1
|
Ford C, de Sena-Tomás C, Wun TTR, Aleman AG, Rangaswamy U, Leyhr J, Nuñez MI, Gao CZ, Nim HT, See M, Coppola U, Waxman JS, Ramialison M, Haitina T, Smeeton J, Sanges R, Targoff KL. Nkx2.7 is a conserved regulator of craniofacial development. Nat Commun 2025; 16:3802. [PMID: 40268889 PMCID: PMC12019251 DOI: 10.1038/s41467-025-58821-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
Craniofacial malformations arise from developmental defects in the head, face, and neck with phenotypes such as 22q11.2 deletion syndrome illustrating a developmental link between cardiovascular and craniofacial morphogenesis. NKX2-5 is a key cardiac transcription factor associated with congenital heart disease and mouse models of Nkx2-5 deficiency highlight roles in cardiac development. In zebrafish, nkx2.5 and nkx2.7 are paralogues in the NK4 family expressed in cardiomyocytes and pharyngeal arches. Despite shared cellular origins of cardiac and craniofacial tissues, the function of NK4 factors in head and neck patterning has not been elucidated. Molecular evolutionary analysis of NK4 genes shows that nkx2.5 and nkx2.7 are ohnologs resulting from whole genome duplication events. Nkx2.7 serves as a previously unappreciated regulator of branchiomeric muscle and cartilage formation for which nkx2.5 cannot fully compensate. Mechanistically, our results highlight that Nkx2.7 patterns the cranial neural crest and functions upstream of Endothelin1 to inhibit Notch signals. Together, our studies shed light on an evolutionarily conserved Nkx transcription factor with unique functions in vertebrate craniofacial development, advancing our understanding of congenital head and neck deformities.
Collapse
Affiliation(s)
- Caitlin Ford
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer, 08036, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, 08028, Barcelona, Spain
| | - Tint Tha Ra Wun
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Angelika G Aleman
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Uday Rangaswamy
- Functional and Structural Genomics, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
| | - Jake Leyhr
- Department of Organismal Biology, Uppsala University, 75236, Uppsala, Sweden
| | - María I Nuñez
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Cynthia Zehui Gao
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Computer Science, Columbia University, New York, NY, 10027, USA
| | - Hieu T Nim
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Michael See
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Biological Sciences, Florida Gulf Coast University, Fort Myers, FL, 33965, 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
| | - Mirana Ramialison
- The Novo Nordisk Foundation Center for Stem Cell Medicine & Stem Cell Biology, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
- Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, VIC, 3052, Australia
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, 75236, Uppsala, Sweden
| | - Joanna Smeeton
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Rehabilitation and Regenerative Medicine, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Remo Sanges
- Functional and Structural Genomics, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, 34136, Trieste, Italy
- Central RNA Laboratory, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152, Genova, Italy
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
2
|
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
|
3
|
Allouh MZ, Rizvi SFA, Alamri A, Jimoh Y, Aouda S, Ouda ZH, Hamad MIK, Perez-Cruet M, Chaudhry GR. Mesenchymal stromal/stem cells from perinatal sources: biological facts, molecular biomarkers, and therapeutic promises. Stem Cell Res Ther 2025; 16:127. [PMID: 40055783 PMCID: PMC11889844 DOI: 10.1186/s13287-025-04254-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
The use of mesenchymal stem cells (MSCs) from perinatal tissue sources has gained attention due to their availability and lack of significant ethical or moral concerns. These cells have a higher proliferative capability than adult MSCs and less immunogenic or tumorigenesis risk than fetal and embryonic stem cells. Additionally, they do not require invasive isolation methods like fetal and adult MSCs. We reviewed the main biological and therapeutic aspects of perinatal MSCs in a three-part article. In the first part, we revised the main biological features and characteristics of MSCs and the advantages of perinatal MSCs over other types of SCs. In the second part, we provided a detailed molecular background for the main biomarkers that can be used to identify MSCs. In the final part, we appraised the therapeutic application of perinatal MSCs in four major degenerative disorders: degenerative disc disease, retinal degenerative diseases, ischemic heart disease, and neurodegenerative diseases. In conclusion, there is no single specific molecular marker to identify MSCs. We recommend using at least two positive markers of stemness (CD29, CD73, CD90, or CD105) and two negative markers (CD34, CD45, or CD14) to exclude the hematopoietic origin. Moreover, utilizing perinatal MSCs for managing degenerative diseases presents a promising therapeutic approach. This review emphasizes the significance of employing more specialized progenitor cells that originated from the perinatal MSCs. The review provides scientific evidence from the literature that applying these progenitor cells in therapeutic procedures provides a greater regenerative capacity than the original primitive MSCs. Finally, this review provides a valuable reference for researchers exploring perinatal MSCs and their therapeutic applications.
Collapse
Affiliation(s)
- Mohammed Z Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE.
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| | - Syed Faizan Ali Rizvi
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Ali Alamri
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Yusuf Jimoh
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA
| | - Salma Aouda
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, UAE
| | - Zakaria H Ouda
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE
| | - Mohammad I K Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, P. O. Box: 15551, Al Ain, UAE
| | - Mick Perez-Cruet
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
- Department of Neurosurgery, Corewell Health, Royal Oak, MI, USA
| | - G Rasul Chaudhry
- OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
- Department of Biological Sciences, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
4
|
Wu Y, Yu W, Song Z, He J, Li Z, Chen Q, Wang S, Li P, Cheng S. The Acute Toxicity and Cardiotoxic Effects of Levofloxacin on Zebrafish ( Danio rerio). TOXICS 2025; 13:122. [PMID: 39997937 PMCID: PMC11861866 DOI: 10.3390/toxics13020122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025]
Abstract
Emerging contaminants refer to chemical substances that have not been widely regulated but possess the potential to cause adverse effects on both the environment and human health. Antibiotics, as emerging contaminants, pose significant threats to ecosystems and human health due to their widespread use and persistence in the environment. Levofloxacin, a broad-spectrum fluoroquinolone antibiotic, is commonly employed in the treatment of bacterial infections, and has been frequently detected in environmental matrices and freshwater systems. In this study, we assessed the effects of levofloxacin on hatchability, mortality rates, malformations, behavioral changes, and cardiac development in zebrafish embryos by exposing them to varying concentrations of levofloxacin (0, 0.5, 1, 2, 4, and 8 mM). Our results demonstrate that levofloxacin exposure significantly impaired the growth and development of zebrafish larvae, particularly at higher concentrations. Notable effects included reduced body length, abnormal yolk sac and swim bladder development, pericardial edema, prolonged distances between the sinus venosus and arteriolar bulb (SV-BA), and disruptions in heart rate. Quantitative PCR analysis further revealed that levofloxacin exposure significantly upregulated the expression of key cardiac development genes in zebrafish larvae, including nppa, myh6, cacna1ab, myl7, gata4, nkx2.5, tbx2b, and tbx5b. These findings indicate that levofloxacin exposure exerts significant toxic effects on both embryonic and larval growth as well as heart development and gene expression in zebrafish. This study provides critical insights into the potential ecological risks posed by levofloxacin along with other antibiotics while laying a foundation for further investigation into their toxicological mechanisms.
Collapse
Affiliation(s)
- Yixiao Wu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qi Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shiwei Wang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ping Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.W.); (W.Y.); (Z.S.); (J.H.); (Z.L.); (Q.C.); (S.W.); (P.L.)
- The Second Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
- School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
5
|
Yoon Y, Lee Y, Cho M. Acute toxicity assessment and QSAR modeling of zebrafish embryos exposed to methyl paraben and its halogenated byproducts. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 370:122844. [PMID: 39405839 DOI: 10.1016/j.jenvman.2024.122844] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/21/2024] [Accepted: 10/06/2024] [Indexed: 11/17/2024]
Abstract
Halogenated methyl parabens are formed readily during water chlorination, with or without bromide ion presence. However, research gaps persist in in vivo toxicological assessments of vertebrates exposed to halo-MePs. To address this gap, this study evaluated acute toxicities at 24-96 h-post-fertilization in zebrafish embryos exposed to methyl paraben and its mono- or di-halogenated derivatives, using various apical endpoints. Significant enhanced toxic effects were confirmed for halo-MePs compared to MeP on embryo coagulation (3-19 fold), heartbeat rate decrement (11-80 fold), deformity rate increment (9-68 fold), and hatching failure (4-33 fold), with parentheses indicating the determined toxic potency ratios. Moreover, halo-MePs showed a significantly higher increase in biochemical levels of reactive oxygen species, catalase, superoxide dismutase, and malondialdehyde, while acetylcholinesterase activity was inhibited compared to NT and MeP. The experimental toxic potencies (log(1/EC50 or LC50)) were compared with the predicted ones (log(1/EC50 or LC50, baseline)) using the baseline toxicity Quantitative Structure-Activity Relationship previously established for zebrafish embryos. Halo-MePs were specific (or reactive) toxicants based on their toxic ratios of more than 10 for apical endpoints including heartbeat rate, deformity rate, and hatching rate, while MeP acted as a baseline toxicant. Overall, this study presents the comprehensive toxicological assessment of halo-MePs in zebrafish embryos, contributing to an essential in vivo toxicity database for halogenated phenolic contaminants in aquatic ecosystems.
Collapse
Affiliation(s)
- Younggun Yoon
- GwangJu Institute, 55, Jingoksandanjungang-ro, Gwangsan-gu, Gwangju, 62465, Republic of Korea; Gyeongnam Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology (KIT), Gyeongsangnam-do, 52834, Republic of Korea; Division of Biotechnology, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea; School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| | - Yunho Lee
- School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| | - Min Cho
- Division of Biotechnology, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk, 54596, Republic of Korea.
| |
Collapse
|
6
|
Sloutskin A, Itzhak D, Vogler G, Pozeilov H, Ideses D, Alter H, Adato O, Shachar H, Doniger T, Shohat-Ophir G, Frasch M, Bodmer R, Duttke SH, Juven-Gershon T. From promoter motif to cardiac function: a single DPE motif affects transcription regulation and organ function in vivo. Development 2024; 151:dev202355. [PMID: 38958007 DOI: 10.1242/dev.202355] [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: 09/18/2023] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
Transcription initiates at the core promoter, which contains distinct core promoter elements. Here, we highlight the complexity of transcriptional regulation by outlining the effect of core promoter-dependent regulation on embryonic development and the proper function of an organism. We demonstrate in vivo the importance of the downstream core promoter element (DPE) in complex heart formation in Drosophila. Pioneering a novel approach using both CRISPR and nascent transcriptomics, we show the effects of mutating a single core promoter element within the natural context. Specifically, we targeted the downstream core promoter element (DPE) of the endogenous tin gene, encoding the Tinman transcription factor, a homologue of human NKX2-5 associated with congenital heart diseases. The 7 bp substitution mutation results in massive perturbation of the Tinman regulatory network that orchestrates dorsal musculature, which is manifested as physiological and anatomical changes in the cardiac system, impaired specific activity features, and significantly compromised viability of adult flies. Thus, a single motif can have a critical impact on embryogenesis and, in the case of DPE, functional heart formation.
Collapse
Affiliation(s)
- Anna Sloutskin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dekel Itzhak
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hadar Pozeilov
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Diana Ideses
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Hadar Alter
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Orit Adato
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Hadar Shachar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tirza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Galit Shohat-Ophir
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Manfred Frasch
- Division of Developmental Biology, Department of Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen 91058, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sascha H Duttke
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
| | - Tamar Juven-Gershon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
7
|
Yoon Y, Cho M. Detrimental impacts and QSAR baseline toxicity assessment of Japanese medaka embryos exposed to methylparaben and its halogenated byproducts. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:171448. [PMID: 38453088 DOI: 10.1016/j.scitotenv.2024.171448] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
Despite the theoretical risk of forming halogenated methylparabens (halo-MePs) during water chlorination in the absence or presence of bromide ions, there remains a lack of in vivo toxicological assessments on vertebrate organisms for halo-MePs. This research addresses these gaps by investigating the lethal (assessed by embryo coagulation) or sub-lethal (assessed by hatching success/heartbeat rate) toxicity and teratogenicity (assessed by deformity rate) of MeP and its mono- and di-halogen derivatives (Cl- or Br-) using Japanese medaka embryos. In assessing selected apical endpoints to discern patterns in physiological or biochemical alterations, heightened toxic impacts were observed for halo-MePs compared to MeP. These include a higher incidence of embryo coagulation (4-36 fold), heartbeat rate decrement (11-36 fold), deformity rate increment (32-223 fold), hatching success decrement (11-59 fold), and an increase in Reactive Oxygen Species (ROS) level (1.2-7.4 fold)/Catalase (CAT) activity (1.7-2.8 fold). Experimentally determined LC50 values are correlated and predicted using a Quantitative Structure Activity Relationship (QSAR) based on the speciation-corrected liposome-water distribution ratio (Dlipw, pH 7.5). The QSAR baseline toxicity aligns well with (sub)lethal toxicity and teratogenicity, as evidenced by toxic ratio (TR) analysis showing TR < 10 for MeP exposure in all cases, while significant specific or reactive toxicity was found for halo-MeP exposure, with TR > 10 observed (excepting three values). Our extensive findings contribute novel insights into the intricate interplay of embryonic toxicity during the early-life-stage of Japanese medaka, with a specific focus on highlighting the potential hazards associated with halo-MePs compared to the parent compound MeP.
Collapse
Affiliation(s)
- Younggun Yoon
- Gyeongnam Department of Environmental Toxicology and Chemistry, Korea Institute of Toxicology (KIT), Gyeongsangnam-do, 52834, South Korea; Division of Biotechnology, SELS Center, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea.
| | - Min Cho
- Division of Biotechnology, SELS Center, College of Environmental and Bioresource Sciences, Jeonbuk National University, Iksan, Jeonbuk 54596, South Korea.
| |
Collapse
|
8
|
Beisaw A, Wu CC. Cardiomyocyte maturation and its reversal during cardiac regeneration. Dev Dyn 2024; 253:8-27. [PMID: 36502296 DOI: 10.1002/dvdy.557] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/03/2022] [Accepted: 12/03/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. Due to the limited proliferative and regenerative capacity of adult cardiomyocytes, the lost myocardium is not replenished efficiently and is replaced by a fibrotic scar, which eventually leads to heart failure. Current therapies to cure or delay the progression of heart failure are limited; hence, there is a pressing need for regenerative approaches to support the failing heart. Cardiomyocytes undergo a series of transcriptional, structural, and metabolic changes after birth (collectively termed maturation), which is critical for their contractile function but limits the regenerative capacity of the heart. In regenerative organisms, cardiomyocytes revert from their terminally differentiated state into a less mature state (ie, dedifferentiation) to allow for proliferation and regeneration to occur. Importantly, stimulating adult cardiomyocyte dedifferentiation has been shown to promote morphological and functional improvement after myocardial infarction, further highlighting the importance of cardiomyocyte dedifferentiation in heart regeneration. Here, we review several hallmarks of cardiomyocyte maturation, and summarize how their reversal facilitates cardiomyocyte proliferation and heart regeneration. A detailed understanding of how cardiomyocyte dedifferentiation is regulated will provide insights into therapeutic options to promote cardiomyocyte de-maturation and proliferation, and ultimately heart regeneration in mammals.
Collapse
Affiliation(s)
- Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Heidelberg, Germany
| | - Chi-Chung Wu
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
9
|
Cao C, Li L, Zhang Q, Li H, Wang Z, Wang A, Liu J. Nkx2.5: a crucial regulator of cardiac development, regeneration and diseases. Front Cardiovasc Med 2023; 10:1270951. [PMID: 38124890 PMCID: PMC10732152 DOI: 10.3389/fcvm.2023.1270951] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Cardiomyocytes fail to regenerate after birth and respond to mitotic signals through cellular hypertrophy rather than cellular proliferation. Necrotic cardiomyocytes in the infarcted ventricular tissue are eventually replaced by fibroblasts, generating scar tissue. Cardiomyocyte loss causes localized systolic dysfunction. Therefore, achieving the regeneration of cardiomyocytes is of great significance for cardiac function and development. Heart development is a complex biological process. An integral cardiac developmental network plays a decisive role in the regeneration of cardiomyocytes. During this process, genetic epigenetic factors, transcription factors, signaling pathways and small RNAs are involved in regulating the developmental process of the heart. Cardiomyocyte-specific genes largely promote myocardial regeneration, among which the Nkx2.5 transcription factor is one of the earliest markers of cardiac progenitor cells, and the loss or overexpression of Nkx2.5 affects cardiac development and is a promising candidate factor. Nkx2.5 affects the development and function of the heart through its multiple functional domains. However, until now, the specific mechanism of Nkx2.5 in cardiac development and regeneration is not been fully understood. Therefore, this article will review the molecular structure, function and interaction regulation of Nkx2.5 to provide a new direction for cardiac development and the treatment of heart regeneration.
Collapse
Affiliation(s)
- Ce Cao
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Qian Zhang
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haoran Li
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ziyan Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Aoao Wang
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
| | - Jianxun Liu
- Institute of Basic Medical Sciences of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing Key Laboratory of Chinese Materia Pharmacology, National Clinical Research Center of Traditional Chinese Medicine for Cardiovascular Diseases, Beijing, China
- Institute of Chinese Medicine Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
10
|
Sloutskin A, Itzhak D, Vogler G, Ideses D, Alter H, Shachar H, Doniger T, Frasch M, Bodmer R, Duttke SH, Juven-Gershon T. A single DPE core promoter motif contributes to in vivo transcriptional regulation and affects cardiac function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.11.544490. [PMID: 37398300 PMCID: PMC10312617 DOI: 10.1101/2023.06.11.544490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Transcription is initiated at the core promoter, which confers specific functions depending on the unique combination of core promoter elements. The downstream core promoter element (DPE) is found in many genes related to heart and mesodermal development. However, the function of these core promoter elements has thus far been studied primarily in isolated, in vitro or reporter gene settings. tinman (tin) encodes a key transcription factor that regulates the formation of the dorsal musculature and heart. Pioneering a novel approach utilizing both CRISPR and nascent transcriptomics, we show that a substitution mutation of the functional tin DPE motif within the natural context of the core promoter results in a massive perturbation of Tinman's regulatory network orchestrating dorsal musculature and heart formation. Mutation of endogenous tin DPE reduced the expression of tin and distinct target genes, resulting in significantly reduced viability and an overall decrease in adult heart function. We demonstrate the feasibility and importance of characterizing DNA sequence elements in vivo in their natural context, and accentuate the critical impact a single DPE motif has during Drosophila embryogenesis and functional heart formation.
Collapse
Affiliation(s)
- Anna Sloutskin
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Dekel Itzhak
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Diana Ideses
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Alter
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Hadar Shachar
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tirza Doniger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Manfred Frasch
- Division of Developmental Biology, Department of Biology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sascha H Duttke
- School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Tamar Juven-Gershon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
11
|
Wu Y, Wang J, Xia Y, Tang K, Xu J, Wang A, Hu S, Wen L, Wang B, Yao W, Wang J. Toxic effects of isofenphos-methyl on zebrafish embryonic development. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114723. [PMID: 36871354 DOI: 10.1016/j.ecoenv.2023.114723] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/26/2023] [Accepted: 03/02/2023] [Indexed: 06/18/2023]
Abstract
Isofenphos-methyl (IFP) is widely used as an organophosphorus for controlling underground insects and nematodes. However, excessive use of IFP may pose potential risks to the environment and humans, but little information is available on its sublethal toxicity to aquatic organisms. To address this knowledge gap, the current study exposed zebrafish embryos to 2, 4, and 8 mg/L IFP within 6-96 h past fertilization (hpf) and measured mortality, hatching, developmental abnormalities, oxidative stress, gene expressions, and locomotor activity. The results showed that IFP exposure reduced the rates of heart and survival rate, hatchability, and body length of embryos and induced uninflated swim bladder and developmental malformations. Reduction in locomotive behavior and inhibition of AChE activity indicated that IFP exposure may induce behavioral defects and neurotoxicity in zebrafish larvae. IFP exposure also led to pericardial edema, longer venous sinus-arterial bulb (SV-BA) distance, and apoptosis of the heart cells. Moreover, IFP exposure increased the accumulation of reactive oxygen species (ROS) and the content of malonaldehyde (MDA), also elevated the levels of antioxidant enzymes of superoxide dismutase (SOD) and catalase (CAT), but decreased glutathione (GSH) levels in zebrafish embryos. The relative expressions of heart development-related genes (nkx2.5, nppa, gata4, and tbx2b), apoptosis-related genes (bcl2, p53, bax, and puma), and swim bladder development-related genes (foxA3, anxa5b, mnx1, and has2) were significantly altered by IFP exposure. Collectively, our results indicated that IFP induced developmental toxicity and neurotoxicity to zebrafish embryos and the mechanisms may be relevant to the activation of oxidative stress and reduction of acetylcholinesterase (AChE) content.
Collapse
Affiliation(s)
- Yuanzhao Wu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Jiawen Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Yumei Xia
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Kaiqin Tang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Jincheng Xu
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Anli Wang
- Fuli Institute of Food Science, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China; National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Hangzhou 310058, Zhejiang, China
| | - Shundi Hu
- The Research Institute of Advanced Technologies, Ningbo University, Ningbo 315211, China
| | - Luhong Wen
- The Research Institute of Advanced Technologies, Ningbo University, Ningbo 315211, China
| | - Binjie Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Weixuan Yao
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China
| | - Jiye Wang
- Key Laboratory of Drug Prevention and Control Technology of Zhejiang Province, The Department of Criminal Science and Technology, Zhejiang Police College, Hangzhou 310051, Zhejiang, China.
| |
Collapse
|
12
|
DeMoya RA, Forman-Rubinsky RE, Fontaine D, Shin J, Watkins SC, Lo C, Tsang M. Sin3a Associated Protein 130kDa, sap130, plays an evolutionary conserved role in zebrafish heart development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.30.534737. [PMID: 37034673 PMCID: PMC10081270 DOI: 10.1101/2023.03.30.534737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Hypoplastic left heart syndrome (HLHS) is a congenital heart disease where the left ventricle is reduced in size. A forward genetic screen in mice identified SIN3A associated protein 130kDa ( Sap130 ), a protein in the chromatin modifying SIN3A/HDAC1 complex, as a gene contributing to the digenic etiology of HLHS. Here, we report the role of zebrafish sap130 genes in heart development. Loss of sap130a, one of two Sap130 orthologs, resulted in smaller ventricle size, a phenotype reminiscent to the hypoplastic left ventricle in mice. While cardiac progenitors were normal during somitogenesis, diminution of the ventricle size suggest the Second Heart Field (SHF) was the source of the defect. To explore the role of sap130a in gene regulation, transcriptome profiling was performed after the heart tube formation to identify candidate pathways and genes responsible for the small ventricle phenotype. Genes involved in cardiac differentiation and cell communication were dysregulated in sap130a , but not in sap130b mutants. Confocal light sheet analysis measured deficits in cardiac output in MZsap130a supporting the notion that cardiomyocyte maturation was disrupted. Lineage tracing experiments revealed a significant reduction of SHF cells in the ventricle that resulted in increased outflow tract size. These data suggest that sap130a is involved in cardiogenesis via regulating the accretion of SHF cells to the growing ventricle and in their subsequent maturation for cardiac function. Further, genetic studies revealed an interaction between hdac1 and sap130a , in the incidence of small ventricles. These studies highlight the conserved role of Sap130a and Hdac1 in zebrafish cardiogenesis.
Collapse
Affiliation(s)
- Ricardo A DeMoya
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| | - Rachel E Forman-Rubinsky
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| | - Deon Fontaine
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| | - Joseph Shin
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| | - Simon C Watkins
- Department of Cell Biology and Molecular Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Cecilia Lo
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| | - Michael Tsang
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh PA 15213, USA
| |
Collapse
|
13
|
Inácio JM, Nunes MM, Almeida M, Cristo F, Anjos R, Belo JA. Gene-Edited Human-Induced Pluripotent Stem Cell Lines to Elucidate DAND5 Function throughout Cardiac Differentiation. Cells 2023; 12:520. [PMID: 36831187 PMCID: PMC9954670 DOI: 10.3390/cells12040520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
(1) Background: The contribution of gene-specific variants for congenital heart disease, one of the most common congenital disabilities, is still far from our complete understanding. Here, we applied a disease model using human-induced pluripotent stem cells (hiPSCs) to evaluate the function of DAND5 on human cardiomyocyte (CM) differentiation and proliferation. (2) Methods: Taking advantage of our DAND5 patient-derived iPSC line, we used CRISPR-Cas9 gene-editing to generate a set of isogenic hiPSCs (DAND5-corrected and DAND5 full-mutant). The hiPSCs were differentiated into CMs, and RT-qPCR and immunofluorescence profiled the expression of cardiac markers. Cardiomyocyte proliferation was analysed by flow cytometry. Furthermore, we used a multi-electrode array (MEA) to study the functional electrophysiology of DAND5 hiPSC-CMs. (3) Results: The results indicated that hiPSC-CM proliferation is affected by DAND5 levels. Cardiomyocytes derived from a DAND5 full-mutant hiPSC line are more proliferative when compared with gene-corrected hiPSC-CMs. Moreover, parallel cardiac differentiations showed a differential cardiac gene expression profile, with upregulated cardiac progenitor markers in DAND5-KO hiPSC-CMs. Microelectrode array (MEA) measurements demonstrated that DAND5-KO hiPSC-CMs showed prolonged field potential duration and increased spontaneous beating rates. In addition, conduction velocity is reduced in the monolayers of hiPSC-CMs with full-mutant genotype. (4) Conclusions: The absence of DAND5 sustains the proliferation of hiPSC-CMs, which alters their electrophysiological maturation properties. These results using DAND5 hiPSC-CMs consolidate the findings of the in vitro and in vivo mouse models, now in a translational perspective. Altogether, the data will help elucidate the molecular mechanism underlying this human heart disease and potentiates new therapies for treating adult CHD.
Collapse
Affiliation(s)
- José M. Inácio
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Mafalda M. Nunes
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Micael Almeida
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Fernando Cristo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| | - Rui Anjos
- Hospital de Santa Cruz, Centro Hospitalar Lisboa Ocidental, 1449-005 Lisboa, Portugal
| | - José A. Belo
- Stem Cells and Development Laboratory, iNOVA4Health, NOVA Medical School/Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082 Lisboa, Portugal
| |
Collapse
|
14
|
Lu J, Wang W, Zhang C, Xu W, Chen W, Tao L, Li Z, Cheng J, Zhang Y. Characterization of glyphosate-induced cardiovascular toxicity and apoptosis in zebrafish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 851:158308. [PMID: 36030873 DOI: 10.1016/j.scitotenv.2022.158308] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/21/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
Glyphosate, the most widely used herbicide, presents new hazards to human health. The developmental toxicity of glyphosate, especially its cardiovascular toxicity, needs to be closely monitored. To understand how glyphosate affects development, we performed toxicity tests on zebrafish embryos that were continuously exposed to glyphosate. The results indicated that glyphosate affected the overall development of zebrafish embryos, including mortality, hatching abnormalities, and decreased body length. At the same time, zebrafish embryos exposed to glyphosate exhibited cardiac malformations, including enlarged chambers, thinned ventricular walls, and rhythm disturbances. In addition, defective intersegmental vasculature occurred after glyphosate exposure, indicating impaired angiogenesis. Mechanistically, apoptosis clustered in the heart and vascular regions and levels of ATP and apoptosis-related genes including caspase-3, caspase-9, bax, and bcl-2 were altered. In summary, the data showed that cardiovascular toxicity caused by glyphosate exposure may be related to apoptosis. Our study provides evidence for a link between glyphosate exposure and cardiovascular developmental toxicity. This raises concerns regarding the health risks of the glyphosate.
Collapse
Affiliation(s)
- Jian Lu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weiguo Wang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Cheng Zhang
- Department of Pathology, UT southwestern Medical Center, Dallas, TX 75390, United States
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Weidong Chen
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zhong Li
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiagao Cheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
15
|
de Sena-Tomás C, Aleman AG, Ford C, Varshney A, Yao D, Harrington JK, Saúde L, Ramialison M, Targoff KL. Activation of Nkx2.5 transcriptional program is required for adult myocardial repair. Nat Commun 2022; 13:2970. [PMID: 35624100 PMCID: PMC9142600 DOI: 10.1038/s41467-022-30468-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 05/03/2022] [Indexed: 11/09/2022] Open
Abstract
The cardiac developmental network has been associated with myocardial regenerative potential. However, the embryonic signals triggered following injury have yet to be fully elucidated. Nkx2.5 is a key causative transcription factor associated with human congenital heart disease and one of the earliest markers of cardiac progenitors, thus it serves as a promising candidate. Here, we show that cardiac-specific RNA-sequencing studies reveal a disrupted embryonic transcriptional profile in the adult Nkx2.5 loss-of-function myocardium. nkx2.5-/- fish exhibit an impaired ability to recover following ventricular apex amputation with diminished dedifferentiation and proliferation. Complex network analyses illuminate that Nkx2.5 is required to provoke proteolytic pathways necessary for sarcomere disassembly and to mount a proliferative response for cardiomyocyte renewal. Moreover, Nkx2.5 targets embedded in these distinct gene regulatory modules coordinate appropriate, multi-faceted injury responses. Altogether, our findings support a previously unrecognized, Nkx2.5-dependent regenerative circuit that invokes myocardial cell cycle re-entry, proteolysis, and mitochondrial metabolism to ensure effective regeneration in the teleost heart.
Collapse
Affiliation(s)
- Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Angelika G Aleman
- Department of Physiology & Cellular Biophysics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Caitlin Ford
- Department of Genetics & Development, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Akriti Varshney
- Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
| | - Di Yao
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Jamie K Harrington
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA
| | - Leonor Saúde
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028, Lisboa, Portugal
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute & Systems Biology Institute Australia, Monash University, Clayton, VIC, 3800, Australia
- Murdoch Children's Research Institute & Department of Peadiatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, New York, NY, 10032, USA.
- Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA.
| |
Collapse
|
16
|
Abstract
The application of next-generation sequencing to study congenital heart disease (CHD) is increasingly providing new insights into the causes and mechanisms of this prevalent birth anomaly. Whole-exome sequencing analysis identifies damaging gene variants altering single or contiguous nucleotides that are assigned pathogenicity based on statistical analyses of families and cohorts with CHD, high expression in the developing heart and depletion of damaging protein-coding variants in the general population. Gene classes fulfilling these criteria are enriched in patients with CHD and extracardiac abnormalities, evidencing shared pathways in organogenesis. Developmental single-cell transcriptomic data demonstrate the expression of CHD-associated genes in particular cell lineages, and emerging insights indicate that genetic variants perturb multicellular interactions that are crucial for cardiogenesis. Whole-genome sequencing analyses extend these observations, identifying non-coding variants that influence the expression of genes associated with CHD and contribute to the estimated ~55% of unexplained cases of CHD. These approaches combined with the assessment of common and mosaic genetic variants have provided a more complete knowledge of the causes and mechanisms of CHD. Such advances provide knowledge to inform the clinical care of patients with CHD or other birth defects and deepen our understanding of the complexity of human development. In this Review, we highlight known and candidate CHD-associated human genes and discuss how the integration of advances in developmental biology research can provide new insights into the genetic contributions to CHD.
Collapse
Affiliation(s)
- Sarah U Morton
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Daniel Quiat
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Boston, MA, USA.
| |
Collapse
|
17
|
Wan M, Huang L, Liu J, Liu F, Chen G, Ni H, Xiong G, Liao X, Lu H, Xiao J, Tao Q, Cao Z. Cyclosporine A Induces Cardiac Developmental Toxicity in Zebrafish by Up-Regulation of Wnt Signaling and Oxidative Stress. Front Pharmacol 2021; 12:747991. [PMID: 34867350 PMCID: PMC8633111 DOI: 10.3389/fphar.2021.747991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/22/2021] [Indexed: 12/03/2022] Open
Abstract
Due to the widely application of Cyclosporine A (CsA) as an immunosuppressant in clinic, it is necessary to study its potential toxicity. Therefore, we used zebrafish as a model animal to evaluate the toxicity of CsA on embryonic development. Exposure of zebrafish embryos to CsA at concentrations of 5 mg/L, 10 mg/L, and 15 mg/L from 12 hpf to 72 hpf resulted in abnormal embryonic development, including cardiac malformation, pericardial edema, decreased heart rate, decreased blood flow velocity, deposition at yolk sac, shortened body length, and increased distance between venous sinus and arterial bulb (SV-BA). The expression of genes related to cardiac development was disordered, and the apoptotic genes were up-regulated. Oxidative stress level was up-regulated and accumulated in pericardium in a dose-dependent manner. Astaxanthin (ATX) treatment could significantly alleviate zebrafish heart defects. CsA induced up-regulation of Wnt signaling in zebrafish, and IWR-1, an inhibitor of Wnt signaling pathway, could effectively rescue the heart defects induced by CsA. Together, our study indicated that CsA induced cardiac developmental toxicity in zebrafish larvae through up-regulating oxidative stress and Wnt signaling, contributing to a more comprehensive evaluation of the safety of the drug.
Collapse
Affiliation(s)
- Mengqi Wan
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang University, Nanchang, China
| | - Ling Huang
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Jieping Liu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Fasheng Liu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Guilan Chen
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Huiwen Ni
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Guanghua Xiong
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Huiqiang Lu
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, China
| | - Qiang Tao
- Department of General Surgery, The Affiliated Children's Hospital of Nanchang University, Nanchang, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Jiangxi Key Laboratory of Developmental Biology of Organs, College of Life Sciences, Jinggangshan University, Ji'an, China
| |
Collapse
|
18
|
Stutt N, Song M, Wilson MD, Scott IC. Cardiac specification during gastrulation - The Yellow Brick Road leading to Tinman. Semin Cell Dev Biol 2021; 127:46-58. [PMID: 34865988 DOI: 10.1016/j.semcdb.2021.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
The question of how the heart develops, and the genetic networks governing this process have become intense areas of research over the past several decades. This research is propelled by classical developmental studies and potential clinical applications to understand and treat congenital conditions in which cardiac development is disrupted. Discovery of the tinman gene in Drosophila, and examination of its vertebrate homolog Nkx2.5, along with other core cardiac transcription factors has revealed how cardiac progenitor differentiation and maturation drives heart development. Careful observation of cardiac morphogenesis along with lineage tracing approaches indicated that cardiac progenitors can be divided into two broad classes of cells, namely the first and second heart fields, that contribute to the heart in two distinct waves of differentiation. Ample evidence suggests that the fate of individual cardiac progenitors is restricted to distinct cardiac structures quite early in development, well before the expression of canonical cardiac progenitor markers like Nkx2.5. Here we review the initial specification of cardiac progenitors, discuss evidence for the early patterning of cardiac progenitors during gastrulation, and consider how early gene expression programs and epigenetic patterns can direct their development. A complete understanding of when and how the developmental potential of cardiac progenitors is determined, and their potential plasticity, is of great interest developmentally and also has important implications for both the study of congenital heart disease and therapeutic approaches based on cardiac stem cell programming.
Collapse
Affiliation(s)
- Nathan Stutt
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Mengyi Song
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Michael D Wilson
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada
| | - Ian C Scott
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S1A8, Canada.
| |
Collapse
|
19
|
Li M, Yu T, Lai J, Han X, Hu J, Deng Z, Li D, Ye Z, Wang S, Hu C, Xu X. Ethoprophos induces cardiac toxicity in zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:113029. [PMID: 34847436 DOI: 10.1016/j.ecoenv.2021.113029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/21/2021] [Accepted: 11/24/2021] [Indexed: 06/13/2023]
Abstract
Ethoprophos is an effective and widely pesticide that used in controlling nemathelminth and soil insect. However, ethoprophos has been frequently detected in environment and freshwater. The potential toxicity to aquatic organisms is still not be explored. In this study, zebrafish embryo model was used to evaluated the toxicity of ethoprophos during cardiovascular developmental process of zebrafish. Zebrafish embryos were separately exposed to 10 mg/L, 20 mg/L, 30 mg/L, 40 mg/L and 50 mg/L of ethoprophos exposure at 96 h post-fertilization (hpf), which induced cardiac defects, such as low heart rate, pericardium edema and long SV-BA distance, but had no influence to vascular development. Mechanistically, the expression of cardiac-related genes were abnormal. Moreover, ethoprophos exposure significantly increased oxidative stress in zebrafish embryos by inhibiting the production of antioxidant enzyme (SOD) and activating reactive oxygen species. Expectedly, some apoptosis genes were induced and the apoptotic cardiomyocytes were detected by acridine orange staining. In addition, ethoprophos exposure also inhibited the expression of genes in wnt signaling pathway, such as β-catenin, Axin2, GSK3β and Sox9b. BML284, an activator of wnt signaling pathway, can rescue the cardiotoxic effect of embryos. These results indicated that oxidative stress and blocking wnt signaling pathway were molecular basis of ethoprophos-induced injure in zebrafish. Generally, our study showed that ethoprophos exposure led to severe cardiotoxicity to zebrafish embryo.
Collapse
Affiliation(s)
- Meifeng Li
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Tingting Yu
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Jingli Lai
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Xue Han
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Jihuan Hu
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, China
| | - Dongming Li
- Fuzhou Medical College, Nanchang University, Fuzhou 344000, China
| | - Zuocheng Ye
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Shanghong Wang
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Chengyu Hu
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China.
| | - Xiaowen Xu
- School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China; State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, Jiangxi, China.
| |
Collapse
|
20
|
Kikel-Coury NL, Brandt JP, Correia IA, O’Dea MR, DeSantis DF, Sterling F, Vaughan K, Ozcebe G, Zorlutuna P, Smith CJ. Identification of astroglia-like cardiac nexus glia that are critical regulators of cardiac development and function. PLoS Biol 2021; 19:e3001444. [PMID: 34793438 PMCID: PMC8601506 DOI: 10.1371/journal.pbio.3001444] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/18/2021] [Indexed: 01/09/2023] Open
Abstract
Glial cells are essential for functionality of the nervous system. Growing evidence underscores the importance of astrocytes; however, analogous astroglia in peripheral organs are poorly understood. Using confocal time-lapse imaging, fate mapping, and mutant genesis in a zebrafish model, we identify a neural crest-derived glial cell, termed nexus glia, which utilizes Meteorin signaling via Jak/Stat3 to drive differentiation and regulate heart rate and rhythm. Nexus glia are labeled with gfap, glast, and glutamine synthetase, markers that typically denote astroglia cells. Further, analysis of single-cell sequencing datasets of human and murine hearts across ages reveals astrocyte-like cells, which we confirm through a multispecies approach. We show that cardiac nexus glia at the outflow tract are critical regulators of both the sympathetic and parasympathetic system. These data establish the crucial role of glia on cardiac homeostasis and provide a description of nexus glia in the PNS.
Collapse
Affiliation(s)
- Nina L. Kikel-Coury
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Jacob P. Brandt
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Isabel A. Correia
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Michael R. O’Dea
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Dana F. DeSantis
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Felicity Sterling
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Kevin Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Gulberk Ozcebe
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J. Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
21
|
Bornhorst D, Abdelilah-Seyfried S. Strong as a Hippo's Heart: Biomechanical Hippo Signaling During Zebrafish Cardiac Development. Front Cell Dev Biol 2021; 9:731101. [PMID: 34422841 PMCID: PMC8375320 DOI: 10.3389/fcell.2021.731101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/20/2021] [Indexed: 11/13/2022] Open
Abstract
The heart is comprised of multiple tissues that contribute to its physiological functions. During development, the growth of myocardium and endocardium is coupled and morphogenetic processes within these separate tissue layers are integrated. Here, we discuss the roles of mechanosensitive Hippo signaling in growth and morphogenesis of the zebrafish heart. Hippo signaling is involved in defining numbers of cardiac progenitor cells derived from the secondary heart field, in restricting the growth of the epicardium, and in guiding trabeculation and outflow tract formation. Recent work also shows that myocardial chamber dimensions serve as a blueprint for Hippo signaling-dependent growth of the endocardium. Evidently, Hippo pathway components act at the crossroads of various signaling pathways involved in embryonic zebrafish heart development. Elucidating how biomechanical Hippo signaling guides heart morphogenesis has direct implications for our understanding of cardiac physiology and pathophysiology.
Collapse
Affiliation(s)
- Dorothee Bornhorst
- Stem Cell Program, Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA, United States.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, United States
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
22
|
Perumal E, Eswaran S, Parvin R, Balasubramanian S. Mitigation of arsenic induced developmental cardiotoxicity by ferulic acid in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2021; 244:109021. [PMID: 33631344 DOI: 10.1016/j.cbpc.2021.109021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/29/2022]
Abstract
We investigated whether ferulic acid (FA), a nutraceutical could mitigate the arsenic (As) induced cardiotoxicity. Zebrafish larvae (60 and 72 h post-fertilization [hpf]) were used to study the effect of FA on As at different time points (24 and 48 h after exposure). The FA exposure was given as pre-treatment (60 hpf) and simultaneous treatment (72 hpf) to translate the results for As contaminated areas. To accomplish this, the lethality assay was done, and based on the results, the dosage for As (1 mM) and FA (30 μM) was fixed. The FA intervention (30 μM) as 12 h pre-treatment (60 hpf) and simultaneous treatment along with As (72 hpf) decreased the As content in zebrafish larvae as evidenced by inductively coupled plasma-mass spectrometry. As exposure showed congenital deformities especially cardiac malformations in zebrafish larvae after 24 and 48 h. These teratogenic effects induced by As were reduced by FA supplementation in both groups. Also, o-dianisidine staining demonstrated that As treated larvae encountered abnormal cardiac function with reduced blood circulation, while FA supplementation reversed these effects. The acetylcholinesterase activity, a biomarker of As-induced cardiotoxicity was also found to be decreased in As group, which was rescued by FA. The modulation in the expression of the genes involved in cardiogenesis (nkx2.5, bmp2b, gata4, gata5, myh6, myl7, and tnnt2) further confirmed the ameliorative effect of FA on As induced malformations.
Collapse
Affiliation(s)
- Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| | - Sangavi Eswaran
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Reshma Parvin
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | | |
Collapse
|
23
|
Rödel CJ, Abdelilah-Seyfried S. A zebrafish toolbox for biomechanical signaling in cardiovascular development and disease. Curr Opin Hematol 2021; 28:198-207. [PMID: 33714969 DOI: 10.1097/moh.0000000000000648] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW The zebrafish embryo has emerged as a powerful model organism to investigate the mechanisms by which biophysical forces regulate vascular and cardiac cell biology during development and disease. A versatile arsenal of methods and tools is available to manipulate and analyze biomechanical signaling. This review aims to provide an overview of the experimental strategies and tools that have been utilized to study biomechanical signaling in cardiovascular developmental processes and different vascular disease models in the zebrafish embryo. Within the scope of this review, we focus on work published during the last two years. RECENT FINDINGS Genetic and pharmacological tools for the manipulation of cardiac function allow alterations of hemodynamic flow patterns in the zebrafish embryo and various types of transgenic lines are available to report endothelial cell responses to biophysical forces. These tools have not only revealed the impact of biophysical forces on cardiovascular development but also helped to establish more accurate models for cardiovascular diseases including cerebral cavernous malformations, hereditary hemorrhagic telangiectasias, arteriovenous malformations, and lymphangiopathies. SUMMARY The zebrafish embryo is a valuable vertebrate model in which in-vivo manipulations of biophysical forces due to cardiac contractility and blood flow can be performed. These analyses give important insights into biomechanical signaling pathways that control endothelial and endocardial cell behaviors. The technical advances using this vertebrate model will advance our understanding of the impact of biophysical forces in cardiovascular pathologies.
Collapse
Affiliation(s)
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Potsdam, Germany
- Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
24
|
Nguyen TH, Nguyen PD, Quetin-Leclercq J, Muller M, Ly Huong DT, Pham HT, Kestemont P. Developmental toxicity of Clerodendrum cyrtophyllum turcz ethanol extract in zebrafish embryo. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:113538. [PMID: 33144170 DOI: 10.1016/j.jep.2020.113538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/18/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Clerodendrum cyrtophyllum Turcz has been used in traditional medicine for the treatment of various diseases. In spite of its therapeutic applications, research on its toxicity and teratogenicity is still limited. AIM OF THE STUDY The study aimed to investigate the developmental toxicity of the ethanol extract of C. cyrtophyllum (EE) in zebrafish embryo model. MATERIAL AND METHODS Major compounds from crude ethanol extract of Clerodendron cyrtophyllum Turcz leaves were determined using HPLC-DAD-Orbitrap-MS analysis. The developmental toxicity of EE were investigated using zebrafish embryo model. Zebrafish embryos at 6 h post-fertilization (hpf) were treated with EE at different concentrations. Egg coagulation, mortality, hatching, yolk sac edema, pericardial edema and teratogenicity were recorded each day for during a 5-day exposure. At time point 120 hpf, body length, pericardial area, heartbeat and yolk sac area were assessed. In order to elucidate molecular mechanisms for the developmental toxicity of EE, we further evaluated the effects of the EE on the expression of genes involved on signaling pathways affecting fish embryo's development such as heart development (gata5, myl7, myh6, has2, hand2, nkx 2.5), oxidative stress (cat, sod1, gpx4, gstp2), wnt pathway (β-catenin, wnt3a, wnt5, wnt8a, wnt11), or cell apoptosis (p53, bax, bcl2, casp3, casp8, casp9, apaf-1, gadd45bb) using qRT-PCR analysis. RESULTS Our results demonstrated that three major components including acteoside, cirsilineol and cirsilineol-4'-O-β-D-glucopyranoside were identified from EE. EE exposure during 6-96 h post-fertilization (hpf) at doses ranging from 80 to 200 μg/mL increased embryo mortality and reduced hatching rate. EE exposure at 20 and 40 μg/mL until 72-120 hpf induced a series of malformations, including yolk sac edema, pericardial edema, spine deformation, shorter body length. Based on two prediction models using a teratogenic index (TI), a 25% lethality concentration (LD25) and the no observed-adverse-effect level (NOAEL), EE is considered as teratogenic for zebrafish embryos with TI (LC50/EC50) and LD25/NOAEC values at 96 hpf reaching 3.87 and 15.73 respectively. The mRNA expression levels of p53, casp8, bax/bcl2, gstp2, nkx2.5, wnt3a, wnt11, gadd45bb and gata5 were significantly upregulated by EE exposure at 20 and 40 μg/mL while the expression of wnt5, hand2 and bcl2 were downregulated. CONCLUSIONS These results provide evidence for toxicity effects of EE to embryo stages and provide an insight into the potential toxicity mechanisms on embryonic development.
Collapse
Affiliation(s)
- Thu Hang Nguyen
- Research Unit in Environmental and Evolutionary Biology (URBE), Institute of Life, Earth and Environment (ILEE), University of Namur, 5000, Namur, Belgium; Pharmacology Department, Hanoi University of Pharmacy, Ha Noi, 100000, Viet Nam.
| | - Phuc-Dam Nguyen
- Department of Chemistry Education, School of Education, Can Tho University, Can Tho City, Viet Nam; Louvain Drug Research Institute (LDRI) Pharmacognosy Research Group, Universite Catholique de Louvain, B-1200, Brussels, Belgium.
| | - Joëlle Quetin-Leclercq
- Louvain Drug Research Institute (LDRI) Pharmacognosy Research Group, Universite Catholique de Louvain, B-1200, Brussels, Belgium.
| | - Marc Muller
- Dept. Life Sciences, GIGA-R, Lab. for Organogenesis and Regeneration, University of Liege, 4000, Liège 1, Belgium.
| | | | - Hai The Pham
- Department of Mỉcrobiology and Center for Life Science Research (CELIFE), Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, 100000, Viet Nam.
| | - Patrick Kestemont
- Research Unit in Environmental and Evolutionary Biology (URBE), Institute of Life, Earth and Environment (ILEE), University of Namur, 5000, Namur, Belgium.
| |
Collapse
|
25
|
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
|
26
|
Meng Y, Zhong K, Xiao J, Huang Y, Wei Y, Tang L, Chen S, Wu J, Ma J, Cao Z, Liao X, Lu H. Exposure to pyrimethanil induces developmental toxicity and cardiotoxicity in zebrafish. CHEMOSPHERE 2020; 255:126889. [PMID: 32388256 DOI: 10.1016/j.chemosphere.2020.126889] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/21/2020] [Accepted: 04/23/2020] [Indexed: 05/27/2023]
Abstract
Pyrimethanil is a broad-spectrum fungicide commonly used in the prevention and treatment of Botrytis cinerea. However, little information is available in the literature to show the toxicity of Pyrimethanil to cardiac development. In this study, we used an experimental animal model to explore the developmental and cardiac toxicity of Pyrimethanil in aquatic vertebrates; we exposed zebrafish embryos to Pyrimethanil at concentrations of 2, 4, and 6 mg/L from 5.5 to 72 h post fertilisation. We found that Pyrimethanil caused a decrease in the hatching rate, heart rate, and survival rate of zebrafish embryos. Pyrimethanil exposure also resulted in pericardial and yolk sac edema, spinal deformity, and heart loop failure. Moreover, Pyrimethanil increased reactive oxygen stress levels and heightened the activity of superoxide dismutase and catalase. Alterations were induced in the transcription of apoptosis-related genes (p53, Bax, Bcl2, Casp 9, and Casp6l1) and heart development-related genes (Tbx2b, Gata4, Myh6, Vmhc, Nppa, Bmp2b, Bpm 4, and Bpm 10). Our data showed that the activation of Wnt signalling by BML-284 could partially rescue the malformed phenotype caused by Pyrimethanil. Our results provide new evidence for Pyrimethanil's toxicity and the danger of its residues in the environment and agricultural products.
Collapse
Affiliation(s)
- Yunlong Meng
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Keyuan Zhong
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juhua Xiao
- Department of Ultrasound, Jiangxi Provincial Maternal and Child Health Hospital, Nanchang, 330006, Jiangxi, China
| | - Yong Huang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - You Wei
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Lin Tang
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Suping Chen
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Juan Wu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Jinze Ma
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China
| | - Zigang Cao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Xinjun Liao
- Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China
| | - Huiqiang Lu
- Center for Drug Screening and Research, School of Geography and Environmental Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; College of Chemistry and Chemical Engineering, Gannan Normal University, Ganzhou, 341000, Jiangxi, China; Jiangxi Engineering Laboratory of Zebrafish Modeling and Drug Screening for Human Diseases, Ji'an, 343009, Jiangxi, China; Jiangxi Key Laboratory of Developmental Biology of Organs, Ji'an, 343009, Jiangxi, China.
| |
Collapse
|
27
|
Liu F, Fang Y, Hou X, Yan Y, Xiao H, Zuo D, Wen J, Wang L, Zhou Z, Dang X, Zhou R, Liao B. Enrichment differentiation of human induced pluripotent stem cells into sinoatrial node-like cells by combined modulation of BMP, FGF, and RA signaling pathways. Stem Cell Res Ther 2020; 11:284. [PMID: 32678003 PMCID: PMC7364513 DOI: 10.1186/s13287-020-01794-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/15/2020] [Accepted: 06/29/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Biological pacemakers derived from pluripotent stem cell (PSC) have been considered as a potential therapeutic surrogate for sick sinus syndrome. So it is essential to develop highly efficient strategies for enrichment of sinoatrial node-like cells (SANLCs) as seed cells for biological pacemakers. It has been reported that BMP, FGF, and RA signaling pathways are involved in specification of different cardiomyocyte subtypes, pacemaker, ventricular, and atrial cells. We aimed to investigate whether combined modulation of BMP, FGF, and RA signaling pathways could enrich the differentiation of SANLC from human pluripotent stem cell (hiPSC). METHODS During the differentiation process from human induced pluripotent stem cell to cardiomyocyte through small molecule-based temporal modulation of the Wnt signaling pathway, signaling of BMP, FGF, and RA was manipulated at cardiac mesoderm stage. qRT-PCR, immunofluorescence, flow cytometry, and whole cell patch clamp were used to identify the SANLC. RESULTS qRT-PCR results showed that manipulating each one of bone morphogenetic protein (BMP), fibroblast growth factor (FGF), and retinoid acid (RA) signaling was effective for the upregulation of SANLC markers. Moreover, combined modulation of these three pathways displayed the best efficiency for the expression of SANLC markers, which was further confirmed at protein level using immunofluorescence and flow cytometry. Finally, the electrophysiological characteristics of upregulated SANLC were verified by patch clamp method. CONCLUSION An efficient transgene-independent differentiation protocol for generating SANLC from hiPSC was developed, in which combined modulating BMP, FGF, and RA signaling at cardiac mesoderm stage generates SANLC at high efficiency. This may serve as a potential approach for biological pacemaker construction.
Collapse
Affiliation(s)
- Feng Liu
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Yibing Fang
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Xiaojie Hou
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Ying Yan
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Haiying Xiao
- The Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Dongchuan Zuo
- The Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Jing Wen
- The Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Linli Wang
- Guangzhou Biocare Institute of Cancer, Guangzhou, 510663, Guangdong, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Xitong Dang
- The Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China
| | - Rui Zhou
- The Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China.
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, 3-319 Zhongshan Road, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
28
|
Budine TE, de Sena-Tomás C, Williams MLK, Sepich DS, Targoff KL, Solnica-Krezel L. Gon4l/Udu regulates cardiomyocyte proliferation and maintenance of ventricular chamber identity during zebrafish development. Dev Biol 2020; 462:223-234. [PMID: 32272116 PMCID: PMC10318589 DOI: 10.1016/j.ydbio.2020.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 01/26/2020] [Accepted: 03/02/2020] [Indexed: 01/03/2023]
Abstract
Vertebrate heart development requires spatiotemporal regulation of gene expression to specify cardiomyocytes, increase the cardiomyocyte population through proliferation, and to establish and maintain atrial and ventricular cardiac chamber identities. The evolutionarily conserved chromatin factor Gon4-like (Gon4l), encoded by the zebrafish ugly duckling (udu) locus, has previously been implicated in cell proliferation, cell survival, and specification of mesoderm-derived tissues including blood and somites, but its role in heart formation has not been studied. Here we report two distinct roles of Gon4l/Udu in heart development: regulation of cell proliferation and maintenance of ventricular identity. We show that zygotic loss of udu expression causes a significant reduction in cardiomyocyte number at one day post fertilization that becomes exacerbated during later development. We present evidence that the cardiomyocyte deficiency in udu mutants results from reduced cell proliferation, unlike hematopoietic deficiencies attributed to TP53-dependent apoptosis. We also demonstrate that expression of the G1/S-phase cell cycle regulator, cyclin E2 (ccne2), is reduced in udu mutant hearts, and that the Gon4l protein associates with regulatory regions of the ccne2 gene during early embryogenesis. Furthermore, udu mutant hearts exhibit a decrease in the proportion of ventricular cardiomyocytes compared to atrial cardiomyocytes, concomitant with progressive reduction of nkx2.5 expression. We further demonstrate that udu and nkx2.5 interact to maintain the proportion of ventricular cardiomyocytes during development. However, we find that ectopic expression of nkx2.5 is not sufficient to restore ventricular chamber identity suggesting that Gon4l regulates cardiac chamber patterning via multiple pathways. Together, our findings define a novel role for zygotically-expressed Gon4l in coordinating cardiomyocyte proliferation and chamber identity maintenance during cardiac development.
Collapse
Affiliation(s)
- Terin E Budine
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carmen de Sena-Tomás
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Margot L K Williams
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Diane S Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
29
|
Wang W, Wang B, Liu Z, Xia X. Developmental toxicity and alteration of gene expression in zebrafish embryo exposed to 6-benzylaminopurine. CHEMOSPHERE 2019; 233:336-346. [PMID: 31176896 DOI: 10.1016/j.chemosphere.2019.05.261] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 06/09/2023]
Abstract
6-benzylaminopurine (6-BA) is widely used in agriculture and horticulture as plant growth regulator. Its excessive use may pose a potential risk to both environment and human health, which is causing great concern. This study was undertaken to assess the acute developmental toxicity of 6-BA to zebrafish embryos based on OECD protocols and mortality, hatching rate and malformation were investigated. Results showed that the 96 h-LC50 and 96 h- EC50 values were 63.29 mg/L and 41.86 mg/L, respectively. No mortality or teratogenic effects were found at concentrations lower than 10 mg/L 6-BA at concentrations higher than 50 mg/L significantly inhibited hatchability and embryo development, induced serious toxicity characterized by morphologic abnormalities (elongated pericardium, heart and yolk sac edema, spine curvature) and functional failure (slow spontaneous movement and heart rate, growth retardation, yolk sac absorption retention). Moreover, 6-BA-induced apoptosis was observed in embryos by the acridine orange staining and confirmed by the apoptotic-related genes, all of which p53 was significantly up-regulated at concentrations higher than 10 mg/L, bax at concentrations higher than 12.5 mg/L, while bcl2 was down-regulated at concentrations higher than 25 mg/L. As for genes of cardiac development, qPCR results demonstrated that nkx2.5, gata5, and amhc were significantly down-regulated at concentrations higher than 25 mg/L, vmhc and atp2a2a at concentration of 50 mg/L, in contrast, hand2 was up-regulated at concentration of 50 mg/L. Our data indicate that 6-BA induces a dose-dependent toxicity resulting in apoptosis through the involvement of p53-dependent pathways and hindering normal heart development in zebrafish embryos.
Collapse
Affiliation(s)
- Weixiang Wang
- School of Food and Bioengineering, Xihua University, Chengdu, PR China.
| | - Binrong Wang
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| | - Zihao Liu
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| | - Xuemei Xia
- School of Food and Bioengineering, Xihua University, Chengdu, PR China
| |
Collapse
|
30
|
Biomechanical signaling within the developing zebrafish heart attunes endocardial growth to myocardial chamber dimensions. Nat Commun 2019; 10:4113. [PMID: 31511517 PMCID: PMC6739419 DOI: 10.1038/s41467-019-12068-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022] Open
Abstract
Intra-organ communication guides morphogenetic processes that are essential for an organ to carry out complex physiological functions. In the heart, the growth of the myocardium is tightly coupled to that of the endocardium, a specialized endothelial tissue that lines its interior. Several molecular pathways have been implicated in the communication between these tissues including secreted factors, components of the extracellular matrix, or proteins involved in cell-cell communication. Yet, it is unknown how the growth of the endocardium is coordinated with that of the myocardium. Here, we show that an increased expansion of the myocardial atrial chamber volume generates higher junctional forces within endocardial cells. This leads to biomechanical signaling involving VE-cadherin, triggering nuclear localization of the Hippo pathway transcriptional regulator Yap1 and endocardial proliferation. Our work suggests that the growth of the endocardium results from myocardial chamber volume expansion and ends when the tension on the tissue is relaxed. It is unknown how endocardium growth is coordinated with that of the myocardium in the zebrafish. Here, the authors show that myocardial chamber volume expansion causes increased endocardial tissue tension, which in turn triggers Hippo signaling-mediated proliferation within the endocardium.
Collapse
|
31
|
Pawlak M, Kedzierska KZ, Migdal M, Karim AN, Ramilowski JA, Bugajski L, Hashimoto K, Marconi A, Piwocka K, Carninci P, Winata CL. Dynamics of cardiomyocyte transcriptome and chromatin landscape demarcates key events of heart development. Genome Res 2019; 29:506-519. [PMID: 30760547 PMCID: PMC6396412 DOI: 10.1101/gr.244491.118] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/09/2019] [Indexed: 12/19/2022]
Abstract
Organogenesis involves dynamic regulation of gene transcription and complex multipathway interactions. Despite our knowledge of key factors regulating various steps of heart morphogenesis, considerable challenges in understanding its mechanism still exist because little is known about their downstream targets and interactive regulatory network. To better understand transcriptional regulatory mechanism driving heart development and the consequences of its disruption in vivo, we performed time-series analyses of the transcriptome and genome-wide chromatin accessibility in isolated cardiomyocytes (CMs) from wild-type zebrafish embryos at developmental stages corresponding to heart tube morphogenesis, looping, and maturation. We identified genetic regulatory modules driving crucial events of heart development that contained key cardiac TFs and are associated with open chromatin regions enriched for DNA sequence motifs belonging to the family of the corresponding TFs. Loss of function of cardiac TFs Gata5, Tbx5a, and Hand2 affected the cardiac regulatory networks and caused global changes in chromatin accessibility profile, indicating their role in heart development. Among regions with differential chromatin accessibility in mutants were highly conserved noncoding elements that represent putative enhancers driving heart development. The most prominent gene expression changes, which correlated with chromatin accessibility modifications within their proximal promoter regions, occurred between heart tube morphogenesis and looping, and were associated with metabolic shift and hematopoietic/cardiac fate switch during CM maturation. Our results revealed the dynamic regulatory landscape throughout heart development and identified interactive molecular networks driving key events of heart morphogenesis.
Collapse
Affiliation(s)
- Michal Pawlak
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Katarzyna Z Kedzierska
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Maciej Migdal
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Abu Nahia Karim
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | | | - Lukasz Bugajski
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, 02-093 Warsaw, Poland
| | - Kosuke Hashimoto
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
| | - Aleksandra Marconi
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
| | - Katarzyna Piwocka
- Nencki Institute of Experimental Biology, Laboratory of Cytometry, 02-093 Warsaw, Poland
| | - Piero Carninci
- RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045 Japan
| | - Cecilia L Winata
- International Institute of Molecular and Cell Biology in Warsaw, Laboratory of Zebrafish Developmental Genomics, 02-109 Warsaw, Poland
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| |
Collapse
|
32
|
smarce1 mutants have a defective endocardium and an increased expression of cardiac transcription factors in zebrafish. Sci Rep 2018; 8:15369. [PMID: 30337622 PMCID: PMC6194089 DOI: 10.1038/s41598-018-33746-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
SWI/SNF or BAF chromatin-remodeling complexes are polymorphic assemblies of homologous subunit families that remodel nucleosomes and facilitate tissue-specific gene regulation during development. BAF57/SMARCE1 is a BAF complex subunit encoded in animals by a single gene and is a component of all mammalian BAF complexes. In vivo, the loss of SMARCE1 would lead to the formation of deficient combinations of the complex which might present limited remodeling activities. To address the specific contribution of SMARCE1 to the function of the BAF complex, we generated CRISPR/Cas9 mutations of smarce1 in zebrafish. Smarce1 mutants showed visible defects at 72 hpf, including smaller eyes, abnormal body curvature and heart abnormalities. Gene expression analysis revealed that the mutant embryos displayed defects in endocardial development since early stages, which led to the formation of a misshapen heart tube. The severe morphological and functional cardiac problems observed at 4 dpf were correlated with the substantially increased expression of different cardiac transcription factors. Additionally, we showed that Smarce1 binds to cis-regulatory regions of the gata5 gene and is necessary for the recruitment of the BAF complex to these regions.
Collapse
|
33
|
Colombo S, de Sena-Tomás C, George V, Werdich AA, Kapur S, MacRae CA, Targoff KL. Nkx genes establish second heart field cardiomyocyte progenitors at the arterial pole and pattern the venous pole through Isl1 repression. Development 2018; 145:dev.161497. [PMID: 29361575 DOI: 10.1242/dev.161497] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/04/2017] [Indexed: 12/28/2022]
Abstract
NKX2-5 is the most commonly mutated gene associated with human congenital heart defects (CHDs), with a predilection for cardiac pole abnormalities. This homeodomain transcription factor is a central regulator of cardiac development and is expressed in both the first and second heart fields (FHF and SHF). We have previously revealed essential functions of nkx2.5 and nkx2.7, two Nkx2-5 homologs expressed in zebrafish cardiomyocytes, in maintaining ventricular identity. However, the differential roles of these genes in the specific subpopulations of the anterior (aSHF) and posterior (pSHF) SHFs have yet to be fully defined. Here, we show that Nkx genes regulate aSHF and pSHF progenitors through independent mechanisms. We demonstrate that Nkx genes restrict proliferation of aSHF progenitors in the outflow tract, delimit the number of pSHF progenitors at the venous pole and pattern the sinoatrial node acting through Isl1 repression. Moreover, optical mapping highlights the requirement for Nkx gene dose in establishing electrophysiological chamber identity and in integrating the physiological connectivity of FHF and SHF cardiomyocytes. Ultimately, our results may shed light on the discrete errors responsible for NKX2-5-dependent human CHDs of the cardiac outflow and inflow tracts.
Collapse
Affiliation(s)
- Sophie Colombo
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Carmen de Sena-Tomás
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Vanessa George
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Andreas A Werdich
- Brigham and Women's Hospital/Harvard Medical School, Cardiovascular Division, 75 Francis Street, Thorn 11, Boston, MA 02115, USA
| | - Sunil Kapur
- Brigham and Women's Hospital/Harvard Medical School, Cardiovascular Division, 75 Francis Street, Thorn 11, Boston, MA 02115, USA
| | - Calum A MacRae
- Brigham and Women's Hospital/Harvard Medical School, Cardiovascular Division, 75 Francis Street, Thorn 11, Boston, MA 02115, USA
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
34
|
Duong TB, Ravisankar P, Song YC, Gafranek JT, Rydeen AB, Dohn TE, Barske LA, Crump JG, Waxman JS. Nr2f1a balances atrial chamber and atrioventricular canal size via BMP signaling-independent and -dependent mechanisms. Dev Biol 2017; 434:7-14. [PMID: 29157563 DOI: 10.1016/j.ydbio.2017.11.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/15/2017] [Accepted: 11/16/2017] [Indexed: 12/18/2022]
Abstract
Determination of appropriate chamber size is critical for normal vertebrate heart development. Although Nr2f transcription factors promote atrial maintenance and differentiation, how they determine atrial size remains unclear. Here, we demonstrate that zebrafish Nr2f1a is expressed in differentiating atrial cardiomyocytes. Zebrafish nr2f1a mutants have smaller atria due to a specific reduction in atrial cardiomyocyte (AC) number, suggesting it has similar requirements to Nr2f2 in mammals. Furthermore, the smaller atria in nr2f1a mutants are derived from distinct mechanisms that perturb AC differentiation at the chamber poles. At the venous pole, Nr2f1a enhances the rate of AC differentiation. Nr2f1a also establishes the atrial-atrioventricular canal (AVC) border through promoting the differentiation of mature ACs. Without Nr2f1a, AVC markers are expanded into the atrium, resulting in enlarged endocardial cushions (ECs). Inhibition of Bmp signaling can restore EC development, but not AC number, suggesting that Nr2f1a concomitantly coordinates atrial and AVC size through both Bmp-dependent and independent mechanisms. These findings provide insight into conserved functions of Nr2f proteins and the etiology of atrioventricular septal defects (AVSDs) associated with NR2F2 mutations in humans.
Collapse
Affiliation(s)
- Tiffany B Duong
- Molecular and Developmental Biology Master's Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States; The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Padmapriyadarshini Ravisankar
- The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Yuntao Charlie Song
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States; The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Jacob T Gafranek
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States; The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Ariel B Rydeen
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States; The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Tracy E Dohn
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States; The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Lindsey A Barske
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, United States
| | - Joshua S Waxman
- The Heart Institute and Molecular Cardiovascular Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Developmental Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
35
|
Hill JT, Demarest B, Gorsi B, Smith M, Yost HJ. Heart morphogenesis gene regulatory networks revealed by temporal expression analysis. Development 2017; 144:3487-3498. [PMID: 28807900 DOI: 10.1242/dev.154146] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
During embryogenesis the heart forms as a linear tube that then undergoes multiple simultaneous morphogenetic events to obtain its mature shape. To understand the gene regulatory networks (GRNs) driving this phase of heart development, during which many congenital heart disease malformations likely arise, we conducted an RNA-seq timecourse in zebrafish from 30 hpf to 72 hpf and identified 5861 genes with altered expression. We clustered the genes by temporal expression pattern, identified transcription factor binding motifs enriched in each cluster, and generated a model GRN for the major gene batteries in heart morphogenesis. This approach predicted hundreds of regulatory interactions and found batteries enriched in specific cell and tissue types, indicating that the approach can be used to narrow the search for novel genetic markers and regulatory interactions. Subsequent analyses confirmed the GRN using two mutants, Tbx5 and nkx2-5, and identified sets of duplicated zebrafish genes that do not show temporal subfunctionalization. This dataset provides an essential resource for future studies on the genetic/epigenetic pathways implicated in congenital heart defects and the mechanisms of cardiac transcriptional regulation.
Collapse
Affiliation(s)
- Jonathon T Hill
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA .,Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Bradley Demarest
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Bushra Gorsi
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - Megan Smith
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| | - H Joseph Yost
- Molecular Medicine Program, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
36
|
Harrington JK, Sorabella R, Tercek A, Isler JR, Targoff KL. Nkx2.5 is essential to establish normal heart rate variability in the zebrafish embryo. Am J Physiol Regul Integr Comp Physiol 2017; 313:R265-R271. [PMID: 28615160 DOI: 10.1152/ajpregu.00223.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/11/2022]
Abstract
Heart rate variability (HRV) has become an important clinical marker of cardiovascular health and a research measure for the study of the cardiac conduction system and its autonomic controls. While the zebrafish (Danio rerio) is an ideal vertebrate model for understanding heart development, HRV has only recently been investigated in this system. We have previously demonstrated that nkx2.5 and nkx2.7, two homologues of Nkx2-5 expressed in zebrafish cardiomyocytes, play vital roles in maintaining cardiac chamber-specific characteristics. Given observed defects in ventricular and atrial chamber identities in nkx2.5-/- embryos coupled with conduction system abnormalities in murine models of Nkx2.5 insufficiency, we postulated that reduced HRV would serve as a marker of poor cardiac health in nkx2.5 mutants and in other zebrafish models of human congenital heart disease. Using live video image acquisition, we derived beat-to-beat intervals to compare HRV in wild-type and nkx2.5-/- embryos. Our data illustrate that the nkx2.5 loss-of-function model exhibits increased heart rate and decreased HRV when compared with wild type during embryogenesis. These findings validate HRV analysis as a useful quantitative tool for assessment of cardiac health in zebrafish and underscore the importance of nkx2.5 in maintaining normal heart rate and HRV during early conduction system development.
Collapse
Affiliation(s)
- Jamie K Harrington
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Robert Sorabella
- Division of Cardiothoracic Surgery, Department of Surgery, College of Physicians and Surgeons, Columbia University, New York, New York; and
| | - Abigail Tercek
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Joseph R Isler
- Division of Neonatology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York;
| |
Collapse
|
37
|
Jung JH, Lee EH, Choi KM, Yim UH, Ha SY, An JG, Kim M. Developmental toxicity in flounder embryos exposed to crude oils derived from different geographical regions. Comp Biochem Physiol C Toxicol Pharmacol 2017; 196:19-26. [PMID: 28274761 DOI: 10.1016/j.cbpc.2017.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 01/28/2023]
Abstract
Crude oils from distinct geographical regions have distinct chemical compositions, and, as a result, their toxicity may be different. However, developmental toxicity of crude oils derived from different geographical regions has not been extensively characterized. In this study, flounder embryos were separately exposed to effluents contaminated by three crude oils including: Basrah Light (BLO), Pyrenees (PCO), and Sakhalin Vityaz (SVO), in addition to a processed fuel oil (MFO-380), to measure developmental toxicity and for gene expressions. Each oil possessed a distinct chemical composition. Edema defect was highest in embryos exposed to PCO and MFO-380 that both have a greater fraction of three-ring PAHs (33% and 22%, respectively) compared to BLO and SVO. Observed caudal fin defects were higher in embryos exposed to SVO and MFO-380, which are both dominated by naphthalenes (81% and 52%, respectively). CYP1A gene expressions were also highest in embryos exposed to SVO and MFO-380. Higher incidence of cardiotoxicity and lower nkx 2.5 expression were detected in embryos exposed to PCO. Unique gene expression profiles were observed in embryos exposed to crude oils with distinct compositions. This study demonstrates that crude oils of different geographical origins with different compositional characteristics induce developmental toxicity to different degrees.
Collapse
Affiliation(s)
- Jee-Hyun Jung
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Eun-Hee Lee
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea
| | - Kwang-Min Choi
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea
| | - Un Hyuk Yim
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Sung Yong Ha
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea
| | - Joon Geon An
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea
| | - Moonkoo Kim
- Oil and POPs Research Group, Korea Institute of Ocean Science and Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
38
|
Jung JH, Ko J, Lee EH, Choi KM, Kim M, Yim UH, Lee JS, Shim WJ. RNA seq- and DEG-based comparison of developmental toxicity in fish embryos of two species exposed to Iranian heavy crude oil. Comp Biochem Physiol C Toxicol Pharmacol 2017; 196:1-10. [PMID: 28257923 DOI: 10.1016/j.cbpc.2017.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/21/2017] [Accepted: 02/23/2017] [Indexed: 02/05/2023]
Abstract
To determine and compare the toxic effects of Iranian heavy crude oil (IHCO) on the embryonic development of two fish species, we examined transcriptome profiles using RNA-seq. The assembled contigs were 66,070 unigenes in olive flounder embryos and 76,498 unigenes in spotted seabass embryos. In the differential gene expression (DEG) profiles, olive flounder embryos showed different up- and down-regulated patterns than spotted seabass embryos in response to fresh IHCO (FIHCO) and weathered IHCO (WIHCO). In this work, we categorized DEG profiles into six pathways: ribosome, oxidative phosphorylation, Parkinson's disease, Alzheimer's disease, Huntington's disease, and cardiac muscle contraction, validating the expression patterns of 13 DEGs using real-time quantitative RT-PCR. The expression of the CYP1A, CYP1B1, and CYP1C1 genes in spotted seabass embryos was higher than in olive flounder embryos, whereas genes related to cell processing, development, and the immune system showed the opposite trend. Orthologous gene cluster analysis showed that olive flounder embryos were sensitive (fold change of genes with cutoff P<0.05) to both FIHCO and WIHCO, but spotted seabass embryos exhibited higher sensitivity to WIHCO than FIHCO, indicating that species-specific differences are likely to be reflected in population levels after oil spills. Overall, our study provides new insight on the different embryonic susceptibilities of two marine fish species to FIHCO and WIHCO and a better understanding of the underlying molecular mechanisms via RNA-seq and DEGs.
Collapse
Affiliation(s)
- Jee-Hyun Jung
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea.
| | - Junsu Ko
- Theragen Etex Bio Institute Inc., Suwon 16229, Republic of Korea
| | - Eun-Hee Lee
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea
| | - Kwang-Min Choi
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea
| | - Moonkoo Kim
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Un Hyuk Yim
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Won Joon Shim
- Oil & POPs Research Group, Korea Institute of Ocean Science & Technology, Geoje 53201, Republic of Korea; Department of Marine Environmental Science, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
39
|
Yalcin HC, Amindari A, Butcher JT, Althani A, Yacoub M. Heart function and hemodynamic analysis for zebrafish embryos. Dev Dyn 2017; 246:868-880. [PMID: 28249360 DOI: 10.1002/dvdy.24497] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/24/2017] [Accepted: 02/24/2017] [Indexed: 12/28/2022] Open
Abstract
The Zebrafish has emerged to become a powerful vertebrate animal model for cardiovascular research in recent years. Its advantages include easy genetic manipulation, transparency, small size, low cost, and the ability to survive without active circulation at early stages of development. Sequencing the whole genome and identifying ortholog genes with human genome made it possible to induce clinically relevant cardiovascular defects via genetic approaches. Heart function and disturbed hemodynamics need to be assessed in a reliable manner for these disease models in order to reveal the mechanobiology of induced defects. This effort requires precise determination of blood flow patterns as well as hemodynamic stress (i.e., wall shear stress and pressure) levels within the developing heart. While traditional approach involves time-lapse brightfield microscopy to track cell and tissue movements, in more recent studies fast light-sheet fluorescent microscopes are utilized for that purpose. Integration of more complicated techniques like particle image velocimetry and computational fluid dynamics modeling for hemodynamic analysis holds a great promise to the advancement of the Zebrafish studies. Here, we discuss the latest developments in heart function and hemodynamic analysis for Zebrafish embryos and conclude with our future perspective on dynamic analysis of the Zebrafish cardiovascular system. Developmental Dynamics 246:868-880, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Armin Amindari
- Faculty of Mechanical Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Jonathan T Butcher
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha, Qatar
| | - Magdi Yacoub
- Imperial College, NHLI, Heart Science Centre, Harefield, Middlesex, UB9 6JH, United Kingdom
| |
Collapse
|
40
|
The pro-inflammatory signalling regulator Stat4 promotes vasculogenesis of great vessels derived from endothelial precursors. Nat Commun 2017; 8:14640. [PMID: 28256502 PMCID: PMC5338034 DOI: 10.1038/ncomms14640] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 01/17/2017] [Indexed: 01/05/2023] Open
Abstract
Vasculogenic defects of great vessels (GVs) are a major cause of congenital cardiovascular diseases. However, genetic regulators of endothelial precursors in GV vasculogenesis remain largely unknown. Here we show that Stat4, a transcription factor known for its regulatory role of pro-inflammatory signalling, promotes GV vasculogenesis in zebrafish. We find stat4 transcripts highly enriched in nkx2.5+ endothelial precursors in the pharynx and demonstrate that genetic ablation of stat4 causes stenosis of pharyngeal arch arteries (PAAs) by suppressing PAAs 3–6 angioblast development. We further show that stat4 is a downstream target of nkx2.5 and that it autonomously promotes proliferation of endothelial precursors of the mesoderm. Mechanistically, stat4 regulates the emerging PAA angioblasts by inhibiting the expression of hdac3 and counteracting the effect of stat1a. Altogether, our study establishes a role for Stat4 in zebrafish great vessel development, and suggests that Stat4 may serve as a therapeutic target for GV defects. Stat4 is a transcription factor known to regulate pro-inflammatory signalling. Here, Meng et al. show that Stat4 is not only regulating inflammation but it is also crucial for great vessels development and endothelial precursor proliferation in zebrafish, by inhibiting the expression of hdac3 and counteracting the effect of Stat1a.
Collapse
|
41
|
Iklé JM, Tavares ALP, King M, Ding H, Colombo S, Firulli BA, Firulli AB, Targoff KL, Yelon D, Clouthier DE. Nkx2.5 regulates endothelin converting enzyme-1 during pharyngeal arch patterning. Genesis 2017; 55. [PMID: 28109039 DOI: 10.1002/dvg.23021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/11/2022]
Abstract
In gnathostomes, dorsoventral (D-V) patterning of neural crest cells (NCC) within the pharyngeal arches is crucial for the development of hinged jaws. One of the key signals that mediate this process is Endothelin-1 (EDN1). Loss of EDN1 binding to the Endothelin-A receptor (EDNRA) results in loss of EDNRA signaling and subsequent facial birth defects in humans, mice and zebrafish. A rate-limiting step in this crucial signaling pathway is the conversion of immature EDN1 into a mature active form by Endothelin converting enzyme-1 (ECE1). However, surprisingly little is known about how Ece1 transcription is induced or regulated. We show here that Nkx2.5 is required for proper craniofacial development in zebrafish and acts in part by upregulating ece1 expression. Disruption of nkx2.5 in zebrafish embryos results in defects in both ventral and dorsal pharyngeal arch-derived elements, with changes in ventral arch gene expression consistent with a disruption in Ednra signaling. ece1 mRNA rescues the nkx2.5 morphant phenotype, indicating that Nkx2.5 functions through modulating Ece1 expression or function. These studies illustrate a new function for Nkx2.5 in embryonic development and provide new avenues with which to pursue potential mechanisms underlying human facial disorders.
Collapse
Affiliation(s)
- Jennifer M Iklé
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| | - Andre L P Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| | - Marisol King
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| | - Hailei Ding
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| | - Sophie Colombo
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, 10032
| | - Beth A Firulli
- Departments of Anatomy and Medical, Biochemistry, and Molecular Genetics, Indiana Medical School, Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Division of Pediatric Cardiology, Indianapolis, 46202
| | - Anthony B Firulli
- Departments of Anatomy and Medical, Biochemistry, and Molecular Genetics, Indiana Medical School, Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Division of Pediatric Cardiology, Indianapolis, 46202
| | - Kimara L Targoff
- Division of Cardiology, Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, 10032
| | - Deborah Yelon
- Division of Biological Sciences, University of California, San Diego, La Jolla, California, 92093
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045
| |
Collapse
|
42
|
Grant MG, Patterson VL, Grimes DT, Burdine RD. Modeling Syndromic Congenital Heart Defects in Zebrafish. Curr Top Dev Biol 2017; 124:1-40. [DOI: 10.1016/bs.ctdb.2016.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Cardiomyocyte proliferation in zebrafish and mammals: lessons for human disease. Cell Mol Life Sci 2016; 74:1367-1378. [PMID: 27812722 PMCID: PMC5357290 DOI: 10.1007/s00018-016-2404-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/14/2016] [Accepted: 10/28/2016] [Indexed: 01/08/2023]
Abstract
Cardiomyocytes proliferate profusely during early development and for a brief period after birth in mammals. Within a month after birth, this proliferative capability is dramatically reduced in mammals unlike lower vertebrates where it persists into adult life. The zebrafish, for example, retains the ability to regenerate the apex of the heart following resection by a mechanism predominantly driven by cardiomyocyte proliferation. Differences in proliferative capacity of cardiomyocytes in adulthood between mammals and lower vertebrates are closely liked to ontogenetic or phylogenetic factors. Elucidation of these factors has the potential to provide enormous benefits if they lead to the development of therapeutic strategies that facilitate cardiomyocyte proliferation. In this review, we highlight the differences between Mammalian and Zebrafish cardiomyocytes, which could explain at least in part the different proliferative capacities in these two species. We discuss the advantages of the zebrafish as a model of cardiomyocyte proliferation, particularly at the embryonic stage. We also identify a number of key molecular pathways with potential to reveal key steps in switching cardiomyocytes from a quiescent to a proliferative phenotype.
Collapse
|
44
|
Su W, Zhu P, Wang R, Wu Q, Wang M, Zhang X, Mei L, Tang J, Kumar M, Wang X, Su L, Dong N. Congenital heart diseases and their association with the variant distribution features on susceptibility genes. Clin Genet 2016; 91:349-354. [PMID: 27426723 DOI: 10.1111/cge.12835] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 07/11/2016] [Accepted: 07/13/2016] [Indexed: 01/07/2023]
Abstract
Congenital heart disease (CHD), one of the causes of childhood morbidity and mortality, is mainly triggered by a combination of environmental and genetic factors. Several susceptible genes, such as NKX2-5, GATA4 and TBX5, have been reported as closely related to heart and vessel development. CHD subtypes are classified into diverse clinical phenotypes, such as atrial septal defects (ASD), ventricular septal defects (VSD), tetralogy of Fallot (TOF), and Holt-Oram syndrome (HOS). Here, we summarize the associations of the genetic variants in these three genes with CHD subtypes. CHD-associated variants of NKX2-5 locate mainly in the tinman domain and the homeodomain. Mutations in the homeodomain are correlated with ASD and atrioventricular (AV) block subtypes. VSD-associated variants of GATA4 are mainly at its terminal ends. Variants of TBX5 gene are primarily in exons 3, 4, 5 and 7 and highly associated with HOS subtype. Hence, the variant distribution of NKX2-5, GATA4 and TBX5 are tightly associated with particular CHD subtypes. Further structure-modelling analysis revealed that these mutated amino acid residuals maintain their DNA-binding ability and structural stability. Therefore structural features of these genes may be used to predict the high risk of CHD subtypes in infants.
Collapse
Affiliation(s)
- W Su
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - P Zhu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - R Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Q Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - M Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - X Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - L Mei
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Tang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - M Kumar
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - X Wang
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - L Su
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - N Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Abstract
Cardiac transcription factors orchestrate the complex cellular and molecular events required to produce a functioning heart. Misregulation of the cardiac transcription program leads to embryonic developmental defects and is associated with human congenital heart diseases. Recent studies have expanded our understanding of the regulation of cardiac gene expression at an additional layer, involving the coordination of epigenetic and transcriptional regulators. In this review, we highlight and discuss discoveries made possible by the genetic and embryological tools available in the zebrafish model organism, with a focus on the novel functions of cardiac transcription factors and epigenetic and transcriptional regulatory proteins during cardiogenesis.
Collapse
|
46
|
Cardiac sodium channel regulator MOG1 regulates cardiac morphogenesis and rhythm. Sci Rep 2016; 6:21538. [PMID: 26903377 PMCID: PMC4763225 DOI: 10.1038/srep21538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/26/2016] [Indexed: 01/09/2023] Open
Abstract
MOG1 was initially identified as a protein that interacts with the small GTPase Ran involved in transport of macromolecules into and out of the nucleus. In addition, we have established that MOG1 interacts with the cardiac sodium channel Nav1.5 and regulates cell surface trafficking of Nav1.5. Here we used zebrafish as a model system to study the in vivo physiological role of MOG1. Knockdown of mog1 expression in zebrafish embryos significantly decreased the heart rate (HR). Consistently, the HR increases in embryos with over-expression of human MOG1. Compared with wild type MOG1 or control EGFP, mutant MOG1 with mutation E83D associated with Brugada syndrome significantly decreases the HR. Interestingly, knockdown of mog1 resulted in abnormal cardiac looping during embryogenesis. Mechanistically, knockdown of mog1 decreases expression of hcn4 involved in the regulation of the HR, and reduces expression of nkx2.5, gata4 and hand2 involved in cardiac morphogenesis. These data for the first time revealed a novel role that MOG1, a nucleocytoplasmic transport protein, plays in cardiac physiology and development.
Collapse
|
47
|
Curcumin-mediated cardiac defects in mouse is associated with a reduced histone H3 acetylation and reduced expression of cardiac transcription factors. Cardiovasc Toxicol 2015; 14:162-9. [PMID: 24323078 DOI: 10.1007/s12012-013-9240-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Histone acetylation plays an important role in heart development. However, the mechanism(s) remains unclear. This study was designed to evaluate the effect of curcumin-caused histone hypo-acetylation on the development of mouse embryonic heart and the expression of cardiac transcription factors in vivo. The results showed that curcumin treatment significantly decreased histone acetylase activity and histone acetylation level in mouse embryonic heart. In curcumin-treated mice, the hearts on E11.5 were smaller with thinner ventricular wall and a delayed development of trabeculae and ventricular septum compared with the controls. The ventricular septum was complete on E14.5; however, the ventricular wall and septum were thinner with fewer trabeculae than those in the controls. On E17.5, the cardiac structure appeared normal, but the ventricular wall and septum were thinner. The expression of GATA4, Nkx2.5 and Mef2c in the heart on E11.5 and E14.5 was decreased significantly as compared to the controls. There was no significant difference in Mef2c expression on E17.5 between curcumin-treated group and the controls, while GATA4 and Nkx2.5 expression remained significantly reduced. These results indicate that inhibition of histone acetylation by curcumin can reduce the expression of the cardiac transcription factors resulting in an abnormal heart development in mice.
Collapse
|
48
|
George V, Colombo S, Targoff KL. An early requirement for nkx2.5 ensures the first and second heart field ventricular identity and cardiac function into adulthood. Dev Biol 2014; 400:10-22. [PMID: 25536398 DOI: 10.1016/j.ydbio.2014.12.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 12/02/2014] [Accepted: 12/16/2014] [Indexed: 10/24/2022]
Abstract
Temporally controlled mechanisms that define the unique features of ventricular and atrial cardiomyocyte identities are essential for the construction of a coordinated, morphologically intact heart. We have previously demonstrated an important role for nkx genes in maintaining ventricular identity, however, the specific timing of nkx2.5 function in distinct cardiomyocyte populations has yet to be elucidated. Here, we show that heat-shock induction of a novel transgenic line, Tg(hsp70l:nkx2.5-EGFP), during the initial stages of cardiomyocyte differentiation leads to rescue of chamber shape and identity in nkx2.5(-/-) embryos as chambers emerge. Intriguingly, our findings link an early role of this essential cardiac transcription factor with a later function. Moreover, these data reveal that nkx2.5 is also required in the second heart field as the heart tube forms, reflecting the temporal delay in differentiation of this population. Thus, our results support a model in which nkx genes induce downstream targets that are necessary to maintain chamber-specific identity in both early- and late-differentiating cardiomyocytes at discrete stages in cardiac morphogenesis. Furthermore, we show that overexpression of nkx2.5 during the first and second heart field development not only rescues the mutant phenotype, but also is sufficient for proper function of the adult heart. Taken together, these results shed new light on the stage-dependent mechanisms that sculpt chamber-specific cardiomyocytes and, therefore, have the potential to improve in vitro generation of ventricular cells to treat myocardial infarction and congenital heart disease.
Collapse
Affiliation(s)
- Vanessa George
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032 USA
| | - Sophie Colombo
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032 USA
| | - Kimara L Targoff
- Division of Pediatric Cardiology, Department of Pediatrics, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032 USA.
| |
Collapse
|
49
|
Schupp MO, Waas M, Chun CZ, Ramchandran R. Transcriptional inhibition of etv2 expression is essential for embryonic cardiac development. Dev Biol 2014; 393:71-83. [PMID: 24984259 PMCID: PMC4137469 DOI: 10.1016/j.ydbio.2014.06.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 04/08/2014] [Accepted: 06/18/2014] [Indexed: 10/25/2022]
Abstract
E-twenty six variant 2 (Etv2) transcription factor participates in cardiac, vascular-endothelial and blood cell lineage specification decisions during embryonic development. Previous studies have identified genomic elements in the etv2 locus responsible for vascular endothelial cell specification. Using transgenic analysis in zebrafish, we report here an etv2 proximal promoter fragment that prevents transgene misexpression in myocardial progenitor cells. This inhibition of etv2 expression in the cardiac progenitor population is partly mediated by Scl and Nkx2.5, likely through direct binding to the etv2 promoter, and cis-regulatory elements located in the first and second introns. The results identify an etv2 cis-regulatory mechanism controlling cardiovascular fate choice implying that etv2 participates in a transcriptional network mediating developmental plasticity of endothelial progenitor cells during embryonic development.
Collapse
Affiliation(s)
- Marcus-Oliver Schupp
- Medical College of Wisconsin, Department of Pediatrics, CRI Developmental Vascular Biology Program, Translational and Biomedical Research Center, CRI C3420, 8701 Watertown Plank Road, P.O. Box 26509, Milwaukee, WI 53226, USA
| | - Matthew Waas
- Division of Nephrology, Hypertension and Renal Transplantation, Room CG-98, 1600 Archer Road, University of Florida, Gainesville, FL 32610, USA
| | - Chang-Zoon Chun
- Medical College of Wisconsin, Department of Pediatrics, CRI Developmental Vascular Biology Program, Translational and Biomedical Research Center, CRI C3420, 8701 Watertown Plank Road, P.O. Box 26509, Milwaukee, WI 53226, USA; Division of Nephrology, Hypertension and Renal Transplantation, Room CG-98, 1600 Archer Road, University of Florida, Gainesville, FL 32610, USA
| | - Ramani Ramchandran
- Medical College of Wisconsin, Department of Pediatrics, CRI Developmental Vascular Biology Program, Translational and Biomedical Research Center, CRI C3420, 8701 Watertown Plank Road, P.O. Box 26509, Milwaukee, WI 53226, USA.
| |
Collapse
|
50
|
Han Y, Zhang JP, Qian JQ, Hu CQ. Cardiotoxicity evaluation of anthracyclines in zebrafish (Danio rerio). J Appl Toxicol 2014; 35:241-52. [PMID: 24853142 DOI: 10.1002/jat.3007] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 02/10/2014] [Indexed: 12/14/2022]
Abstract
Drug-induced cardiotoxicity is a leading factor for drug withdrawals, and limits drug efficacy and clinical use. Therefore, new alternative animal models and methods for drug safety evaluation have been given great attention. Anthracyclines (ANTs) are widely prescribed anticancer agents that have a cumulative dose relationship with cardiotoxicity. We performed experiments to study the toxicity of ANTs in early developing zebrafish embryos, especially their effects on the heart. LC50 values for daunorubicin, pirarubicin, doxorubicin (DOX), epirubicin and DOX-liposome at 72 h post-fertilization were 122.7 μM, 111.9 μM, 31.2 μM, 108.3 μM and 55.8 μM, respectively. At the same time, zebrafish embryos were exposed to ANTs in three exposure stages and induced incomplete looping of the heart tube, pericardia edema and bradycardia in a dose-dependent manner, eventually leading to death. DOX caused the greatest heart defects in the treatment stages and its liposome reduced the effects on the heart, while daunorubicin produced the least toxicity. Genes and proteins related to heart development were also identified to be sensitive to ANT exposure and downregulated by ANTs. It revealed ANTs could disturb the heart formation and development. ANTs induced cardiotoxicity in zebrafish has similar effects in mammalian models, indicating that zebrafish may have a potential value for assessment of drug-induced developmental cardiotoxicity.
Collapse
Affiliation(s)
- Ying Han
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | | | | | | |
Collapse
|