functions of each hapln1 paralog. RESULTS A particular cluster of epicardial cells had the strongest association with regeneration and was marked by expression of hapln1a and hapln1b. The hapln1 paralogs are expressed in epicardial cells that enclose dedifferentiated and proliferating cardiomyocytes during regeneration. Induced genetic depletion of hapln1-expressing cells or genetic inactivation of hapln1b altered deposition of the key extracellular matrix component hyaluronic acid, disrupted cardiomyocyte proliferation, and inhibited heart regeneration. We also found that hapln1-expressing epicardial cells first emerge at the juvenile stage, when they associate with and are required for focused cardiomyocyte expansion events that direct maturation of the ventricular wall. CONCLUSIONS Our findings identify a subset of epicardial cells that emerge in postembryonic zebrafish and sponsor regions of active cardiomyogenesis during cardiac growth and regeneration. We provide evidence that, as the heart achieves its mature structure, these cells facilitate hyaluronic acid deposition to support formation of the compact muscle layer of the ventricle. They are also required, along with the function of hapln1b paralog, in the production and organization of hyaluronic acid-containing matrix in cardiac injury sites, enabling normal cardiomyocyte proliferation and muscle regeneration.
Collapse
Affiliation(s)
- Jisheng Sun
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA (J.S., E.A.P., K.E.S., J.W.)
| | - Elizabeth A Peterson
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA (J.S., E.A.P., K.E.S., J.W.)
| | - Annabel Z Wang
- Duke Regeneration Center, Department of Cell Biology, Duke University Medical Center, Durham, NC (A.Z.W., J.O., K.D.P.)
| | - Jianhong Ou
- Duke Regeneration Center, Department of Cell Biology, Duke University Medical Center, Durham, NC (A.Z.W., J.O., K.D.P.)
| | - Kieko E Smith
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA (J.S., E.A.P., K.E.S., J.W.)
| | - Kenneth D Poss
- Duke Regeneration Center, Department of Cell Biology, Duke University Medical Center, Durham, NC (A.Z.W., J.O., K.D.P.)
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA (J.S., E.A.P., K.E.S., J.W.)
| |
Collapse
|
34
|
Chen J, He J, Luo L. Brain vascular damage-induced lymphatic ingrowth is directed by Cxcl12b/Cxcr4a. Development 2022; 149:275687. [PMID: 35694896 DOI: 10.1242/dev.200729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/06/2022] [Indexed: 12/15/2022]
Abstract
After ischemic stroke, promotion of vascular regeneration without causing uncontrolled vessel growth appears to be the major challenge for pro-angiogenic therapies. The molecular mechanisms underlying how nascent blood vessels (BVs) are correctly guided into the post-ischemic infarction area remain unknown. Here, using a zebrafish cerebrovascular injury model, we show that chemokine signaling provides crucial guidance cues to determine the growing direction of ingrown lymphatic vessels (iLVs) and, in turn, that of nascent BVs. The chemokine receptor Cxcr4a is transcriptionally activated in the iLVs after injury, whereas its ligand Cxcl12b is expressed in the residual central BVs, the destinations of iLV ingrowth. Mutant and mosaic studies indicate that Cxcl12b/Cxcr4a-mediated chemotaxis is necessary and sufficient to determine the growing direction of iLVs and nascent BVs. This study provides a molecular basis for how the vessel directionality of cerebrovascular regeneration is properly determined, suggesting potential application of Cxcl12b/Cxcr4a in the development of post-ischemic pro-angiogenic therapies.
Collapse
Affiliation(s)
- Jingying Chen
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China.,University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, 400715 Chongqing, China.,University of Chinese Academy of Sciences (Chongqing), Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Beibei, 400714 Chongqing, China
| |
Collapse
|
35
|
Rathjen T, Kunkemoeller B, Cederquist CT, Wang X, Lockhart SM, Patti JC, Willenbrock H, Olsen GS, Povlsen GK, Beck HC, Rasmussen LM, Li Q, Park K, King GL, Rask-Madsen C. Endothelial Cell Insulin Signaling Regulates CXCR4 (C-X-C Motif Chemokine Receptor 4) and Limits Leukocyte Adhesion to Endothelium. Arterioscler Thromb Vasc Biol 2022; 42:e217-e227. [PMID: 35652755 PMCID: PMC9371472 DOI: 10.1161/atvbaha.122.317476] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND An activated, proinflammatory endothelium is a key feature in the development of complications of obesity and type 2 diabetes and can be caused by insulin resistance in endothelial cells. METHODS We analyzed primary human endothelial cells by RNA sequencing to discover novel insulin-regulated genes and used endothelial cell culture and animal models to characterize signaling through CXCR4 (C-X-C motif chemokine receptor 4) in endothelial cells. RESULTS CXCR4 was one of the genes most potently regulated by insulin, and this was mediated by PI3K (phosphatidylinositol 3-kinase), likely through FoxO1, which bound to the CXCR4 promoter. CXCR4 mRNA in CD31+ cells was 77% higher in mice with diet-induced obesity compared with lean controls and 37% higher in db/db mice than db/+ controls, consistent with upregulation of CXCR4 in endothelial cell insulin resistance. SDF-1 (stromal cell-derived factor-1)-the ligand for CXCR4-increased leukocyte adhesion to cultured endothelial cells. This effect was lost after deletion of CXCR4 by gene editing while 80% of the increase was prevented by treatment of endothelial cells with insulin. In vivo microscopy of mesenteric venules showed an increase in leukocyte rolling after intravenous injection of SDF-1, but most of this response was prevented in transgenic mice with endothelial overexpression of IRS-1 (insulin receptor substrate-1). CONCLUSIONS Endothelial cell insulin signaling limits leukocyte/endothelial cell interaction induced by SDF-1 through downregulation of CXCR4. Improving insulin signaling in endothelial cells or inhibiting endothelial CXCR4 may reduce immune cell recruitment to the vascular wall or tissue parenchyma in insulin resistance and thereby help prevent several vascular complications.
Collapse
Affiliation(s)
- Thomas Rathjen
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.).,Novo Nordisk A/S, Måløv, Denmark (T.R., H.W., G.S.O., G.K.P.)
| | - Britta Kunkemoeller
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Carly T Cederquist
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Xuanchun Wang
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Sam M Lockhart
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - James C Patti
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | | | | | | | | | | | - Qian Li
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Kyoungmin Park
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - George L King
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| | - Christian Rask-Madsen
- Joslin Diabetes Center and Harvard Medical School, Boston, MA (T.R., B.K., C.T.C., X.W., S.M.L., J.C.P., Q.L., K.P., G.L.K., C.R.-M.)
| |
Collapse
|
36
|
Sun J, Peterson EA, Jiao C, Chen X, Zhao Y, Wang J. Zebrafish heart regeneration after coronary dysfunction-induced cardiac damage. Dev Biol 2022; 487:57-66. [PMID: 35490764 PMCID: PMC11017783 DOI: 10.1016/j.ydbio.2022.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/03/2022]
Abstract
Over the past 20 years, various zebrafish injury models demonstrated efficient heart regeneration after cardiac tissue loss. However, no established coronary vessel injury methods exist in the zebrafish model, despite coronary endothelial dysfunction occurring in most patients with acute coronary syndrome. This is due to difficulties performing surgery on small coronary vessels and a lack of genetic tools to precisely manipulate coronary cells in zebrafish. We determined that the Notch ligand gene deltaC regulatory sequences drive gene expression in zebrafish coronary endothelial cells, enabling us to overcome these obstacles. We created a deltaC fluorescent reporter line and visualized robust coronary growth during heart development and regeneration. Importantly, this reporter facilitated the visualization of coronary growth without an endocardial background. Moreover, we visualized robust coronary growth on the surface of juvenile hearts and regrowth in the wounded area of adult hearts ex vivo. With this approach, we observed growth inhibition by reported vascular growth antagonists of the VEGF, EGF and Notch signaling pathways. Furthermore, we established a coronary genetic ablation system and observed that severe coronary endothelial cell loss resulted in fish death, whereas fish survived mild coronary cell loss. Coronary cell depletion triggered regenerative responses, which resulted in the restoration of damaged cardiac tissues within several weeks. Overall, our work demonstrated the efficacy of using deltaC regulatory elements for high-resolution visualization of the coronary endothelium; screening small molecules for coronary growth effects; and revealed complete recovery in adult zebrafish after coronary-induced heart damage.
Collapse
Affiliation(s)
- Jisheng Sun
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Elizabeth A Peterson
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Cheng Jiao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xin Chen
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Yun Zhao
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Jinhu Wang
- Division of Cardiology, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
37
|
Ross Stewart KM, Walker SL, Baker AH, Riley PR, Brittan M. Hooked on heart regeneration: the zebrafish guide to recovery. Cardiovasc Res 2022; 118:1667-1679. [PMID: 34164652 PMCID: PMC9215194 DOI: 10.1093/cvr/cvab214] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/22/2021] [Indexed: 12/23/2022] Open
Abstract
While humans lack sufficient capacity to undergo cardiac regeneration following injury, zebrafish can fully recover from a range of cardiac insults. Over the past two decades, our understanding of the complexities of both the independent and co-ordinated injury responses by multiple cardiac tissues during zebrafish heart regeneration has increased exponentially. Although cardiomyocyte regeneration forms the cornerstone of the reparative process in the injured zebrafish heart, recent studies have shown that this is dependent on prior neovascularization and lymphangiogenesis, which in turn require epicardial, endocardial, and inflammatory cell signalling within an extracellular milieu that is optimized for regeneration. Indeed, it is the amalgamation of multiple regenerative systems and gene regulatory patterns that drives the much-heralded success of the adult zebrafish response to cardiac injury. Increasing evidence supports the emerging paradigm that developmental transcriptional programmes are re-activated during adult tissue regeneration, including in the heart, and the zebrafish represents an optimal model organism to explore this concept. In this review, we summarize recent advances from the zebrafish cardiovascular research community with novel insight into the mechanisms associated with endogenous cardiovascular repair and regeneration, which may be of benefit to inform future strategies for patients with cardiovascular disease.
Collapse
Affiliation(s)
- Katherine M Ross Stewart
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Sophie L Walker
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Paul R Riley
- Department of Physiology, Anatomy & Genetics, University of Oxford, Sherrington Building, Sherrington Rd, Oxford OX1 3PT, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
38
|
Abstract
Heart disease is the leading cause of death worldwide. Despite decades of research, most heart pathologies have limited treatments, and often the only curative approach is heart transplantation. Thus, there is an urgent need to develop new therapeutic approaches for treating cardiac diseases. Animal models that reproduce the human pathophysiology are essential to uncovering the biology of diseases and discovering therapies. Traditionally, mammals have been used as models of cardiac disease, but the cost of generating and maintaining new models is exorbitant, and the studies have very low throughput. In the last decade, the zebrafish has emerged as a tractable model for cardiac diseases, owing to several characteristics that made this animal popular among developmental biologists. Zebrafish fertilization and development are external; embryos can be obtained in high numbers, are cheap and easy to maintain, and can be manipulated to create new genetic models. Moreover, zebrafish exhibit an exceptional ability to regenerate their heart after injury. This review summarizes 25 years of research using the zebrafish to study the heart, from the classical forward screenings to the contemporary methods to model mutations found in patients with cardiac disease. We discuss the advantages and limitations of this model organism and introduce the experimental approaches exploited in zebrafish, including forward and reverse genetics and chemical screenings. Last, we review the models used to induce cardiac injury and essential ideas derived from studying natural regeneration. Studies using zebrafish have the potential to accelerate the discovery of new strategies to treat cardiac diseases.
Collapse
Affiliation(s)
- Juan Manuel González-Rosa
- Cardiovascular Research Center, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA
| |
Collapse
|
39
|
Perl E, Ravisankar P, Beerens ME, Mulahasanovic L, Smallwood K, Sasso MB, Wenzel C, Ryan TD, Komár M, Bove KE, MacRae CA, Weaver KN, Prada CE, Waxman JS. Stx4 is required to regulate cardiomyocyte Ca 2+ handling during vertebrate cardiac development. HGG ADVANCES 2022; 3:100115. [PMID: 35599850 PMCID: PMC9114686 DOI: 10.1016/j.xhgg.2022.100115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022] Open
Abstract
Requirements for vesicle fusion within the heart remain poorly understood, despite the multitude of processes that necessitate proper intracellular trafficking within cardiomyocytes. Here, we show that Syntaxin 4 (STX4), a target-Soluble N-ethylmaleimide sensitive factor attachment receptor (t-SNARE) protein, is required for normal vertebrate cardiac conduction and vesicular transport. Two patients were identified with damaging variants in STX4. A patient with a homozygous R240W missense variant displayed biventricular dilated cardiomyopathy, ectopy, and runs of non-sustained ventricular tachycardia, sensorineural hearing loss, global developmental delay, and hypotonia, while a second patient displayed severe pleiotropic abnormalities and perinatal lethality. CRISPR/Cas9-generated stx4 mutant zebrafish exhibited defects reminiscent of these patients' clinical presentations, including linearized hearts, bradycardia, otic vesicle dysgenesis, neuronal atrophy, and touch insensitivity by 3 days post fertilization. Imaging of Vamp2+ vesicles within stx4 mutant zebrafish hearts showed reduced docking to the cardiomyocyte sarcolemma. Optical mapping of the embryonic hearts coupled with pharmacological modulation of Ca2+ handling together support that zebrafish stx4 mutants have a reduction in L-type Ca2+ channel modulation. Transgenic overexpression of zebrafish Stx4R241W, analogous to the first patient's STX4R240W variant, indicated that the variant is hypomorphic. Thus, these data show an in vivo requirement for SNAREs in regulating normal embryonic cardiac function and that variants in STX4 are associated with pleiotropic human disease, including cardiomyopathy.
Collapse
Affiliation(s)
- Eliyahu Perl
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Padmapriyadarshini Ravisankar
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Manu E. Beerens
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Lejla Mulahasanovic
- Praxis für Humangenetik, Tübingen, Baden-Württemberg, Germany,CeGaT GmbH, Tübingen, Baden-Württemberg, Germany
| | - Kelly Smallwood
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Marion Bermúdez Sasso
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Saxony, Germany
| | - Carina Wenzel
- Institute of Pathology, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
| | - Thomas D. Ryan
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA,Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Matej Komár
- Department of Gynecology and Obstetrics, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Saxony, Germany
| | - Kevin E. Bove
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA,Division of Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pathology and Laboratory Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Calum A. MacRae
- Cardiovascular Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA,Genetics and Network Medicine Divisions, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA,Harvard Stem Cell Institute, Boston, MA, USA
| | - K. Nicole Weaver
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Carlos E. Prada
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA
| | - Joshua S. Waxman
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH, USA,Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA,Corresponding author
| |
Collapse
|
40
|
Tian X, Zhou B. Coronary vessel formation in development and regeneration: origins and mechanisms. J Mol Cell Cardiol 2022; 167:67-82. [DOI: 10.1016/j.yjmcc.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
41
|
Kapuria S, Bai H, Fierros J, Huang Y, Ma F, Yoshida T, Aguayo A, Kok F, Wiens KM, Yip JK, McCain ML, Pellegrini M, Nagashima M, Hitchcock PF, Mochizuki N, Lawson ND, Harrison MMR, Lien CL. Heterogeneous pdgfrb+ cells regulate coronary vessel development and revascularization during heart regeneration. Development 2022; 149:274137. [PMID: 35088848 PMCID: PMC8918812 DOI: 10.1242/dev.199752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022]
Abstract
Endothelial cells emerge from the atrioventricular canal to form coronary blood vessels in juvenile zebrafish hearts. We find that pdgfrb is first expressed in the epicardium around the atrioventricular canal and later becomes localized mainly in the mural cells. pdgfrb mutant fish show severe defects in mural cell recruitment and coronary vessel development. Single-cell RNA sequencing analyses identified pdgfrb+ cells as epicardium-derived cells (EPDCs) and mural cells. Mural cells associated with coronary arteries also express cxcl12b and smooth muscle cell markers. Interestingly, these mural cells remain associated with coronary arteries even in the absence of Pdgfrβ, although smooth muscle gene expression is downregulated. We find that pdgfrb expression dynamically changes in EPDCs of regenerating hearts. Differential gene expression analyses of pdgfrb+ EPDCs and mural cells suggest that they express genes that are important for regeneration after heart injuries. mdka was identified as a highly upregulated gene in pdgfrb+ cells during heart regeneration. However, pdgfrb but not mdka mutants show defects in heart regeneration after amputation. Our results demonstrate that heterogeneous pdgfrb+ cells are essential for coronary development and heart regeneration.
Collapse
Affiliation(s)
- Subir Kapuria
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Authors for correspondence (; ; )
| | - Haipeng Bai
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Laboratory of Chemical Genomics, School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, People's Republic of China
| | - Juancarlos Fierros
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Department of Biology, California State University, San Bernardino, San Bernardino, CA 92407, USA
| | - Ying Huang
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tyler Yoshida
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Department of Biological Sciences, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA 90007, USA
| | - Antonio Aguayo
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA
| | - Fatma Kok
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Katie M. Wiens
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Science Department, Bay Path University, Longmeadow, MA 01106, USA
| | - Joycelyn K. Yip
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, College of Letters and Sciences, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Mikiko Nagashima
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Peter F. Hitchcock
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Osaka, 564-8565, Japan
| | - Nathan D. Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Michael M. R. Harrison
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Authors for correspondence (; ; )
| | - Ching-Ling Lien
- Department of Surgery, The Saban Research Institute and Heart Institute of Children's Hospital Los Angeles, Los Angeles, CA 90027, USA,Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA,Authors for correspondence (; ; )
| |
Collapse
|
42
|
Hoareau M, El Kholti N, Debret R, Lambert E. Zebrafish as a Model to Study Vascular Elastic Fibers and Associated Pathologies. Int J Mol Sci 2022; 23:2102. [PMID: 35216218 PMCID: PMC8875079 DOI: 10.3390/ijms23042102] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 02/06/2023] Open
Abstract
Many extensible tissues such as skin, lungs, and blood vessels require elasticity to function properly. The recoil of elastic energy stored during a stretching phase is provided by elastic fibers, which are mostly composed of elastin and fibrillin-rich microfibrils. In arteries, the lack of elastic fibers leads to a weakening of the vessel wall with an increased risk to develop cardiovascular defects such as stenosis, aneurysms, and dissections. The development of new therapeutic molecules involves preliminary tests in animal models that recapitulate the disease and whose response to drugs should be as close as possible to that of humans. Due to its superior in vivo imaging possibilities and the broad tool kit for forward and reverse genetics, the zebrafish has become an important model organism to study human pathologies. Moreover, it is particularly adapted to large scale studies, making it an attractive model in particular for the first steps of investigations. In this review, we discuss the relevance of the zebrafish model for the study of elastic fiber-related vascular pathologies. We evidence zebrafish as a compelling alternative to conventional mouse models.
Collapse
Affiliation(s)
- Marie Hoareau
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| | | | | | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Institut de Biologie et Chimie des Protéines, Université Lyon 1, 7, Passage du Vercors, CEDEX 07, F-69367 Lyon, France; (N.E.K.); (R.D.)
| |
Collapse
|
43
|
Riley SE, Feng Y, Hansen CG. Hippo-Yap/Taz signalling in zebrafish regeneration. NPJ Regen Med 2022; 7:9. [PMID: 35087046 PMCID: PMC8795407 DOI: 10.1038/s41536-022-00209-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/14/2021] [Indexed: 12/29/2022] Open
Abstract
The extent of tissue regeneration varies widely between species. Mammals have a limited regenerative capacity whilst lower vertebrates such as the zebrafish (Danio rerio), a freshwater teleost, can robustly regenerate a range of tissues, including the spinal cord, heart, and fin. The molecular and cellular basis of this altered response is one of intense investigation. In this review, we summarise the current understanding of the association between zebrafish regeneration and Hippo pathway function, a phosphorylation cascade that regulates cell proliferation, mechanotransduction, stem cell fate, and tumorigenesis, amongst others. We also compare this function to Hippo pathway activity in the regenerative response of other species. We find that the Hippo pathway effectors Yap/Taz facilitate zebrafish regeneration and that this appears to be latent in mammals, suggesting that therapeutically promoting precise and temporal YAP/TAZ signalling in humans may enhance regeneration and hence reduce morbidity.
Collapse
Affiliation(s)
- Susanna E Riley
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Yi Feng
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Carsten Gram Hansen
- University of Edinburgh Centre for Inflammation Research, Institute for Regeneration and Repair, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
44
|
Chowdhury K, Lai SL, Marín-Juez R. Modulation of VEGFA Signaling During Heart Regeneration in Zebrafish. Methods Mol Biol 2022; 2475:297-312. [PMID: 35451767 DOI: 10.1007/978-1-0716-2217-9_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last decades, myocardial infarction and heart failure have accounted every year for millions of deaths worldwide. After a coronary occlusion, the lack of blood supply to downstream muscle leads to cell death and scarring. To date, several pro-angiogenic factors have been tested to stimulate reperfusion of the affected myocardium, VEGFA being one of the most extensively studied. Given the unsuccessful outcomes of clinical trials, understanding how cardiac revascularization takes place in models with endogenous regenerative capacity holds the key to devising more efficient therapies. Here, we summarize the main findings on VEGFA's role during cardiac repair and regeneration, with a particular focus on zebrafish as a regenerative model. Moreover, we provide a comprehensive overview of available tools to modulate Vegfa expression and action in zebrafish regeneration studies. Understanding the role of Vegfa during zebrafish heart regeneration may help devise efficient therapies and circumvent current limitations in using VEGFA for therapeutic angiogenesis approaches.
Collapse
Affiliation(s)
- Kaushik Chowdhury
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, QC, Canada.
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
45
|
Ma H, Liu Z, Yang Y, Feng D, Dong Y, Garbutt TA, Hu Z, Wang L, Luan C, Cooper CD, Li Y, Welch JD, Qian L, Liu J. Functional coordination of non-myocytes plays a key role in adult zebrafish heart regeneration. EMBO Rep 2021; 22:e52901. [PMID: 34523214 DOI: 10.15252/embr.202152901] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac regeneration occurs primarily through proliferation of existing cardiomyocytes, but also involves complex interactions between distinct cardiac cell types including non-cardiomyocytes (non-CMs). However, the subpopulations, distinguishing molecular features, cellular functions, and intercellular interactions of non-CMs in heart regeneration remain largely unexplored. Using the LIGER algorithm, we assemble an atlas of cell states from 61,977 individual non-CM scRNA-seq profiles isolated at multiple time points during regeneration. This analysis reveals extensive non-CM cell diversity, including multiple macrophage (MC), fibroblast (FB), and endothelial cell (EC) subpopulations with unique spatiotemporal distributions, and suggests an important role for MC in inducing the activated FB and EC subpopulations. Indeed, pharmacological perturbation of MC function compromises the induction of the unique FB and EC subpopulations. Furthermore, we developed computational algorithm Topologizer to map the topological relationships and dynamic transitions between functional states. We uncover dynamic transitions between MC functional states and identify factors involved in mRNA processing and transcriptional regulation associated with the transition. Together, our single-cell transcriptomic analysis of non-CMs during cardiac regeneration provides a blueprint for interrogating the molecular and cellular basis of this process.
Collapse
Affiliation(s)
- Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ziqing Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yuchen Yang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dong Feng
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanhan Dong
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tiffany A Garbutt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Li Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Changfei Luan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Cynthia D Cooper
- School of Molecular Biosciences, Washington State University Vancouver, Vancouver, WA, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA.,Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
46
|
Tsedeke AT, Allanki S, Gentile A, Jimenez-Amilburu V, Rasouli SJ, Guenther S, Lai SL, Stainier DY, Marín-Juez R. Cardiomyocyte heterogeneity during zebrafish development and regeneration. Dev Biol 2021; 476:259-271. [DOI: 10.1016/j.ydbio.2021.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/22/2021] [Accepted: 03/19/2021] [Indexed: 12/31/2022]
|
47
|
Ando K, Shih YH, Ebarasi L, Grosse A, Portman D, Chiba A, Mattonet K, Gerri C, Stainier DYR, Mochizuki N, Fukuhara S, Betsholtz C, Lawson ND. Conserved and context-dependent roles for pdgfrb signaling during zebrafish vascular mural cell development. Dev Biol 2021; 479:11-22. [PMID: 34310924 DOI: 10.1016/j.ydbio.2021.06.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 12/27/2022]
Abstract
Platelet derived growth factor beta and its receptor, Pdgfrb, play essential roles in the development of vascular mural cells, including pericytes and vascular smooth muscle cells. To determine if this role was conserved in zebrafish, we analyzed pdgfb and pdgfrb mutant lines. Similar to mouse, pdgfb and pdgfrb mutant zebrafish lack brain pericytes and exhibit anatomically selective loss of vascular smooth muscle coverage. Despite these defects, pdgfrb mutant zebrafish did not otherwise exhibit circulatory defects at larval stages. However, beginning at juvenile stages, we observed severe cranial hemorrhage and vessel dilation associated with loss of pericytes and vascular smooth muscle cells in pdgfrb mutants. Similar to mouse, pdgfrb mutant zebrafish also displayed structural defects in the glomerulus, but normal development of hepatic stellate cells. We also noted defective mural cell investment on coronary vessels with concomitant defects in their development. Together, our studies support a conserved requirement for Pdgfrb signaling in mural cells. In addition, these zebrafish mutants provide an important model for definitive investigation of mural cells during early embryonic stages without confounding secondary effects from circulatory defects.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan.
| | - Yu-Huan Shih
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Lwaki Ebarasi
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institute, Stockholm, Sweden
| | - Ann Grosse
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Daneal Portman
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Kenny Mattonet
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Claudia Gerri
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany; Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, 564 8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Sendagi Bunkyo-ku, Tokyo, 113 8602, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds Väg 20, SE-751 85, Uppsala, Sweden; Department of Medicine Huddinge (MedH), Karolinska Institutet, Campus Flemingsberg, Neo, Blickagången 16, Hiss S, Plan 7, SE-141 57, Huddinge, Sweden
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01650, United States.
| |
Collapse
|
48
|
Stone OA, Zhou B, Red-Horse K, Stainier DYR. Endothelial ontogeny and the establishment of vascular heterogeneity. Bioessays 2021; 43:e2100036. [PMID: 34145927 DOI: 10.1002/bies.202100036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
The establishment of distinct cellular identities was pivotal during the evolution of Metazoa, enabling the emergence of an array of specialized tissues with different functions. In most animals including vertebrates, cell specialization occurs in response to a combination of intrinsic (e.g., cellular ontogeny) and extrinsic (e.g., local environment) factors that drive the acquisition of unique characteristics at the single-cell level. The first functional organ system to form in vertebrates is the cardiovascular system, which is lined by a network of endothelial cells whose organ-specific characteristics have long been recognized. Recent genetic analyses at the single-cell level have revealed that heterogeneity exists not only at the organ level but also between neighboring endothelial cells. Thus, how endothelial heterogeneity is established has become a key question in vascular biology. Drawing upon evidence from multiple organ systems, here we will discuss the role that lineage history may play in establishing endothelial heterogeneity.
Collapse
Affiliation(s)
- Oliver A Stone
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kristy Red-Horse
- Department of Biology, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
49
|
Li W, Liu C, Burns N, Hayashi J, Yoshida A, Sajja A, González-Hernández S, Gao JL, Murphy PM, Kubota Y, Zou YR, Nagasawa T, Mukouyama YS. Alterations in the spatiotemporal expression of the chemokine receptor CXCR4 in endothelial cells cause failure of hierarchical vascular branching. Dev Biol 2021; 477:70-84. [PMID: 34015362 DOI: 10.1016/j.ydbio.2021.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/18/2022]
Abstract
The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.
Collapse
Affiliation(s)
- Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, USA
| | - Nathan Burns
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Jeffery Hayashi
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Atsufumi Yoshida
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Aparna Sajja
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Sara González-Hernández
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA
| | - Ji-Liang Gao
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yoshiaki Kubota
- Department of Anatomy, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yong-Rui Zou
- The Feinstein Institute for Medical Research, Center for Autoimmune and Musculoskeletal Diseases, Manhasset, NY 11030, USA
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences, Graduate School of Medicine, Immunology Frontier Research Center, World Premier International Research Center, Osaka University, Osaka 565-0871, Japan
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Development Biology Center, USA.
| |
Collapse
|
50
|
Grivas D, González-Rajal Á, de la Pompa JL. Midkine-a Regulates the Formation of a Fibrotic Scar During Zebrafish Heart Regeneration. Front Cell Dev Biol 2021; 9:669439. [PMID: 34026760 PMCID: PMC8138450 DOI: 10.3389/fcell.2021.669439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/07/2021] [Indexed: 01/22/2023] Open
Abstract
Unlike the hearts of mammals, the adult zebrafish heart regenerates after injury. Heart cryoinjury in zebrafish triggers the formation of a fibrotic scar that gradually degrades, leading to regeneration. Midkine-a (Mdka) is a multifunctional cytokine that is activated after cardiac injury. Here, we investigated the role of mdka in zebrafish heart regeneration. We show that mdka expression was induced at 1-day post-cryoinjury (dpci) throughout the epicardial layer, whereas by 7 dpci expression had become restricted to the epicardial cells covering the injured area. To study the role of mdka in heart regeneration, we generated mdka-knock out (KO) zebrafish strains. Analysis of injured hearts showed that loss of mdka decreased endothelial cell proliferation and resulted in an arrest in heart regeneration characterized by retention of a collagenous scar. Transcriptional analysis revealed increases in collagen transcription and intense TGFβ signaling activity. These results reveal a critical role for mdka in fibrosis regulation during heart regeneration.
Collapse
Affiliation(s)
- Dimitrios Grivas
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, Madrid, Spain.,Developmental Biology, Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Álvaro González-Rajal
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Ciber de Enfermedades Cardiovasculares, Madrid, Spain
| |
Collapse
|