1
|
Griffin AHC, Small AM, Johnson RD, Medina AM, Kollar KT, Nazir RA, McGuire AM, Schumacher JA. Retinoic acid promotes second heart field addition and regulates ventral aorta patterning in zebrafish. Dev Biol 2025; 522:143-155. [PMID: 40147741 DOI: 10.1016/j.ydbio.2025.03.013] [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: 07/21/2024] [Revised: 03/09/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Retinoic acid (RA) signaling is used reiteratively during vertebrate heart development. Its earliest known role is to restrict formation of the earlier-differentiating first heart field (FHF) progenitors, while promoting the differentiation of second heart field (SHF) progenitors that give rise to the arterial pole of the ventricle and outflow tract (OFT). However, requirements for RA signaling at later stages of cardiogenesis remain poorly understood. Here, we investigated the role of RA signaling after the later differentiating SHF cells have begun to add to the OFT. We found that inhibiting RA production in zebrafish beginning at 26 hours post fertilization (hpf) produced embryos that have smaller ventricles with fewer ventricular cardiomyocytes, and reduced number of smooth muscle cells in the bulbus arteriosus (BA) of the OFT. Our results suggest that the deficiency of the ventricular cardiomyocytes is due to reduced SHF addition to the arterial pole. In contrast to smaller ventricles and BA, later RA deficiency also results in a dramatically elongated posterior branch of the adjacent ventral aorta, which is surrounded by an increased number of smooth muscle cells. Altogether, our results reveal that RA signaling is required during the period of SHF addition to promote addition of ventricular cardiomyocytes, partition smooth muscle cells onto the BA and posterior ventral aorta, and to establish proper ventral aorta anterior-posterior patterning.
Collapse
Affiliation(s)
| | - Allison M Small
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Riley D Johnson
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Anna M Medina
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Kiki T Kollar
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | - Ridha A Nazir
- Department of Biology, Miami University, Oxford, OH, 45056, USA
| | | | - Jennifer A Schumacher
- Department of Biology, Miami University, Oxford, OH, 45056, USA; Department of Biological Sciences, Miami University, Hamilton, OH, 45011, USA.
| |
Collapse
|
2
|
Ampuja M, Ericsson S, Paatero I, Chowdhury I, Villman J, Broberg M, Ramste A, Balboa D, Ojala T, Chong JX, Bamshad MJ, Priest JR, Varjosalo M, Kivelä R, Helle E. The ERBB2 c.1795C>T, p.Arg599Cys variant is associated with left ventricular outflow tract obstruction defects in humans. HGG ADVANCES 2025; 6:100446. [PMID: 40329538 DOI: 10.1016/j.xhgg.2025.100446] [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: 07/16/2024] [Revised: 04/30/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
Non-syndromic congenital heart defects (CHDs) are occasionally familial and left ventricular outflow tract obstruction (LVOTO) defects are among the subtypes with the highest hereditability. The aim of this study was to evaluate the pathogenicity of a heterozygous ERBB2 variant c.1795C>T, p.Arg599Cys identified in three families with LVOTO defects. Variant detection was done with exome sequencing. Western blotting, digital PCR, mass spectrometry (MS), MS microscopy, and flow cytometry were used to study the function of the ERBB2 variant c.1795C>T. Cardiac structure and function were studied in zebrafish embryos expressing human ERBB2 wild type or c.1795C>T. Proband-derived human induced pluripotent stem cell cardiomyocytes (hiPS-CMs) and endothelial cells (hiPS-ECs) were used for transcriptomic analyses. While phosphorylation of the ERBB2 p.Arg599Cys receptor was not altered, the variant affected dramatically the binding partners of the protein, indicating mislocalization of the mutant ERBB2 from plasma membrane to endoplasmic reticulum. Expression of human ERBB2 p.Arg599Cys in zebrafish embryos resulted in cardiomyocyte hypertrophy, increased cardiac wall thickness, and impaired fractional shortening. Transcriptomic analyses of hiPS-ECs and hiPS-CMs from an individual with the c.1795C>T variant showed aberrant expression of genes related to cardiovascular system development and abnormal response to oxidative stress in both cell types. In conclusion, the heterozygous variant ERBB2 c.1795C>T, p.Arg599Cys leads to abnormal cellular localization of the ERBB2 receptor and induces structural changes and dysfunction in the zebrafish embryo heart. This evidence expands previous findings from animal studies to humans and suggests variants in ERBB2 may be associated with CHD.
Collapse
Affiliation(s)
- Minna Ampuja
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sabina Ericsson
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilkka Paatero
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Iftekhar Chowdhury
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jenna Villman
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Martin Broberg
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Amanda Ramste
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Diego Balboa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tiina Ojala
- Children's Hospital, Paediatric Research Centre, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jessica X Chong
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Michael J Bamshad
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James R Priest
- Tenaya Therapeutics, 171 Oyster Point Boulevard Suite 500, South San Francisco, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Riikka Kivelä
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Wihuri Research Institute, Helsinki, Finland; Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland
| | - Emmi Helle
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Children's Hospital, Paediatric Research Centre, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland.
| |
Collapse
|
3
|
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
|
4
|
Mukherjee S, Das S, Das S, Gupta S, Hui SP, Sengupta A, Ghosh A. Pyruvate plus uridine augments mitochondrial respiration and prevents cardiac hypertrophy in zebrafish and H9c2 cells. J Cell Sci 2025; 138:jcs263653. [PMID: 40270134 DOI: 10.1242/jcs.263653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
Dysfunction of mitochondrial pyruvate oxidation and aberrant respiratory chain components are common in cardiac defects. However, the precise role of mitochondrial respiration in cardiomyocyte hypertrophy is unclear. Phenylephrine (PE) treatment of rat neonatal H9c2 cardiomyocytes promotes significant hypertrophy with decreased mitochondrial oxygen consumption rate (OCR), membrane potential, respiratory subunit NDUFB8, UQCRC2 and ATP5A (ATP5F1A) expression, and accumulation of reactive oxygen species (ROS). Surprisingly, a 60% reduction in cell survival was observed in PE-treated cells relative to control cells when grown under the respiratory-proficient galactose medium. To revert H9c2 hypertrophy and increase survival, we performed a screening with compounds that boost mitochondrial OCR and scavenge ROS, and identified pyruvate plus uridine as the best hit. As corroboration of the in vitro study, supplementation of pyruvate plus uridine significantly prevented PE-induced cardiac hypertrophy, pericardial edema and bradycardia symptoms in zebrafish embryos. Moreover, pyruvate plus uridine decreased the ventricular and atrial area in cardiomyocyte-specific GFP transgenic Tg (myl7:HRAS-EGFP) lines. Using in vitro and in vivo models, we show that boosting of mitochondrial respiration through pyruvate supplementation and scavenging ROS through uridine supplementation jointly ameliorate cardiac hypertrophy and bradycardia symptoms.
Collapse
Affiliation(s)
- Soumyajit Mukherjee
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Shreya Das
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Surajit Das
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Samudra Gupta
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Subhra Prakash Hui
- S.N. Pradhan Centre for Neurosciences, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Arunima Sengupta
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Alok Ghosh
- Department of Biochemistry, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| |
Collapse
|
5
|
Angom RS, Singh M, Muhammad H, Varanasi SM, Mukhopadhyay D. Zebrafish as a Versatile Model for Cardiovascular Research: Peering into the Heart of the Matter. Cells 2025; 14:531. [PMID: 40214485 PMCID: PMC11988917 DOI: 10.3390/cells14070531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. A total of 17.5 million people died of CVDs in the year 2012, accounting for 31% of all deaths globally. Vertebrate animal models have been used to understand cardiac disease biology, as the cellular, molecular, and physiological aspects of human CVDs can be replicated closely in these organisms. Zebrafish is a popular model organism offering an arsenal of genetic tools that allow the rapid in vivo analysis of vertebrate gene function and disease conditions. It has a short breeding cycle, high fecundity, optically transparent embryos, rapid internal organ development, and easy maintenance. This review aims to give readers an overview of zebrafish cardiac biology and a detailed account of heart development in zebrafish and its comparison with humans and the conserved genetic circuitry. We also discuss the contributions made in CVD research using the zebrafish model. The first part of this review focuses on detailed information on the morphogenetic and differentiation processes in early cardiac development. The overlap and divergence of the human heart's genetic circuitry, structure, and physiology are emphasized wherever applicable. In the second part of the review, we overview the molecular tools and techniques available to dissect gene function and expression in zebrafish, with special mention of the use of these tools in cardiac biology.
Collapse
Affiliation(s)
- Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Meghna Singh
- Department of Pathology and Lab Medicine, University of California, Los Angeles, CA 92093, USA;
| | - Huzaifa Muhammad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Sai Manasa Varanasi
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, College of Medicine and Science, Jacksonville, FL 32224, USA; (R.S.A.); (H.M.); (S.M.V.)
| |
Collapse
|
6
|
Li X, Yang S, Wang L, Zhang X, Zhang A, Wang Y, Shi DL, Li H. Zinc Finger Protein Znf296 Is a Cardiac-Specific Splicing Regulator Required for Cardiomyocyte Formation. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00078-1. [PMID: 40122456 DOI: 10.1016/j.ajpath.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/10/2025] [Accepted: 02/19/2025] [Indexed: 03/25/2025]
Abstract
Heart formation and function are tightly regulated at transcriptional and post-transcriptional levels. The dysfunction of cardiac cell-specific regulatory genes leads to various heart diseases. Heart failure is one of the most severe and complex cardiovascular diseases, which could be fatal if not treated promptly. However, the exact causes of heart failure are still unclear, especially at the level of single-gene causation. Here, an essential role is uncovered for the zinc finger protein Znf296 in heart development and cardiac contractile function. Specifically, znf296-deficient zebrafish embryos display heart defects characterized by decreased systolic and diastolic capacities of the ventricle and atrium. This is associated with reduced numbers and disrupted structural integrity of cardiomyocytes, including disorganized cytoskeleton and absence of sarcomeres. Mechanistically, the loss of Znf296 alters the alternative splicing of a subset of genes important for heart development and disease, such as mef2ca, sparc, tpm2, camk2g1, tnnt3b, and pdlim5b. Furthermore, it is demonstrated that Znf296 biochemically and functionally interacts with Myt1la in regulating cardiac-specific splicing and heart development. Importantly, it is shown that ZNF296 also regulates alternative splicing in human cardiomyocytes to maintain structural integrity. These results suggest that Znf296 plays a conserved role for the differentiation of cardiomyocytes and the proper function of the cardiovascular system.
Collapse
Affiliation(s)
- Xianpeng Li
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Institute of Brain Science and Brain-Inspired Research, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuaiqi Yang
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China
| | - Lu Wang
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China
| | - Xiangmin Zhang
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China
| | - Ailong Zhang
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China
| | - Yunchao Wang
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China
| | - De-Li Shi
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China; Sorbonne Université, Institut de Biologie Paris-Seine, UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France.
| | - Hongyan Li
- College of Marine Life Sciences, Institute of Evolution and Marine Biodiversity and Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Ocean University of China, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
7
|
Wang X, Kang X, Li B, Chen C, Chen L, Liu D. High Glucose Treatment Induces Nuclei Aggregation of Microvascular Endothelial Cells via the foxo1a- klf2a Pathway. Arterioscler Thromb Vasc Biol 2025; 45:398-411. [PMID: 39882604 PMCID: PMC11856006 DOI: 10.1161/atvbaha.124.321719] [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: 11/05/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Hyperglycemia is a major contributor to endothelial dysfunction and blood vessel damage, leading to severe diabetic microvascular complications. Despite the growing body of research on the underlying mechanisms of endothelial cell (EC) dysfunction, the available drugs based on current knowledge fall short of effectively alleviating these complications. Therefore, our endeavor to explore novel insights into the cellular and molecular mechanisms of endothelial dysfunction is crucial for the field. METHODS In this study, we performed a high-resolution imaging and time-lapse imaging analysis of the behavior of ECs in Tg(kdrl:ras-mCherry::fli1a:nGFP) zebrafish embryos upon high glucose treatment. Genetic manipulation and chemical biology approaches were utilized to analyze the underlying mechanism of high glucose-induced nuclei aggregation and aberrant migration of zebrafish ECs and cultured human ECs. Bioinformatical analysis of single-cell RNA-sequencing data and molecular biological techniques was performed to identify the target genes of foxo1a. RESULTS In this study, we observed that the high glucose treatment resulted in nuclei aggregation of ECs in zebrafish intersegmental vessels. Additionally, the aberrant migration of microvascular ECs in high glucose-treated embryos, which might be a cause of nuclei aggregation, was discovered. High glucose induced aggregation of vascular endothelial nuclei via foxo1a downregulation in zebrafish embryos. Then, we revealed that high glucose resulted in the downregulation of foxo1a expression and increased the expression of its direct downstream effector, klf2a, through which the aberrant migration and aggregation of vascular endothelial nuclei were caused. CONCLUSIONS High glucose treatment caused the nuclei of ECs to aggregate in vivo, which resembles the crowded nuclei of ECs in microaneurysms. High glucose suppresses foxo1a expression and increases the expression of its downstream effector, klf2a, thereby causing the aberrant migration and aggregation of vascular endothelial nuclei. Our findings provide a novel insight into the mechanism of microvascular complications in hyperglycemia.
Collapse
Affiliation(s)
- Xiaoning Wang
- Research Center of Clinical Medicine, Affiliated Hospital (X.W., D.L.), Nantong University, China
| | - Xinyi Kang
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| | - Bowen Li
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
| | - Changsheng Chen
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
| | - Liping Chen
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| | - Dong Liu
- Research Center of Clinical Medicine, Affiliated Hospital (X.W., D.L.), Nantong University, China
- School of Life Science, Nantong Laboratory of Development and Diseases, The Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration (B.L., C.C., D.L.), Nantong University, China
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Nantong University, China (X.K., L.C., D.L.)
| |
Collapse
|
8
|
Li S, Li X, Zhao R, Jiang T, Ou Q, Huang H, Tang J. Esketamine induces embryonic and cardiac malformation through regulating the nkx2.5 and gata4 in zebrafish. Sci Rep 2025; 15:7187. [PMID: 40021926 PMCID: PMC11871044 DOI: 10.1038/s41598-025-91315-2] [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: 08/25/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
Esketamine (EK) has been widely used in the treatment of depression, but the effects of EK prenatal treatment on embryonic heart development have been rarely reported. This study assesses the effects of varying concentrations of EK on embryonic development and cardiogenesis to determine the teratogenic concentration in the zebrafish model, centering on the interaction between the genes nkx2.5 and gata4 to elucidate the mechanisms underlying cardiac morphogenesis. Zebrafish embryos were classified into six distinct groups and exposed to either a vehicle or EK to ascertain the median lethal concentration (LC50) at 48 and 72 h post-fertilization (hpf) analyzing mortality rate data. Embryonic and cardiac morphologies were assessed utilizing live embryo imaging techniques and stereo microscopy. Nkx2.5 and gata4 were identified via whole-mount in situ hybridization (WISH) and reverse transcription quantitative polymerase chain reaction (RT-qPCR). Exposure to EK leads to significant teratogenic effects on zebrafish embryos, which are both concentration- and time-dependent. The 48 h- and 72 h-LC50 of EK for zebrafish embryos were 1.30 (95% CI 0.92, 1.60) millimolar (mM) and 0.71 (95% CI 0.46, 1.01) mM, respectively. A significant reduction in heart rates and body length were observed and the distance between the sinus venosus and bulbar artery (SV-BA) was found expanded, the pericardial edema area showed significant swelling, and the body axis curvature was more pronounced in the EK exposure groups. Both WISH an RT-qPCR analysis showed nkx2.5 staining intensity and expression significantly decreased, while gata4 assay results were in the opposite direction. Our findings indicate that exposure of zebrafish embryos to EK results in embryonic and cardiac malformations, primarily due to the down-regulation of nkx2.5 and the over-expression of gata4. Equilibrium maintenance and compensatory mechanisms are crucial in spatiotemporal gene regulation.
Collapse
Affiliation(s)
- Shuang Li
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiang Li
- Department of Anesthesiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Rui Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Tingyu Jiang
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qiuyun Ou
- Second Clinical College of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Huansen Huang
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China.
| | - Jiancheng Tang
- Department of Anesthesiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China.
| |
Collapse
|
9
|
Hong T, Park J, An G, Song J, Song G, Lim W. Evaluation of organ developmental toxicity of environmental toxicants using zebrafish embryos. Mol Cells 2024; 47:100144. [PMID: 39489379 PMCID: PMC11635654 DOI: 10.1016/j.mocell.2024.100144] [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: 08/08/2024] [Revised: 10/04/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024] Open
Abstract
There is increasing global concern about environmental pollutants, such as heavy metals, plastics, pharmaceuticals, personal care products, and pesticides, which have been detected in a variety of environments and are likely to be exposed to nontarget organisms, including humans. Various animal models have been utilized for toxicity assessment, and zebrafish are particularly valuable for studying the toxicity of various compounds owing to their similarity to other aquatic organisms and 70% genetic similarity to humans. Their development is easy to observe, and transgenic models for organs such as the heart, liver, blood vessels, and nervous system enable efficient studies of organ-specific toxicity. This suggests that zebrafish are a valuable tool for evaluating toxicity in specific organs and forecasting the potential impacts on other nontarget species. This review describes organ toxicity caused by various toxic substances and their mechanisms in zebrafish.
Collapse
Affiliation(s)
- Taeyeon Hong
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Junho Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Garam An
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jisoo Song
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
10
|
Saberigarakani A, Patel RP, Almasian M, Zhang X, Brewer J, Hassan SS, Chai J, Lee J, Fei B, Yuan J, Carroll K, Ding Y. Volumetric imaging and computation to explore contractile function in zebrafish hearts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.14.623621. [PMID: 39605398 PMCID: PMC11601419 DOI: 10.1101/2024.11.14.623621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Despite advancements in cardiovascular engineering, heart diseases remain a leading cause of mortality. The limited understanding of the underlying mechanisms of cardiac dysfunction at the cellular level restricts the development of effective screening and therapeutic methods. To address this, we have developed a framework that incorporates light field detection and individual cell tracking to capture real-time volumetric data in zebrafish hearts, which share structural and electrical similarities with the human heart and generate 120 to 180 beats per minute. Our results indicate that the in-house system achieves an acquisition speed of 200 volumes per second, with resolutions of up to 5.02 ± 0.54 µm laterally and 9.02 ± 1.11 µm axially across the entire depth, using the estimated-maximized-smoothed deconvolution method. The subsequent deep learning-based cell trackers enable further investigation of contractile dynamics, including cellular displacement and velocity, followed by volumetric tracking of specific cells of interest from end-systole to end-diastole in an interactive environment. Collectively, our strategy facilitates real-time volumetric imaging and assessment of contractile dynamics across the entire ventricle at the cellular resolution over multiple cycles, providing significant potential for exploring intercellular interactions in both health and disease.
Collapse
|
11
|
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
|
12
|
Zhang X, Saberigarakani A, Almasian M, Hassan S, Nekkanti M, Ding Y. 4D Light-sheet Imaging of Zebrafish Cardiac Contraction. J Vis Exp 2024:10.3791/66263. [PMID: 38251787 PMCID: PMC10939705 DOI: 10.3791/66263] [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] [Indexed: 01/23/2024] Open
Abstract
Zebrafish is an intriguing model organism known for its remarkable cardiac regeneration capacity. Studying the contracting heart in vivo is essential for gaining insights into structural and functional changes in response to injuries. However, obtaining high-resolution and high-speed 4-dimensional (4D, 3D spatial + 1D temporal) images of the zebrafish heart to assess cardiac architecture and contractility remains challenging. In this context, an in-house light-sheet microscope (LSM) and customized computational analysis are used to overcome these technical limitations. This strategy, involving LSM system construction, retrospective synchronization, single cell tracking, and user-directed analysis, enables one to investigate the micro-structure and contractile function across the entire heart at the single-cell resolution in the transgenic Tg(myl7:nucGFP) zebrafish larvae. Additionally, we are able to further incorporate microinjection of small molecule compounds to induce cardiac injury in a precise and controlled manner. Overall, this framework allows one to track physiological and pathophysiological changes, as well as the regional mechanics at the single-cell level during cardiac morphogenesis and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, The University of Texas at Dallas
| | | | - Milad Almasian
- Department of Bioengineering, The University of Texas at Dallas
| | - Sohail Hassan
- Department of Bioengineering, The University of Texas at Dallas
| | - Manasa Nekkanti
- Department of Bioengineering, The University of Texas at Dallas
| | - Yichen Ding
- Department of Bioengineering, The University of Texas at Dallas; Center for Imaging and Surgical Innovation, The University of Texas at Dallas; Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center;
| |
Collapse
|
13
|
Nagorska A, Zaucker A, Lambert F, Inman A, Toral-Perez S, Gorodkin J, Wan Y, Smutny M, Sampath K. Translational control of furina by an RNA regulon is important for left-right patterning, heart morphogenesis and cardiac valve function. Development 2023; 150:dev201657. [PMID: 38032088 PMCID: PMC10730018 DOI: 10.1242/dev.201657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023]
Abstract
Heart development is a complex process that requires asymmetric positioning of the heart, cardiac growth and valve morphogenesis. The mechanisms controlling heart morphogenesis and valve formation are not fully understood. The pro-convertase FurinA functions in heart development across vertebrates. How FurinA activity is regulated during heart development is unknown. Through computational analysis of the zebrafish transcriptome, we identified an RNA motif in a variant FurinA transcript harbouring a long 3' untranslated region (3'UTR). The alternative 3'UTR furina isoform is expressed prior to organ positioning. Somatic deletions in the furina 3'UTR lead to embryonic left-right patterning defects. Reporter localisation and RNA-binding assays show that the furina 3'UTR forms complexes with the conserved RNA-binding translational repressor, Ybx1. Conditional ybx1 mutant embryos show premature and increased Furin reporter expression, abnormal cardiac morphogenesis and looping defects. Mutant ybx1 hearts have an expanded atrioventricular canal, abnormal sino-atrial valves and retrograde blood flow from the ventricle to the atrium. This is similar to observations in humans with heart valve regurgitation. Thus, the furina 3'UTR element/Ybx1 regulon is important for translational repression of FurinA and regulation of heart development.
Collapse
Affiliation(s)
- Agnieszka Nagorska
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Andreas Zaucker
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Finnlay Lambert
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore 138672
| | - Angus Inman
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Sara Toral-Perez
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Jan Gorodkin
- Center for non-coding RNAs in Technology and Health, Department of Veterinary and Animal Sciences, Faculty for Health and Medical Sciences, University of Copenhagen, Grønnega °rdsvej 3, 1870 Frederiksberg C, Denmark
| | - Yue Wan
- Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore 138672
| | - Michael Smutny
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Mechanochemical Cell Biology, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Karuna Sampath
- Warwick Medical School, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Mechanochemical Cell Biology, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
- Centre for Early Life, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK
| |
Collapse
|
14
|
Zhang X, Almasian M, Hassan SS, Jotheesh R, Kadam VA, Polk AR, Saberigarakani A, Rahat A, Yuan J, Lee J, Carroll K, Ding Y. 4D Light-sheet imaging and interactive analysis of cardiac contractility in zebrafish larvae. APL Bioeng 2023; 7:026112. [PMID: 37351330 PMCID: PMC10283270 DOI: 10.1063/5.0153214] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023] Open
Abstract
Despite ongoing efforts in cardiovascular research, the acquisition of high-resolution and high-speed images for the purpose of assessing cardiac contraction remains challenging. Light-sheet fluorescence microscopy (LSFM) offers superior spatiotemporal resolution and minimal photodamage, providing an indispensable opportunity for the in vivo study of cardiac micro-structure and contractile function in zebrafish larvae. To track the myocardial architecture and contractility, we have developed an imaging strategy ranging from LSFM system construction, retrospective synchronization, single cell tracking, to user-directed virtual reality (VR) analysis. Our system enables the four-dimensional (4D) investigation of individual cardiomyocytes across the entire atrium and ventricle during multiple cardiac cycles in a zebrafish larva at the cellular resolution. To enhance the throughput of our model reconstruction and assessment, we have developed a parallel computing-assisted algorithm for 4D synchronization, resulting in a nearly tenfold enhancement of reconstruction efficiency. The machine learning-based nuclei segmentation and VR-based interaction further allow us to quantify cellular dynamics in the myocardium from end-systole to end-diastole. Collectively, our strategy facilitates noninvasive cardiac imaging and user-directed data interpretation with improved efficiency and accuracy, holding great promise to characterize functional changes and regional mechanics at the single cell level during cardiac development and regeneration.
Collapse
Affiliation(s)
- Xinyuan Zhang
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Milad Almasian
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Sohail S. Hassan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Rosemary Jotheesh
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Vinay A. Kadam
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Austin R. Polk
- Department of Computer Science, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Alireza Saberigarakani
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Aayan Rahat
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Jie Yuan
- Department of Bioengineering, Erik Jonsson School of Engineering and Computer Science, The University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Juhyun Lee
- Department of Bioengineering, The University of Texas at Arlington, Arlington, Texas 76019, USA
| | - Kelli Carroll
- Department of Biology, Austin College, Sherman, Texas 75090, USA
| | - Yichen Ding
- Author to whom correspondence should be addressed:. Tel.: 972–883-7217
| |
Collapse
|
15
|
Baillie JS, Gendernalik A, Garrity DM, Bark D, Quinn TA. The in vivo study of cardiac mechano-electric and mechano-mechanical coupling during heart development in zebrafish. Front Physiol 2023; 14:1086050. [PMID: 37007999 PMCID: PMC10060984 DOI: 10.3389/fphys.2023.1086050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
In the adult heart, acute adaptation of electrical and mechanical activity to changes in mechanical load occurs via feedback processes known as “mechano-electric coupling” and “mechano-mechanical coupling.” Whether this occurs during cardiac development is ill-defined, as acutely altering the heart’s mechanical load while measuring functional responses in traditional experimental models is difficult, as embryogenesis occurs in utero, making the heart inaccessible. These limitations can be overcome with zebrafish, as larvae develop in a dish and are nearly transparent, allowing for in vivo manipulation and measurement of cardiac structure and function. Here we present a novel approach for the in vivo study of mechano-electric and mechano-mechanical coupling in the developing zebrafish heart. This innovative methodology involves acute in vivo atrial dilation (i.e., increased atrial preload) in larval zebrafish by injection of a controlled volume into the venous circulation immediately upstream of the heart, combined with optical measurement of the acute electrical (change in heart rate) and mechanical (change in stroke area) response. In proof-of-concept experiments, we applied our new method to 48 h post-fertilisation zebrafish, which revealed differences between the electrical and mechanical response to atrial dilation. In response to an acute increase in atrial preload there is a large increase in atrial stroke area but no change in heart rate, demonstrating that in contrast to the fully developed heart, during early cardiac development mechano-mechanical coupling alone drives the adaptive increase in atrial output. Overall, in this methodological paper we present our new experimental approach for the study of mechano-electric and mechano-mechanical coupling during cardiac development and demonstrate its potential for understanding the essential adaptation of heart function to acute changes in mechanical load.
Collapse
Affiliation(s)
| | - Alex Gendernalik
- Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | | | - David Bark
- Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, United States
| | - T. Alexander Quinn
- Physiology & Biophysics, Dalhousie University, Halifax, NS, Canada
- Biomedical Engineering, Dalhousie University, Halifax, NS, Canada
- *Correspondence: T. Alexander Quinn,
| |
Collapse
|
16
|
Rajesh V, Kokilavani A, Jayaseelan S, Gomathi S, Vishali K, Kumudhavalli MV. Embryonic exposure to acetyl-L-carnitine protects against valproic acid-induced cardiac malformation in zebrafish model. Amino Acids 2023:10.1007/s00726-023-03256-7. [PMID: 36894749 DOI: 10.1007/s00726-023-03256-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023]
Abstract
Worldwide, estimated counts of about 7.9 million children are born with serious birth defects. In addition to genetic factors, prenatal exposure to drugs and environmental toxicants represents a major contributing factor to congenital malformations. In earlier investigation, we explored cardiac malformation caused by valproic acid (VPA) during early developing stages of zebrafish. Since heart depends on mitochondrial fatty acid oxidative metabolism for energy demands in which carnitine shuttle has a major role, the present study aimed to investigate the effect of acetyl-L-carnitine (AC) against VPA-induced cardiac malformation in developing zebrafish. Initially, AC was subjected to toxicological evaluation, and two micromolar concentrations (25 µM and 50 µM) were selected for evaluation. A sub-lethal concentration of VPA (50 µM) was selected to induce cardiac malformation. The embryos were grouped and the drug exposures were made at 2.5 h post-fertilization (hpf). The cardiac development and functioning was monitored. A progressive decline in cardiac functioning was noted in group exposed to VPA 50 µM. At 96 hpf and 120 hpf, the morphology of heart was severely affected with the chambers which became elongated and string-like accompanied by histological changes. Acridine orange staining showed accumulation of apoptotic cells. Group exposed to VPA 50 µM with AC 50 µM showed a significant reduction in pericardial sac edema with morphological, functional and histological recovery in developing heart. Moreover, reduced number of apoptotic cells was noted. The improvement with AC might be due to restoration of carnitine homeostasis for cardiac energy metabolism in developing heart.
Collapse
Affiliation(s)
- Venugopalan Rajesh
- Department of Pharmacology, The Erode College of Pharmacy and Research Institute, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India.
| | - Annadurai Kokilavani
- Department of Pharmacology, The Erode College of Pharmacy and Research Institute, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Subramanian Jayaseelan
- Department of Pharmaceutical Analysis, The Erode College of Pharmacy and Research Institute, Veppampalayam, Vallipurathampalayam (Po), Erode, Tamil Nadu, 638112, India
| | - Swaminathan Gomathi
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Ottacamund, Nilgiris District, Ooty, Tamil Nadu, 643001, India
| | - Korrapati Vishali
- Department of Pharmacology, Vignan's Foundation for Science, Technology and Research, Vadlamudi, Guntur, Andhra Pradesh, 522213, India
| | - Manni Venkatachari Kumudhavalli
- Department of Pharmaceutical Chemistry, Vinayaka Mission's College of Pharmacy, Kondappanaickenpatti, Yercaud Main Road, Salem, Tamil Nadu, 636008, India
| |
Collapse
|
17
|
Yahya I, Brand-Saberi B, Morosan-Puopolo G. Chicken embryo as a model in second heart field development. Heliyon 2023; 9:e14230. [PMID: 36923876 PMCID: PMC10009738 DOI: 10.1016/j.heliyon.2023.e14230] [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: 10/12/2022] [Revised: 01/30/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Previously, a single source of progenitor cells was thought to be responsible for the formation of the cardiac muscle. However, the second heart field has recently been identified as an additional source of myocardial progenitor cells. The chicken embryo, which develops in the egg, outside the mother can easily be manipulated in vivo and in vitro. Hence, it was an excellent model for establishing the concept of the second heart field. Here, our review will focus on the chicken model, specifically its role in understanding the second heart field. In addition to discussing historical aspects, we provide an overview of recent findings that have helped to define the chicken second heart field progenitor cells. A better understanding of the second heart field development will provide important insights into the congenital malformations affecting cardiac muscle formation and function.
Collapse
Affiliation(s)
- Imadeldin Yahya
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
- Department of Anatomy, Faculty of Veterinary Medicine, University of Khartoum, Khartoum, 11115, Sudan
- Corresponding author. Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801 Bochum, Germany.
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, 44801, Bochum, Germany
| | | |
Collapse
|
18
|
Brown W, Wesalo J, Tsang M, Deiters A. Engineering Small Molecule Switches of Protein Function in Zebrafish Embryos. J Am Chem Soc 2023; 145:2395-2403. [PMID: 36662675 DOI: 10.1021/jacs.2c11366] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Precise temporally regulated protein function directs the highly complex processes that make up embryo development. The zebrafish embryo is an excellent model organism to study development, and conditional control over enzymatic activity is desirable to target chemical intervention to specific developmental events and to investigate biological mechanisms. Surprisingly few, generally applicable small molecule switches of protein function exist in zebrafish. Genetic code expansion allows for site-specific incorporation of unnatural amino acids into proteins that contain caging groups that are removed through addition of small molecule triggers such as phosphines or tetrazines. This broadly applicable control of protein function was applied to activate several enzymes, including a GTPase and a protease, with temporal precision in zebrafish embryos. Simple addition of the small molecule to the media produces robust and tunable protein activation, which was used to gain insight into the development of a congenital heart defect from a RASopathy mutant of NRAS and to control DNA and protein cleavage events catalyzed by a viral recombinase and a viral protease, respectively.
Collapse
Affiliation(s)
- Wes Brown
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Joshua Wesalo
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
19
|
Sutlive J, Seyyedhosseinzadeh H, Ao Z, Xiu H, Choudhury S, Gou K, Guo F, Chen Z. Mechanics of morphogenesis in neural development: In vivo, in vitro, and in silico. BRAIN MULTIPHYSICS 2023. [DOI: 10.1016/j.brain.2022.100062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
20
|
Li W, Guo S, Miao N. Transcriptional responses of fluxapyroxad-induced dysfunctional heart in zebrafish (Danio rerio) embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:90034-90045. [PMID: 35864390 DOI: 10.1007/s11356-022-21981-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 07/08/2022] [Indexed: 06/15/2023]
Abstract
Fluxapyroxad (FLU) is a succinate dehydrogenase inhibitor (SDHI) fungicide used in controlling crop diseases. Potential toxicity to aquatic organisms is not known. We exposed zebrafish to 1, 2, and 4 μM FLU for 3 days. The embryonic zebrafish showed developmental cardiac defects, including heart malformation, pericardial edema, and heart rate reduction. Compared with the controls, cardiac-specific transcription factors (nkx2.5, myh7, myl7, and myh6) exhibited dysregulated expression patterns after FLU treatment. We next used transcriptome and qRT-PCR analyses to explore the molecular mechanism of FLU cardiotoxicity. The transcriptome analysis and interaction network showed that the downregulated genes were enriched in calcium signaling pathways, adrenergic signaling in cardiomyocytes, and cardiac muscle contraction. FLU exposure repressed the cardio-related calcium signaling pathway, associated with apoptosis in the heart and other manifestations of cardiotoxicity. Thus, the findings provide valuable evidence that FLU exposure causes disruption of cardiac development in zebrafish embryos. Our findings will help to promote a better understanding of the toxicity mechanisms of FLU and act as a reference to explore the rational use and safety of FLU in agriculture.
Collapse
Affiliation(s)
- Wenhua Li
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Shanshan Guo
- Engineering Research Center of Molecular Medicine of Ministry of Education, Key Laboratory of Fujian Molecular Medicine, Key Laboratory of Xiamen Marine and Gene Drugs, Key Laboratory of Precision Medicine and Molecular Diagnosis of Fujian Universities, School of Biomedical Sciences, Huaqiao University, Xiamen, 361021, People's Republic of China
| | - Nan Miao
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, 668 Jimei Road, Xiamen, 361021, People's Republic of China.
| |
Collapse
|
21
|
Yang X, Li L, Shi Y, Wang X, Zhang Y, Jin M, Chen X, Wang R, Liu K. Neurotoxicity of sanguinarine via inhibiting mitophagy and activating apoptosis in zebrafish and PC12 cells. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2022; 188:105259. [PMID: 36464364 DOI: 10.1016/j.pestbp.2022.105259] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 06/17/2023]
Abstract
Sanguinarine, a plant-derived phytoalexin, displays various biological activities, such as insecticidal, antimicrobial, anti-inflammatory, anti-angiogenesis and antitumor effects. But its potential neurotoxicity and the underlying mechanisms has rarely been investigated. Therefore, we aimed to assess the neurotoxicity of sanguinarine using zebrafish model and PC12 cells in this study. The results showed that sanguinarine induced the reduction of the length of dopamine neurons and inhibited the blood vessel in the head area of the zebrafish. Further studies demonstrated that the behavioral phenotype of the larval zebrafish was changed by sanguinarine. In addition, there were more apoptotic cells in the larval zebrafish head area. The mRNA expression levels of β-syn, th, pink1 and parkin, closely related to the nervous function, were changed after sanguinarine treatment. The in vitro studies show that notably increases of ROS and apoptosis levels in PC12 cells were observed after sanguinarine treatment. Moreover, the protein expression of Caspase3, Parp, Bax, Bcl2, α-Syn, Th, PINK1 and Parkin were also altered by sanguinarine. Our data indicated that the inhibition of mitophagy, ROS elevation and apoptosis were involved in the neurotoxicity of sanguinarine. These findings will be useful to understand the toxicity induced by sanguinarine.
Collapse
Affiliation(s)
- Xueliang Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Lei Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yuxin Shi
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xue Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xiqiang Chen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
22
|
Wu YJ, Li ZH, Li JY, Zhou Y, Wang RY, Chen XY, Qing LS, Luo P. Elucidation of the binding mechanism of astragaloside IV derivative with human serum albumin and its cardiotoxicity in zebrafish embryos. Front Pharmacol 2022; 13:987882. [PMID: 36210826 PMCID: PMC9537572 DOI: 10.3389/fphar.2022.987882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
LS-102 is a new derivative of astragaloside IV (AGS IV) that has been shown to possess potentially significant cardioprotective effects. However, there are no reports concerning its interaction with human serum albumin (HSA) and toxicology in vertebrates. The present investigation was undertaken to characterize the interaction of AGS IV and LS-102 with HSA using equilibrium dialysis and UHPLC-MS/MS methods, along with computational methods. Notably, the effects of AGS IV and LS-102 were studied in vivo using the zebrafish embryo model. Markers related to embryonic cardiotoxicity and thrombosis were evaluated. We showed that the plasma protein binding rate of AGS IV (94.04%–97.42%) was significantly higher than that of LS-102 (66.90%–69.35%). Through site marker competitive experiments and molecular docking, we found that AGS IV and LS-102 were located at the interface of subdomains IIA and IIIA, but the site I might be the primary binding site. Molecular dynamics revealed that AGS IV showed a higher binding free energy mainly due to the stronger hydrophobic and hydrogen bonding interactions. Moreover, the secondary structure implied no obvious effect on the protein structure and conformation during the binding of LS-102. LS-102 significantly ameliorated the astramizole-induced heart rate slowing, increased SV-BA spacing, and prevented arachidonic acid-induced thrombosis in zebrafish. To our knowledge, we are the first to reveal that LS-102 binds to HSA with reversible and moderate affinity, indicating its easy diffusion from the circulatory system to the target tissue, thereby providing significant insights into its pharmacokinetic and pharmacodynamic properties when spread in the human body. Our results also provide a reference for the rational clinical application of LS-102 in the cardiovascular field.
Collapse
Affiliation(s)
- You-Jiao Wu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Zhan-Hua Li
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Jiu-Yan Li
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Yan Zhou
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Run-Yue Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Xiao-Yi Chen
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
| | - Lin-Sen Qing
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
- *Correspondence: Lin-Sen Qing, ; Pei Luo,
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macao, SAR, China
- *Correspondence: Lin-Sen Qing, ; Pei Luo,
| |
Collapse
|
23
|
Cardiotoxicity of Zebrafish Induced by 6-Benzylaminopurine Exposure and Its Mechanism. Int J Mol Sci 2022; 23:ijms23158438. [PMID: 35955574 PMCID: PMC9369308 DOI: 10.3390/ijms23158438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/15/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
6-BA is a common plant growth regulator, but its safety has not been conclusive. The heart is one of the most important organs of living organisms, and the cardiogenesis process of zebrafish is similar to that of humans. Therefore, based on wild-type and transgenic zebrafish, we explored the development of zebrafish heart under 6-BA exposure and its mechanism. We found that 6-BA affected larval cardiogenesis, inducing defective expression of key genes for cardiac development (myl7, vmhc, and myh6) and AVC differentiation (bmp4, tbx2b, and notch1b), ultimately leading to weakened cardiac function (heart rate, diastolic speed, systolic speed). Acridine orange staining showed that the degree of apoptosis in zebrafish hearts was significantly increased under 6-BA, and the expression of cell-cycle-related genes was also changed. In addition, HPA axis assays revealed abnormally expressed mRNA levels of genes and significantly increased cortisol contents, which was also consistent with the observed anxiety behavior in zebrafish at 3 dpf. Transcriptional abnormalities of pro- and anti-inflammatory factors in immune signaling pathways were also detected in qPCR experiments. Collectively, we found that 6-BA induced cardiotoxicity in zebrafish, which may be related to altered HPA axis activity and the onset of inflammatory responses under 6-BA treatment.
Collapse
|
24
|
Wang X, Ge X, Qin Y, Liu D, Chen C. Ifi30 Is Required for Sprouting Angiogenesis During Caudal Vein Plexus Formation in Zebrafish. Front Physiol 2022; 13:919579. [PMID: 35910561 PMCID: PMC9325957 DOI: 10.3389/fphys.2022.919579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Interferon-gamma-inducible protein 30 (IFI30) is a critical enzyme that mainly exists in immune cells and functions in reducing protein disulfide bonds in endocytosis-mediated protein degradation. Regardless of this, it is also found to be expressed in vascular system. However, the functions of IFI30 in vascular development remains unknown. Vascular network formation is a tightly controlled process coordinating a series of cell behaviors, including endothelial cell (EC) sprouting, proliferation, and anastomosis. In this work, we analyzed the function of zebrafish Ifi30, orthologous to the human IFI30, in vascular development during embryogenesis. The ifi30 gene was found to be highly expressed in the caudal vein plexus (CVP) region of zebrafish embryos. Morpholino-mediated Ifi30 knockdown in zebrafish resulted in incomplete CVP formation with reduced loop numbers, area, and width. Further analyses implied that Ifi30 deficiency impaired cell behaviors of both ECs and macrophages, including cell proliferation and migration. Here, we demonstrate a novel role of IFI30, which was originally identified as a lysosomal thiol reductase involved in immune responses, in CVP development during embryogenesis. Our results suggest that Ifi30 is required for sprouting angiogenesis during CVP formation, which may offer an insight into the function of human IFI30 in angiogenesis under physiological or pathological conditions.
Collapse
Affiliation(s)
| | | | | | - Dong Liu
- *Correspondence: Dong Liu, ; Changsheng Chen,
| | | |
Collapse
|
25
|
Chen C, Liu D. Establishment of Zebrafish Models for Diabetes Mellitus and Its Microvascular Complications. J Vasc Res 2022; 59:251-260. [PMID: 35378543 DOI: 10.1159/000522471] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/19/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease known to cause several microvascular complications, including diabetic retinopathy, diabetic nephropathy, and diabetic neuropathy. Hyperglycemia plays a key role in inducing diabetic microvascular complications. A cohort of diabetic animal models has been established to study diabetes-related vascular diseases. However, the zebrafish model offers unique advantages in this field. The tiny size and huge offspring numbers of zebrafish make it amenable to perform large-scale analysis or screening. The easily accessible strategies for gene manipulation with morpholino or CRISPR/Cas9 and chemical/drug treatment through microinjection or skin absorption allow establishing the zebrafish DM models by a variety of means. In addition, the transparency of zebrafish embryos makes it accessible to perform in vivo high-resolution imaging of the vascular system. In this review, we focus on the strategies to establish diabetic or hyperglycemic models with zebrafish and the achievements and disadvantages of using zebrafish as a model to study diabetic microvascular complications.
Collapse
Affiliation(s)
- Changsheng Chen
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Medical College, Nantong University, Nantong, China
| | - Dong Liu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Medical College, Nantong University, Nantong, China.,Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, China
| |
Collapse
|
26
|
Baek KI, Chang SS, Chang CC, Roustaei M, Ding Y, Wang Y, Chen J, O'Donnell R, Chen H, Ashby JW, Xu X, Mack JJ, Cavallero S, Roper M, Hsiai TK. Vascular Injury in the Zebrafish Tail Modulates Blood Flow and Peak Wall Shear Stress to Restore Embryonic Circular Network. Front Cardiovasc Med 2022; 9:841101. [PMID: 35369301 PMCID: PMC8971683 DOI: 10.3389/fcvm.2022.841101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Mechano-responsive signaling pathways enable blood vessels within a connected network to structurally adapt to partition of blood flow between organ systems. Wall shear stress (WSS) modulates endothelial cell proliferation and arteriovenous specification. Here, we study vascular regeneration in a zebrafish model by using tail amputation to disrupt the embryonic circulatory loop (ECL) at 3 days post fertilization (dpf). We observed a local increase in blood flow and peak WSS in the Segmental Artery (SeA) immediately adjacent to the amputation site. By manipulating blood flow and WSS via changes in blood viscosity and myocardial contractility, we show that the angiogenic Notch-ephrinb2 cascade is hemodynamically activated in the SeA to guide arteriogenesis and network reconnection. Taken together, ECL amputation induces changes in microvascular topology to partition blood flow and increase WSS-mediated Notch-ephrinb2 pathway, promoting new vascular arterial loop formation and restoring microcirculation.
Collapse
Affiliation(s)
- Kyung In Baek
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shyr-Shea Chang
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, United States
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, United States
| | - Chih-Chiang Chang
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mehrdad Roustaei
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yichen Ding
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yixuan Wang
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Justin Chen
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ryan O'Donnell
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Julianne W. Ashby
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xiaolei Xu
- Zebrafish Genetics, Mayo Clinic, Rochester, MN, United States
| | - Julia J. Mack
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Susana Cavallero
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Marcus Roper
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tzung K. Hsiai
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
27
|
Zebrafish Model-Based Assessment of Indoxyl Sulfate-Induced Oxidative Stress and Its Impact on Renal and Cardiac Development. Antioxidants (Basel) 2022; 11:antiox11020400. [PMID: 35204282 PMCID: PMC8869691 DOI: 10.3390/antiox11020400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
Kidney disease patients may have concurrent chronic kidney disease-associated mineral bone disorder and hypertension. Cardiovascular disease (CVD) and neuropathy occur due to kidney failure-induced accumulation of uremic toxins in the body. Indoxyl sulfate (IS), a product of indole metabolism in the liver, is produced from tryptophan by the intestinal flora and is ultimately excreted through the kidneys. Hemodialysis helps renal failure patients eliminate many nephrotoxins, except for IS, which leads to a poor prognosis. Although the impacts of IS on cardiac and renal development have been well documented using mouse and rat models, other model organisms, such as zebrafish, have rarely been studied. The zebrafish genome shares at least 70% similarity with the human genome; therefore, zebrafish are ideal model organisms for studying vertebrate development, including renal development. In this study, we aimed to investigate the impact of IS on the development of zebrafish embryos, especially cardiac and renal development. At 24 h postfertilization (hpf), zebrafish were exposed to IS at concentrations ranging from 2.5 to 10 mM. IS reduced survival and the hatching rate, caused cardiac edema, increased mortality, and shortened the body length of zebrafish embryos. In addition, IS decreased heart rates and renal function. IS affected zebrafish development via the ROS and MAPK pathways, which subsequently led to inflammation in the embryos. The results suggest that IS interferes with cardiac and renal development in zebrafish embryos, providing new evidence about the toxicity of IS to aquatic organisms and new insights for the assessment of human health risks. Accordingly, we suggest that zebrafish studies can ideally complement mouse model studies to allow the simultaneous and comprehensive investigation of the physiological impacts of uremic endotheliotoxins, such as IS, on cardiac and renal development.
Collapse
|
28
|
Lv F, Ge X, Qian P, Lu X, Liu D, Chen C. Neuron navigator 3 (NAV3) is required for heart development in zebrafish. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:173-183. [PMID: 35039994 DOI: 10.1007/s10695-022-01049-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
As a tightly controlled biological process, cardiogenesis requires the specification and migration of a suite of cell types to form a particular three-dimensional configuration of the heart. Many genetic factors are involved in the formation and maturation of the heart, and any genetic mutations may result in severe cardiac failures. The neuron navigator (NAV) family consists of three vertebrate homologs (NAV1, NAV2, and NAV3) of the neural guidance molecule uncoordinated-53 (UNC-53) in Caenorhabditis elegans. Although they are recognized as neural regulators, their expressions are also detected in many organs, including the heart, kidney, and liver. However, the functions of NAVs, regardless of neural guidance, remain largely unexplored. In our study, we found that nav3 gene was expressed in the cardiac region of zebrafish embryos from 24 to 48 h post-fertilization (hpf) by means of in situ hybridization (ISH) assay. A CRISPR/Cas9-based genome editing method was utilized to delete the nav3 gene in zebrafish and loss of function of Nav3 resulted in a severe deficiency in its cardiac morphology and structure. The similar phenotypic defects of the knockout mutants could recur by nav3 morpholino injection and be rescued by nav3 mRNA injection. Dual-color fluorescence imaging of ventricle and atrium markers further confirmed the disruption of the heart development in nav3-deleted mutants. Although the heart rate was not affected by the deletion of nav3, the heartbeat intensity was decreased in the mutants. All these findings indicate that Nav3 was required for cardiogenesis in developing zebrafish embryos.
Collapse
Affiliation(s)
- Feng Lv
- Nantong Science and Technology College, School of Life Sciences, Nantong University, Nantong, China
| | - Xiaojuan Ge
- Nantong Science and Technology College, School of Life Sciences, Nantong University, Nantong, China
| | - Peipei Qian
- Medical School, Nantong University, Nantong, China
| | - Xiaofeng Lu
- Nantong Science and Technology College, School of Life Sciences, Nantong University, Nantong, China
| | - Dong Liu
- Nantong Science and Technology College, School of Life Sciences, Nantong University, Nantong, China.
| | - Changsheng Chen
- Nantong Science and Technology College, School of Life Sciences, Nantong University, Nantong, China.
| |
Collapse
|
29
|
Wang X, Yang X, Wang J, Li L, Zhang Y, Jin M, Chen X, Sun C, Wang R, Liu K. Cardiotoxicity of sanguinarine via regulating apoptosis and MAPK pathways in zebrafish and HL1 cardiomyocytes. Comp Biochem Physiol C Toxicol Pharmacol 2022; 252:109228. [PMID: 34744004 DOI: 10.1016/j.cbpc.2021.109228] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/14/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022]
Abstract
Sanguinarine, a plant phytoalexin, possesses extensive biological activities including antimicrobial, insecticidal, antitumor, anti-inflammatory and anti-angiogenesis effect. But its cardiotoxicity has rarely been studied. Here, we assess the cardiotoxicity of sanguinarine in vivo using larval zebrafish from 48 hpf to 96 hpf. The results show that sanguinarine caused severe malformation and the dysfunction of the heart including reductions of heart rate, red blood cell number, blood flow dynamics, stroke volume and increase of SV-BA distance, subintestinal venous congestion. Further studies showed that apoptosis in the zebrafish heart region was observed after sanguinarine exposure using TUNEL assay and AO staining method. In addition, the genes, such as sox9b, myl7, nkx2.5 and bmp10, which play crucial parts in the development and the function of the heart, were changed after sanguinarine treatment. caspase3, caspase9, bax and bcl2, apoptosis-related genes, were also altered by sanguinarine. Further studies were performed to study the cardiotoxicity in vitro using cardiomyocytes HL1 cell line. The results showed that remarkable increase of apoptosis and ROS level in HL1 cells were induced by sanguinarine. Moreover, the MAPK pathway (JNK and P38) were notably enhanced and involved in the cardiotoxicity induced by sanguinarine. Our findings will provide better understanding of sanguinarine in the toxic effect on heart.
Collapse
Affiliation(s)
- Xue Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xueliang Yang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Jiazhen Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Lei Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xiqiang Chen
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Chen Sun
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Rongchun Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Shandong Provincial Engineering Laboratory for Biological Testing Technology, 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
30
|
Henderson DJ, Eley L, Turner JE, Chaudhry B. Development of the Human Arterial Valves: Understanding Bicuspid Aortic Valve. Front Cardiovasc Med 2022; 8:802930. [PMID: 35155611 PMCID: PMC8829322 DOI: 10.3389/fcvm.2021.802930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022] Open
Abstract
Abnormalities in the arterial valves are some of the commonest congenital malformations, with bicuspid aortic valve (BAV) occurring in as many as 2% of the population. Despite this, most of what we understand about the development of the arterial (semilunar; aortic and pulmonary) valves is extrapolated from investigations of the atrioventricular valves in animal models, with surprisingly little specifically known about how the arterial valves develop in mouse, and even less in human. In this review, we summarise what is known about the development of the human arterial valve leaflets, comparing this to the mouse where appropriate.
Collapse
Affiliation(s)
- Deborah J. Henderson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | |
Collapse
|
31
|
The effective use of blebbistatin to study the action potential of cardiac pacemaker cells of zebrafish (Danio rerio) during incremental warming. Curr Res Physiol 2022; 5:48-54. [PMID: 35128467 PMCID: PMC8803472 DOI: 10.1016/j.crphys.2022.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Blebbistatin potently inhibits actin-myosin interaction, preventing contractile activity of excitable cells including cardiac myocytes, despite electrical excitation of an action potential (AP). We collected intracellular microelectrode recordings of pacemaker cells located in the sinoatrial region (SAR) of the zebrafish heart at room temperature and during acute warming to investigate whether or not blebbistatin inhibition of contraction significantly alters pacemaker cell electrophysiology. Changes were evaluated based on 16 variables that characterized the AP waveform. None of these AP variables nor the spontaneous heart rate were significantly modified with the application of 10 μM blebbistatin when recordings were made at room temperature. Compared with the control group, the blebbistatin-treated group showed minor changes in the rate of spontaneous diastolic depolarization (P = 0.027) and the 50% and 80% repolarization (P = 0.008 and 0.010, respectively) in the 26°C–29°C temperature bin, but not at higher temperatures. These findings suggest that blebbistatin is an effective excitation-contraction uncoupler that does not appreciably affect APs generated in pacemaking cells of the SAR and can, therefore, be used in zebrafish cardiac studies. Blebbistatin uncouples excitation-contraction in zebrafish cardiomyocytes. Blebbistatin does not modify the pacemaker action potential variables. Temperature does not modify the effect of blebbistatin. First validation of the use of blebbistatin in adult fish. Methodology of intracellular microelectrode recording of zebrafish pacemaker cells.
Collapse
|
32
|
Stoyek MR, Hortells L, Quinn TA. From Mice to Mainframes: Experimental Models for Investigation of the Intracardiac Nervous System. J Cardiovasc Dev Dis 2021; 8:149. [PMID: 34821702 PMCID: PMC8620975 DOI: 10.3390/jcdd8110149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/17/2023] Open
Abstract
The intracardiac nervous system (IcNS), sometimes referred to as the "little brain" of the heart, is involved in modulating many aspects of cardiac physiology. In recent years our fundamental understanding of autonomic control of the heart has drastically improved, and the IcNS is increasingly being viewed as a therapeutic target in cardiovascular disease. However, investigations of the physiology and specific roles of intracardiac neurons within the neural circuitry mediating cardiac control has been hampered by an incomplete knowledge of the anatomical organisation of the IcNS. A more thorough understanding of the IcNS is hoped to promote the development of new, highly targeted therapies to modulate IcNS activity in cardiovascular disease. In this paper, we first provide an overview of IcNS anatomy and function derived from experiments in mammals. We then provide descriptions of alternate experimental models for investigation of the IcNS, focusing on a non-mammalian model (zebrafish), neuron-cardiomyocyte co-cultures, and computational models to demonstrate how the similarity of the relevant processes in each model can help to further our understanding of the IcNS in health and disease.
Collapse
Affiliation(s)
- Matthew R. Stoyek
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
| | - Luis Hortells
- Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg–Bad Krozingen, 79110 Freiburg, Germany;
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - T. Alexander Quinn
- Department of Physiology and Biophysics, Dalhousie University, Halifax, NS 15000, Canada;
- School of Biomedical Engineering, Dalhousie University, Halifax, NS 15000, Canada
| |
Collapse
|
33
|
Duong TB, Waxman JS. Patterning of vertebrate cardiac progenitor fields by retinoic acid signaling. Genesis 2021; 59:e23458. [PMID: 34665508 DOI: 10.1002/dvg.23458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
The influence of retinoic acid (RA) signaling on vertebrate development has a well-studied history. Cumulatively, we now understand that RA signaling has a conserved requirement early in development restricting cardiac progenitors within the anterior lateral plate mesoderm of vertebrate embryos. Moreover, genetic and pharmacological manipulations of RA signaling in vertebrate models have shown that proper heart development is achieved through the deployment of positive and negative feedback mechanisms, which maintain appropriate RA levels. In this brief review, we present a chronological overview of key work that has led to a current model of the critical role for early RA signaling in limiting the generation of cardiac progenitors within vertebrate embryos. Furthermore, we integrate the previous work in mice and our recent findings using zebrafish, which together show that RA signaling has remarkably conserved influences on the later-differentiating progenitor populations at the arterial and venous poles. We discuss how recognizing the significant conservation of RA signaling on the differentiation of these progenitor populations offers new perspectives and may impact future work dedicated to examining vertebrate heart development.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
34
|
Piechowski JM, Bagatto B. Cardiovascular function during early development is suppressed by cinnamon flavored, nicotine-free, electronic cigarette vapor. Birth Defects Res 2021; 113:1215-1223. [PMID: 34487432 DOI: 10.1002/bdr2.1951] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/20/2021] [Accepted: 08/21/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Vaping products continue to remain popular among teens and young adults despite an overall lack of research regarding their potential health effects. While much research focuses on respiratory effects associated with electronic cigarette use, their effects on other systems, including embryonic cardiovascular function and development due to maternal use during pregnancy, also needs to be evaluated. Here, we assessed the impact of nicotine-free, cinnamon and chocolate flavored, electronic cigarette vapor on cardiovascular function during early development by exposing wild-type zebrafish embryos to electronic cigarette vapor. METHODS Vapor was produced from a second-generation style vape pen and was incorporated into dechlorinated water at 0.6, 12, and 25 puffs/L, where one puff equals 55 ml of vapor. Vapor infused water was distributed among flasks to which zebrafish embryos were added. Exposures lasted for 24 hours and cardiovascular videos were recorded. Videos were analyzed and end systolic volume, end diastolic volume, stroke volume, heart rate, cardiac output, red blood cell density, and arterial and venous blood vessel diameters were measured. RESULTS Here, it was found that embryonic exposure to nicotine free, cinnamon, and not chocolate, flavored electronic cigarette vapor at 25 puffs/L significantly decreased all cardiovascular parameters measured, with the exception of blood vessel diameter. No significant effect on any measured parameter was observed at 0.6 or 12 puffs/L with either flavor. CONCLUSION These results indicate that cinnamon flavored electronic cigarette vapor can affect cardiovascular function during early development, even in the absence of nicotine, particularly at elevated exposure concentrations.
Collapse
Affiliation(s)
- Jennifer M Piechowski
- Program in Integrated Bioscience, Department of Biology, The University of Akron, Akron, Ohio, USA
| | - Brian Bagatto
- Program in Integrated Bioscience, Department of Biology, The University of Akron, Akron, Ohio, USA
| |
Collapse
|
35
|
Chakraborty S, Allmon E, Sepúlveda MS, Vlachos PP. Haemodynamic dependence of mechano-genetic evolution of the cardiovascular system in Japanese medaka. J R Soc Interface 2021; 18:20210752. [PMID: 34699728 PMCID: PMC8548083 DOI: 10.1098/rsif.2021.0752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 11/12/2022] Open
Abstract
The progression of cardiac gene expression-wall shear stress (WSS) interplay is critical to identifying developmental defects during cardiovascular morphogenesis. However, mechano-genetics from the embryonic to larval stages are poorly understood in vertebrates. We quantified peak WSS in the heart and tail vessels of Japanese medaka from 3 days post fertilization (dpf) to 14 dpf using in vivo micro-particle image velocimetry flow measurements, and in parallel analysed the expression of five cardiac genes (fgf8, hoxb6b, bmp4, nkx2.5, smyd1). Here, we report that WSS in the atrioventricular canal (AVC), ventricular outflow tract (OFT), and the caudal vessels in medaka peak with inflection points at 6 dpf and 10-11 dpf instead of a monotonic trend. Retrograde flows are captured at the AVC and OFT of the medaka heart for the first time. In addition, all genes were upregulated at 3 dpf and 7 dpf, indicating a possible correlation between the two, with the cardiac gene upregulation preceding WSS increase in order to facilitate cardiac wall remodelling.
Collapse
Affiliation(s)
- Sreyashi Chakraborty
- Department of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Elizabeth Allmon
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Maria S. Sepúlveda
- Department of Forestry and Natural Resources, Purdue University, West Lafayette, IN, USA
| | - Pavlos P. Vlachos
- Department of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
36
|
Advances in Cardiac Development and Regeneration Using Zebrafish as a Model System for High-Throughput Research. J Dev Biol 2021; 9:jdb9040040. [PMID: 34698193 PMCID: PMC8544412 DOI: 10.3390/jdb9040040] [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: 09/01/2021] [Revised: 09/19/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Heart disease is the leading cause of death in the United States and worldwide. Understanding the molecular mechanisms of cardiac development and regeneration will improve diagnostic and therapeutic interventions against heart disease. In this direction, zebrafish is an excellent model because several processes of zebrafish heart development are largely conserved in humans, and zebrafish has several advantages as a model organism. Zebrafish transcriptomic profiles undergo alterations during different stages of cardiac development and regeneration which are revealed by RNA-sequencing. ChIP-sequencing has detected genome-wide occupancy of histone post-translational modifications that epigenetically regulate gene expression and identified a locus with enhancer-like characteristics. ATAC-sequencing has identified active enhancers in cardiac progenitor cells during early developmental stages which overlap with occupancy of histone modifications of active transcription as determined by ChIP-sequencing. CRISPR-mediated editing of the zebrafish genome shows how chromatin modifiers and DNA-binding proteins regulate heart development, in association with crucial signaling pathways. Hence, more studies in this direction are essential to improve human health because they answer fundamental questions on cardiac development and regeneration, their differences, and why zebrafish hearts regenerate upon injury, unlike humans. This review focuses on some of the latest studies using state-of-the-art technology enabled by the elegant yet simple zebrafish.
Collapse
|
37
|
Dong Y, Qian L, Liu J. Molecular and cellular basis of embryonic cardiac chamber maturation. Semin Cell Dev Biol 2021; 118:144-149. [PMID: 33994094 DOI: 10.1016/j.semcdb.2021.04.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/12/2021] [Accepted: 04/26/2021] [Indexed: 10/21/2022]
Abstract
Heart malformation is the leading cause of human birth defects, and many of the congenital heart diseases (CHDs) originate from genetic defects that impact cardiac development and maturation. During development, the vertebrate heart undergoes a series of complex morphogenetic processes that increase its ability to pump blood. One of these processes leads to the formation of the sheet-like muscular projections called trabeculae. Trabeculae increase cardiac output and permit nutrition and oxygen uptake in the embryonic myocardium prior to coronary vascularization without increasing heart size. Cardiac trabeculation is also crucial for the development of the intraventricular fast conduction system. Alterations in cardiac trabecular development can manifest as a variety of congenital defects such as left ventricular noncompaction. In this review, we discuss the latest advances in understanding the molecular and cellular mechanisms underlying cardiac trabecular development.
Collapse
Affiliation(s)
- Yanhan Dong
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
38
|
Zhu J, Liu C, Wang J, Liang Y, Gong X, You L, Ji C, Wang SL, Wang C, Chi X. Difenoconazole induces cardiovascular toxicity through oxidative stress-mediated apoptosis in early life stages of zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 216:112227. [PMID: 33848738 DOI: 10.1016/j.ecoenv.2021.112227] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
Difenoconazole (DIF), a common broad-spectrum triazole fungicide, is associated with an increased risk of cardiovascular diseases. Unfortunately, little attention has been paid to the mechanisms underlying this association. In this study, zebrafish embryos were exposed to DIF (0, 0.3, 0.6 and 1.2 mg/L) from 4 to 96 h post fertilization (hpf) and cardiovascular toxicity was evaluated. Our results showed that DIF decreased hatching rate, survival rate and heart rate, with increased malformation rate. Cardiovascular deformities are the most prominent, including pericardial edema, abnormal cardiac structure and disrupted vascular pattern in two transgenic zebrafish models (myl7:egfp and fli1:egfp). DIF exacerbated oxidative stress by via accumulation of reactive oxygen species (ROS) and inhibition of antioxidant enzyme. Cardiovascular apoptosis was triggered through increased expression of p53, bcl-2, bax and caspase 9, while DIF suppressed the transcription of key genes involved in calcium signaling and cardiac muscle contraction. These adverse outcomes were restored by the antioxidant N-acetyl-L-cysteine (NAC), indicating that oxidative stress played a crucial role in DIF-induced cardiovascular toxicity caused by apoptosis and inhibition of cardiac muscle contraction. Taken together, this study revealed the key role of oxidative stress in DIF-induced cardiovascular toxicity and provided novel insights into strategies to mitigate its toxicity.
Collapse
Affiliation(s)
- Jiansheng Zhu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Chunlan Liu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Jingyu Wang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, Nanjing 210029, PR China
| | - Yinyin Liang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Xing Gong
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Lianghui You
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, Nanjing 210029, PR China
| | - Chenbo Ji
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, Nanjing 210029, PR China
| | - Shou-Lin Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China
| | - Chao Wang
- Key Lab of Modern Toxicology of Ministry of Education, Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, PR China.
| | - Xia Chi
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, PR China; Institute of Pediatrics, Nanjing Medical University, Nanjing 210029, PR China.
| |
Collapse
|
39
|
Duong TB, Holowiecki A, Waxman JS. Retinoic acid signaling restricts the size of the first heart field within the anterior lateral plate mesoderm. Dev Biol 2021; 473:119-129. [PMID: 33607112 DOI: 10.1016/j.ydbio.2021.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/27/2023]
Abstract
Retinoic acid (RA) signaling is required to restrict heart size through limiting the posterior boundary of the vertebrate cardiac progenitor field within the anterior lateral plate mesoderm (ALPM). However, we still do not fully understand how different cardiac progenitor populations that contribute to the developing heart, including earlier-differentiating first heart field (FHF), later-differentiating second heart field (SHF), and neural crest-derived progenitors, are each affected in RA-deficient embryos. Here, we quantified the number of cardiac progenitors and differentiating cardiomyocytes (CMs) in RA-deficient zebrafish embryos. While Nkx2.5+ cells were increased overall in the nascent hearts of RA-deficient embryos, unexpectedly, we found that the major effect within this population was a significant expansion in the number of differentiating FHF CMs. In contrast to the expansion of the FHF, there was a progressive decrease in SHF progenitors at the arterial pole as the heart tube elongated. Temporal differentiation assays and immunostaining in RA-deficient embryos showed that the outflow tracts (OFTs) of the hearts were significantly smaller, containing fewer differentiated SHF-derived ventricular CMs and a complete absence of SHF-derived smooth muscle at later stages. At the venous pole of the heart, pacemaker cells of the sinoatrial node also failed to differentiate in RA-deficient embryos. Interestingly, genetic lineage tracing showed that the number of neural-crest derived CMs was not altered within the enlarged hearts of RA-deficient zebrafish embryos. Altogether, our data show that the enlarged hearts in RA-deficient zebrafish embryos are comprised of an expansion in earlier differentiating FHF-derived CMs coupled with a progressive depletion of the SHF, suggesting RA signaling determines the relative ratios of earlier- and later-differentiation cardiac progenitors within an expanded cardiac progenitor pool.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular Genetics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew Holowiecki
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joshua S Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
40
|
Martin KE, Waxman JS. Atrial and Sinoatrial Node Development in the Zebrafish Heart. J Cardiovasc Dev Dis 2021; 8:jcdd8020015. [PMID: 33572147 PMCID: PMC7914448 DOI: 10.3390/jcdd8020015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/31/2021] [Accepted: 02/04/2021] [Indexed: 12/11/2022] Open
Abstract
Proper development and function of the vertebrate heart is vital for embryonic and postnatal life. Many congenital heart defects in humans are associated with disruption of genes that direct the formation or maintenance of atrial and pacemaker cardiomyocytes at the venous pole of the heart. Zebrafish are an outstanding model for studying vertebrate cardiogenesis, due to the conservation of molecular mechanisms underlying early heart development, external development, and ease of genetic manipulation. Here, we discuss early developmental mechanisms that instruct appropriate formation of the venous pole in zebrafish embryos. We primarily focus on signals that determine atrial chamber size and the specialized pacemaker cells of the sinoatrial node through directing proper specification and differentiation, as well as contemporary insights into the plasticity and maintenance of cardiomyocyte identity in embryonic zebrafish hearts. Finally, we integrate how these insights into zebrafish cardiogenesis can serve as models for human atrial defects and arrhythmias.
Collapse
Affiliation(s)
- Kendall E. Martin
- Molecular Genetics, Biochemistry, and Microbiology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Correspondence:
| |
Collapse
|
41
|
Chen J. NF-Y is critical for the proper growth of zebrafish embryonic heart and its cardiomyocyte proliferation. Genesis 2021; 59:e23408. [PMID: 33417743 DOI: 10.1002/dvg.23408] [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: 03/06/2020] [Revised: 12/27/2020] [Accepted: 12/29/2020] [Indexed: 11/06/2022]
Abstract
The ubiquitous NF-Y gene regulates the expression of different genes in various signaling pathways. However, the function of NF-Y in zebrafish heart development is largely unknown. Previously we identified a same group of cell cycle related gene cluster (CCRG) was downregulated in the embryonic hearts with impeded growth due to various stresses. The promoter regions of these CCRG genes shared a most common motif for NF-Y. Chromatin immunoprecipitation experiment demonstrated that the binding of NF-Y to its motif was real on the CCRG candidate gene promoters. Knockdown of embryonic NF-Y by morpholinos led to a small heart, mimicking the abnormal heart phenotype caused by other stresses. In parallel the expression of certain CCRG candidate genes was reduced in the NF-Y A morphant hearts exposed to malignant environments. Absence of NF-Y A also led to undermine cardiomyocyte proliferation and hence less total number of caridomyocytes per heart. Trans-AM Elisa experiment also found that in the presence of the stresses such as TCDD and TNNT2 MO, the binding capacity of NF-Y A subunit to its core motif was reduced. We conclude that NF-Y sustains proper cardiomyocyte proliferation in the heart, thus it plays a positive role in promoting early zebrafish heart growth.
Collapse
Affiliation(s)
- Jing Chen
- Provincial University Key Laboratory of Cellular Stress Response and Metabolic Regulation, College of Life Sciences, Fujian Normal University, Fuzhou, China
| |
Collapse
|
42
|
Machikhin AS, Volkov MV, Burlakov AB, Khokhlov DD, Potemkin AV. Blood Vessel Imaging at Pre-Larval Stages of Zebrafish Embryonic Development. Diagnostics (Basel) 2020; 10:diagnostics10110886. [PMID: 33143148 PMCID: PMC7692510 DOI: 10.3390/diagnostics10110886] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/25/2023] Open
Abstract
The zebrafish (Danio rerio) is an increasingly popular animal model biological system. In cardiovascular research, it has been used to model specific cardiac phenomena as well as to identify novel therapies for human cardiovascular disease. While the zebrafish cardiovascular system functioning is well examined at larval stages, the mechanisms by which vessel activity is initiated remain a subject of intense investigation. In this research, we report on an in vivo stain-free blood vessel imaging technique at pre-larval stages of zebrafish embryonic development. We have developed the algorithm for the enhancement, alignment and spatiotemporal analysis of bright-field microscopy images of zebrafish embryos. It enables the detection, mapping and quantitative characterization of cardiac activity across the whole specimen. To validate the proposed approach, we have analyzed multiple data cubes, calculated vessel images and evaluated blood flow velocity and heart rate dynamics in the absence of any anesthesia. This non-invasive technique may shed light on the mechanism of vessel activity initiation and stabilization as well as the cardiovascular system’s susceptibility to environmental stressors at early developmental stages.
Collapse
Affiliation(s)
- Alexander S. Machikhin
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
| | - Mikhail V. Volkov
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| | - Alexander B. Burlakov
- Department of Ichthyology, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Demid D. Khokhlov
- Laboratory of Acousto-optical Spectroscopy, Scientific and Technological Center of Unique Instrumentation, Russian Academy of Sciences, 117342 Moscow, Russia;
- Correspondence:
| | - Andrey V. Potemkin
- Department of Applied Optics, University ITMO, 190000 Saint Petersburg, Russia; (M.V.V.); (A.V.P.)
| |
Collapse
|
43
|
Merola C, Perugini M, Conte A, Angelozzi G, Bozzelli M, Amorena M. Embryotoxicity of methylparaben to zebrafish (Danio rerio) early-life stages. Comp Biochem Physiol C Toxicol Pharmacol 2020; 236:108792. [PMID: 32428600 DOI: 10.1016/j.cbpc.2020.108792] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/09/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
Methylparaben (MeP) is widely used as preservative in personal care products, food commodities and pharmaceuticals due to its antimicrobial properties. Its widespread use resulted in the contamination of aquatic environment and raised concerns about the potential adverse effects on human health, especially in the developing organisms. The aim of the present study was to evaluate the embryotoxicity of MeP in zebrafish early-life stages applying the benchmark-dose (BMD) methodology to Fish embryo acute toxicity (FET) tests-OECD guideline 236. Toxic effects were studied by daily evaluation of lethal endpoints, hatching rate and sublethal alterations. Zebrafish fertilized eggs were exposed until 96 h post fertilization (hpf) to five concentrations of MeP: 1 mg/L, 10 mg/L, 30 mg/L, 60 mg/L and 80 mg/L. The lethal concentration 50 (LC 50) was 72.67 mg/L. Indeed, BMD confidence interval (lower bound, BMDL-upper bound, BMDU) was 40.8-57.4 mg/L for lethal endpoints and 16-26.5 mg/L for toxicity index, that includes both lethal and sublethal alterations. Zebrafish embryos exposed to MeP developed sublethal alterations including pericardial edema, yolk edema, blood stasis, reduction in blood circulation, reduced heartbeat and notochord curvature. The number of embryos exposed to the highest concentrations of MeP that reported sublethal alterations increased between 24hpf and 48 hpf-72 hpf-96 hpf. Only zebrafish larvae treated with 30 mg/L of MeP showed behavioural changes. This study highlighted the detrimental effects of MeP on zebrafish early-life stages with attention to its developmental toxicity.
Collapse
Affiliation(s)
- C Merola
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Italy
| | - M Perugini
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Italy.
| | - A Conte
- Istituto Zooprofilattico Sperimentale "G. Caporale", via Campo Boario, 64100 Teramo, Italy
| | - G Angelozzi
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Italy
| | - M Bozzelli
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Italy
| | - M Amorena
- Faculty of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Italy
| |
Collapse
|
44
|
Sun Y, Cao Y, Tong L, Tao F, Wang X, Wu H, Wang M. Exposure to prothioconazole induces developmental toxicity and cardiovascular effects on zebrafish embryo. CHEMOSPHERE 2020; 251:126418. [PMID: 32443233 DOI: 10.1016/j.chemosphere.2020.126418] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 06/11/2023]
Abstract
Prothioconazole is a fungicide that has been widely used in general agriculture and livestock husbandry. This study evaluated the acute toxicity of prothioconazole to zebrafish embryos by assessing their hatching rate and malformation when exposed to different concentrations of prothioconazole. The 96 h-LC50 value of zebrafish embryos was 1.70 mg/L. Upon exposure to 0.85 mg/L, the mortality rate of the embryos significantly increased while their hatching rate decreased significantly. At prothioconazole concentrations higher than 0.43 mg/L, developmental morphologic abnormalities such as heart and yolk-sac edema, spine curvature, tail deformity, shortened body length and decreased eye area were observed. The heart rate of embryos decreased in a dose-dependent fashion during the exposure time. Prothioconazole exposure also resulted in increased rates of cardiac malformation detected by significant increase in the distance between the sinus venosus and bulbus arteriosus and the pericardium area. Moreover, the expression levels of genes related to cardiac development (amhc, vmhc, fli1, hand2, gata4, nkx2.5, tbx5 and atp2a2a) were significantly altered after exposure to prothioconazole. Indeed, this study revealed the adverse effects on the developmental and cardiovascular system of zebrafish embryo caused by prothioconazole. It further elucidated the risk of prothioconazole exposure to vertebrate cardiovascular toxicity. As such, it provides a theoretical foundation for pesticide risk management measures.
Collapse
Affiliation(s)
- Yongqi Sun
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Yi Cao
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Lili Tong
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Fangyi Tao
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Xiaonan Wang
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China
| | - Huiming Wu
- School of Agricultural and Food Science, Zhejiang Agriculture & Forestry University, Hangzhou, China.
| | - Mengcen Wang
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Pesticide & Environmental Toxicology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
45
|
Heigwer J, Kutzner J, Haeussler M, Burkhalter MD, Draebing T, Juergensen L, Katus HA, Philipp M, Westhoff JH, Hassel D. miR-103/107 regulates left-right asymmetry in zebrafish by modulating Kupffer's vesicle development and ciliogenesis. Biochem Biophys Res Commun 2020; 527:432-439. [PMID: 32334837 DOI: 10.1016/j.bbrc.2020.04.066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
In zebrafish, cilia movement within the Kupffer's vesicle (KV) generates a fluid flow responsible for accumulating nodal signals exclusively in the left lateral plate mesoderm, thereby initiating left-right patterning (LRP). Defects in LRP cause devastating congenital disorders including congenital heart malformations due to organ mis-positioning. We identified the miR-103/107 family to be involved in regulating LRP. Depletion of miR-103/107 in zebrafish embryos resulted in malpositioned and malformed visceral organs and hearts due to disturbed LRP gene expression, indicating early defects in LRP. Additionally, loss of miR-103/107 affected KV morphogenesis and cilia formation without disturbing endoderm development. Human fibroblasts depleted of miR-103a/107 often failed to extend cilia or developed shorter cilia, indicating functional conservation between species. We identified arl6, araf and foxH1 as direct targets of miR-103/107 providing a mechanistic link to cilia development and nodal signal titration. We describe a new microRNA family controlling KV development and hence influencing establishment of internal organ asymmetry.
Collapse
Affiliation(s)
- Jana Heigwer
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany; Department of Pediatrics I, University Children's Hospital Heidelberg, 69120, Heidelberg, Germany.
| | - Juliane Kutzner
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Monika Haeussler
- Institute of Biochemistry and Molecular Biology (iBMB), Ulm University, 89081, Ulm, Germany
| | - Martin D Burkhalter
- Institute of Biochemistry and Molecular Biology (iBMB), Ulm University, 89081, Ulm, Germany; Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, 72074, Tübingen, Germany
| | - Thomas Draebing
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Lonny Juergensen
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| | - Melanie Philipp
- Institute of Biochemistry and Molecular Biology (iBMB), Ulm University, 89081, Ulm, Germany; Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, 72074, Tübingen, Germany
| | - Jens H Westhoff
- Department of Pediatrics I, University Children's Hospital Heidelberg, 69120, Heidelberg, Germany
| | - David Hassel
- Department of Internal Medicine III, Cardiology, Heidelberg University Hospital, 69120, Heidelberg, Germany and DZHK (German Centre for Cardiovascular Research), Partner Site, Heidelberg, Mannheim, Germany
| |
Collapse
|
46
|
Rubbini D, Cornet C, Terriente J, Di Donato V. CRISPR Meets Zebrafish: Accelerating the Discovery of New Therapeutic Targets. SLAS DISCOVERY 2020; 25:552-567. [PMID: 32462967 DOI: 10.1177/2472555220926920] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bringing a new drug to the market costs an average of US$2.6 billion and takes more than 10 years from discovery to regulatory approval. Despite the need to reduce cost and time to increase productivity, pharma companies tend to crowd their efforts in the same indications and drug targets. This results in the commercialization of drugs that share the same mechanism of action (MoA) and, in many cases, equivalent efficacies among them-an outcome that helps neither patients nor the balance sheet of the companies trying to bring therapeutics to the same patient population. Indeed, the discovery of new therapeutic targets, based on a deeper understanding of the disease biology, would likely provide more innovative MoAs and potentially greater drug efficacies. It would also bring better chances for identifying appropriate treatments according to the patient's genetic stratification. Nowadays, we count with an enormous amount of unprocessed information on potential disease targets that could be extracted from omics data obtained from patient samples. In addition, hundreds of pharmacological and genetic screenings have been performed to identify innovative drug targets. Traditionally, rodents have been the animal models of choice to perform functional genomic studies. The high experimental cost, combined with the low throughput provided by those models, however, is a bottleneck for discovering and validating novel genetic disease associations. To overcome these limitations, we propose that zebrafish, in conjunction with the use of CRISPR/Cas9 genome-editing tools, could streamline functional genomic processes to bring biologically relevant knowledge on innovative disease targets in a shorter time frame.
Collapse
Affiliation(s)
- Davide Rubbini
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Carles Cornet
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Javier Terriente
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Vincenzo Di Donato
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| |
Collapse
|
47
|
Shrestha R, Lieberth J, Tillman S, Natalizio J, Bloomekatz J. Using Zebrafish to Analyze the Genetic and Environmental Etiologies of Congenital Heart Defects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:189-223. [PMID: 32304074 DOI: 10.1007/978-981-15-2389-2_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Congenital heart defects (CHDs) are among the most common human birth defects. However, the etiology of a large proportion of CHDs remains undefined. Studies identifying the molecular and cellular mechanisms that underlie cardiac development have been critical to elucidating the origin of CHDs. Building upon this knowledge to understand the pathogenesis of CHDs requires examining how genetic or environmental stress changes normal cardiac development. Due to strong molecular conservation to humans and unique technical advantages, studies using zebrafish have elucidated both fundamental principles of cardiac development and have been used to create cardiac disease models. In this chapter we examine the unique toolset available to zebrafish researchers and how those tools are used to interrogate the genetic and environmental contributions to CHDs.
Collapse
Affiliation(s)
- Rabina Shrestha
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Jaret Lieberth
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Savanna Tillman
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | - Joseph Natalizio
- Department of Biology, University of Mississippi, Oxford, MS, USA
| | | |
Collapse
|
48
|
Meneghetti G, Skobo T, Chrisam M, Fontana CM, Facchinello N, Nazio F, Cecconi F, Bonaldo P, Dalla Valle L. Zebrafish ambra1a and ambra1b Silencing Affect Heart Development. Zebrafish 2020; 17:163-176. [PMID: 32320344 DOI: 10.1089/zeb.2020.1860] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In zebrafish, two paralogous genes, activating molecule in beclin-1 (BECN1)-regulated autophagy ambra1a and ambra1b, both required for the autophagic process and during development, encode the protein AMBRA1, a positive regulator of early steps of autophagosome formation. As transcripts for both genes are expressed during embryogenesis in the heart region, in this work, we investigated the effects of ambra1a and ambra1b knockdown on heart development by means of morpholino oligonucleotides (MOs). Silencing of the two proteins by MOs directed against the ATG translation initiation codon affects cardiac morphogenesis, resulting in a small, string-like heart with pericardial edema, whereas treatment with splice-blocking MOs does not lead to overt cardiac phenotypes, thus revealing the relevance of maternally supplied ambra1 transcripts for heart development. Co-injection of both ATG-MOs determines a more severe cardiac phenotype, with prominent pericardial edema. Whole-mount in situ hybridization (WMISH) for myosin light chain 7 (myl7), as well as ambra1 ATG-MO microinjection in zebrafish transgenic line expressing green fluorescent protein in the heart, revealed defects with the heart jogging process followed by imperfect cardiac looping. Moreover, WMISH of homeodomain transcription factor 2 isoform c (pitx2c) transcripts showed both bilateral and reversed pitx2c expression in morphants. The morphants' cardiac phenotypes were effectively rescued by co-injection of MOs with human AMBRA1 (hAMBRA1) messenger RNA (mRNA), pointing at the conservation of Ambra1 functions during evolution. Co-injections of ambra1 ATG-MOs with a hAMBRA1 mRNA mutated in the protein phosphatase 2a (PP2A) binding sites (hAMBRA1PXP) were not able to rescue the cardiac phenotypes, at the difference from wild-type hAMBRA1 mRNA, and treatment of zebrafish embryos with the specific PP2A inhibitor cantharidin resulted in similar developmental cardiac defects. These results suggest a critical role for AMBRA1 in vertebrate heart development, likely involving the binding site for the PP2A phosphatase.
Collapse
Affiliation(s)
| | - Tatjana Skobo
- Department of Biology and University of Padova, Padova, Italy
| | - Martina Chrisam
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | | | - Francesca Nazio
- Department of Pediatric Hemato-Oncology and Cell and Gene therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Cecconi
- Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | |
Collapse
|
49
|
Abstract
As the first organ to form and function in all vertebrates, the heart is crucial to development. Tightly-regulated levels of retinoic acid (RA) are critical for the establishment of the regulatory networks that drive normal cardiac development. Thus, the heart is an ideal organ to investigate RA signaling, with much work remaining to be done in this area. Herein, we highlight the role of RA signaling in vertebrate heart development and provide an overview of the field's inception, its current state, and in what directions it might progress so that it may yield fruitful insight for therapeutic applications within the domain of regenerative medicine.
Collapse
|
50
|
Shen R, Yu Y, Lan R, Yu R, Yuan Z, Xia Z. The cardiovascular toxicity induced by high doses of gatifloxacin and ciprofloxacin in zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 254:112861. [PMID: 31454568 DOI: 10.1016/j.envpol.2019.07.029] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 07/04/2019] [Accepted: 07/06/2019] [Indexed: 05/11/2023]
Abstract
As a new type of pollutant, fluoroquinolones (FQs) antibiotics are ubiquitous in environment and have some threat to human health and ecological environment. Their ecological toxicity to the environment urgently need to be assessed. Therefore, we firstly explored the toxic effects and possible mechanism of cardiovascular toxicity induced by gatifloxacin (GTFX) and ciprofloxacin (CPFX) using zebrafish model. After 24 h exposure, the zebrafish treated with GTFX showed pericardial edema which was further investigated by histopathological examination, while CPFX exposure did not induce morphological abnormalities. However, both of them induced cardiac dysfunction, such as decreased heart rate and cardiac output which was showed a positive correlation with the concentration. To better understand the possible molecular mechanisms underlying cardiovascular toxicity in zebrafish, we investigated the transcriptional level of genes related to calcium signaling pathway and cardiac muscle contraction. The results indicated that the expression of ATPase (atp2a1l) and cardiac troponin C (tnnc1a) genes were significantly inhibited, the expression of calcium channel (cacna1ab) gene showed slight promoted trend after CPFX exposure. For zebrafish treated with GTFX, the expression of atp2a1l genes was also significantly inhibited, while the expression of tnnc1a genes was slightly inhibited and cacna1ab genes expression had no obvious effect. The present study firstly revealed that GTFX exposure can induce morphological and functional abnormalities on the cardiovascular system of zebrafish. Though CPFX exposure did not induce morphological abnormalities, the function of cardiovascular system was still damaged. Mechanistically, this toxicity might result from the pressure of down-regulation of genes associated with calcium signaling pathway and cardiac muscle contraction. The results of this study can provide a valuable theoretical basis for the establishment of FQs environmental quality standards in water environment, environmental drug regulation and risk management.
Collapse
Affiliation(s)
- Rong Shen
- College of Bioengineering, Chongqing University, Chongqing 400044, PR China; College of Bioengineering, Beijing Polytechnic, Beijing 100176, PR China
| | - Yichang Yu
- Research Center of Environmental Engineering Technology, Chongqing Academy of Environmental Science, Chongqing 401120, PR China
| | - Rong Lan
- College of Bioengineering, Beijing Polytechnic, Beijing 100176, PR China
| | - Ran Yu
- College of Bioengineering, Beijing Polytechnic, Beijing 100176, PR China
| | - Ze Yuan
- College of Bioengineering, Beijing Polytechnic, Beijing 100176, PR China
| | - Zhining Xia
- College of Bioengineering, Chongqing University, Chongqing 400044, PR China.
| |
Collapse
|