1
|
Burggren W, Abramova R, Bautista NM, Fritsche Danielson R, Dubansky B, Gupta A, Hansson K, Iyer N, Jagadeeswaran P, Jennbacken K, Rydén-Markinhutha K, Patel V, Raman R, Trivedi H, Vazquez Roman K, Williams S, Wang QD. A larval zebrafish model of cardiac physiological recovery following cardiac arrest and myocardial hypoxic damage. Biol Open 2024; 13:bio060230. [PMID: 39263862 DOI: 10.1242/bio.060230] [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: 11/15/2023] [Accepted: 07/22/2024] [Indexed: 09/13/2024] Open
Abstract
Contemporary cardiac injury models in zebrafish larvae include cryoinjury, laser ablation, pharmacological treatment and cardiac dysfunction mutations. Although effective in damaging cardiomyocytes, these models lack the important element of myocardial hypoxia, which induces critical molecular cascades within cardiac muscle. We have developed a novel, tractable, high throughput in vivo model of hypoxia-induced cardiac damage that can subsequently be used in screening cardioactive drugs and testing recovery therapies. Our potentially more realistic model for studying cardiac arrest and recovery involves larval zebrafish (Danio rerio) acutely exposed to severe hypoxia (PO2=5-7 mmHg). Such exposure induces loss of mobility quickly followed by cardiac arrest occurring within 120 min in 5 days post fertilization (dpf) and within 40 min at 10 dpf. Approximately 90% of 5 dpf larvae survive acute hypoxic exposure, but survival fell to 30% by 10 dpf. Upon return to air-saturated water, only a subset of larvae resumed heartbeat, occurring within 4 min (5 dpf) and 6-8 min (8-10 dpf). Heart rate, stroke volume and cardiac output in control larvae before hypoxic exposure were 188±5 bpm, 0.20±0.001 nL and 35.5±2.2 nL/min (n=35), respectively. After briefly falling to zero upon severe hypoxic exposure, heart rate returned to control values by 24 h of recovery. However, reflecting the severe cardiac damage induced by the hypoxic episode, stroke volume and cardiac output remained depressed by ∼50% from control values at 24 h of recovery, and full restoration of cardiac function ultimately required 72 h post-cardiac arrest. Immunohistological staining showed co-localization of Troponin C (identifying cardiomyocytes) and Capase-3 (identifying cellular apoptosis). As an alternative to models employing mechanical or pharmacological damage to the developing myocardium, the highly reproducible cardiac effects of acute hypoxia-induced cardiac arrest in the larval zebrafish represent an alternative, potentially more realistic model that mimics the cellular and molecular consequences of an infarction for studying cardiac tissue hypoxia injury and recovery of function.
Collapse
Affiliation(s)
- Warren Burggren
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Regina Abramova
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Naim M Bautista
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Regina Fritsche Danielson
- SVP and head of Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 50, Sweden
| | - Ben Dubansky
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Avi Gupta
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Kenny Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 50, Sweden
| | - Neha Iyer
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Pudur Jagadeeswaran
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 50, Sweden
| | - Katarina Rydén-Markinhutha
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 50, Sweden
| | - Vishal Patel
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Revathi Raman
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Hersh Trivedi
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Karem Vazquez Roman
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Steven Williams
- Developmental Integrative Biology Research Group, Department of Biological Sciences, University of North Texas, Denton, TX 76205, USA
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 50, Sweden
| |
Collapse
|
2
|
Aydemir U, Mousa AH, Dicko C, Strakosas X, Shameem MA, Hellman K, Yadav AS, Ekström P, Hughes D, Ek F, Berggren M, Arner A, Hjort M, Olsson R. In situ assembly of an injectable cardiac stimulator. Nat Commun 2024; 15:6774. [PMID: 39117721 PMCID: PMC11310494 DOI: 10.1038/s41467-024-51111-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
Without intervention, cardiac arrhythmias pose a risk of fatality. However, timely intervention can be challenging in environments where transporting a large, heavy defibrillator is impractical, or emergency surgery to implant cardiac stimulation devices is not feasible. Here, we introduce an injectable cardiac stimulator, a syringe loaded with a nanoparticle solution comprising a conductive polymer and a monomer that, upon injection, forms a conductive structure around the heart for cardiac stimulation. Following treatment, the electrode is cleared from the body, eliminating the need for surgical extraction. The mixture adheres to the beating heart in vivo without disrupting its normal rhythm. The electrofunctionalized injectable cardiac stimulator demonstrates a tissue-compatible Young's modulus of 21 kPa and a high conductivity of 55 S/cm. The injected electrode facilitates electrocardiogram measurements, regulates heartbeat in vivo, and rectifies arrhythmia. Conductive functionality is maintained for five consecutive days, and no toxicity is observed at the organism, organ, or cellular levels.
Collapse
Affiliation(s)
- Umut Aydemir
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Abdelrazek H Mousa
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Cedric Dicko
- Pure and Applied Biochemistry, Department of Chemistry, Lund University, Lund, Sweden
| | - Xenofon Strakosas
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Muhammad Anwar Shameem
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Karin Hellman
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Amit Singh Yadav
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Peter Ekström
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Damien Hughes
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Magnus Berggren
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, Sweden
| | - Anders Arner
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Martin Hjort
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Roger Olsson
- Chemical Biology & Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden.
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
3
|
Habjan E, Schouten GK, Speer A, van Ulsen P, Bitter W. Diving into drug-screening: zebrafish embryos as an in vivo platform for antimicrobial drug discovery and assessment. FEMS Microbiol Rev 2024; 48:fuae011. [PMID: 38684467 PMCID: PMC11078164 DOI: 10.1093/femsre/fuae011] [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: 11/01/2023] [Revised: 02/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
The rise of multidrug-resistant bacteria underlines the need for innovative treatments, yet the introduction of new drugs has stagnated despite numerous antimicrobial discoveries. A major hurdle is a poor correlation between promising in vitro data and in vivo efficacy in animal models, which is essential for clinical development. Early in vivo testing is hindered by the expense and complexity of existing animal models. Therefore, there is a pressing need for cost-effective, rapid preclinical models with high translational value. To overcome these challenges, zebrafish embryos have emerged as an attractive model for infectious disease studies, offering advantages such as ethical alignment, rapid development, ease of maintenance, and genetic manipulability. The zebrafish embryo infection model, involving microinjection or immersion of pathogens and potential antibiotic hit compounds, provides a promising solution for early-stage drug screening. It offers a cost-effective and rapid means of assessing the efficacy, toxicity and mechanism of action of compounds in a whole-organism context. This review discusses the experimental design of this model, but also its benefits and challenges. Additionally, it highlights recently identified compounds in the zebrafish embryo infection model and discusses the relevance of the model in predicting the compound's clinical potential.
Collapse
Affiliation(s)
- Eva Habjan
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Gina K Schouten
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Alexander Speer
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Peter van Ulsen
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | - Wilbert Bitter
- Department of Medical Microbiology and Infection Control, Amsterdam UMC, Location VU Medical Center,De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
- Section Molecular Microbiology of A-LIFE, Vrije Universiteit, De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
4
|
Sebastian WA, Inoue M, Shimizu N, Sato R, Oguri S, Itonaga T, Kishimoto S, Shiraishi H, Hanada T, Ihara K. Cardiac manifestations of human ACTA2 variants recapitulated in a zebrafish model. J Hum Genet 2024; 69:133-138. [PMID: 38316882 DOI: 10.1038/s10038-024-01221-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/31/2023] [Accepted: 01/19/2024] [Indexed: 02/07/2024]
Abstract
The ACTA2 gene encodes actin α2, a major smooth muscle protein in vascular smooth muscle cells. Missense variants in the ACTA2 gene can cause inherited thoracic aortic diseases with characteristic symptoms, such as dysfunction of smooth muscle cells in the lungs, brain vessels, intestines, pupils, bladder, or heart. We identified a heterozygous missense variant of Gly148Arg (G148R) in a patient with a thoracic aortic aneurysm, dissection, and left ventricular non-compaction. We used zebrafish as an in vivo model to investigate whether or not the variants might cause functional or histopathological abnormalities in the heart. Following the fertilization of one-cell stage embryos, we injected in vitro synthesized ACTA2 mRNA of wild-type, novel variant G148R, or the previously known pathogenic variant Arg179His (R179H). The embryos were maintained and raised for 72 h post-fertilization for a heart analysis. Shortening fractions of heart were significantly reduced in both pathogenic variants. A histopathological evaluation showed that the myocardial wall of ACTA2 pathogenic variants was thinner than that of the wild type, and the total cell number within the myocardium was markedly decreased in all zebrafish with pathogenic variants mRNAs. Proliferating cell numbers were also significantly decreased in the endothelial and myocardial regions of zebrafish with ACTA2 variants compared to the wild type. These results demonstrate the effects of ACTA2 G148R and R179H on the development of left ventricle non-compaction and cardiac morphological abnormalities. Our study highlights the previously unknown significance of the ACTA2 gene in several aspects of cardiovascular development.
Collapse
Affiliation(s)
| | - Masanori Inoue
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Nobuyuki Shimizu
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Ryosuke Sato
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Saori Oguri
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Tomoyo Itonaga
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Shintaro Kishimoto
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan
| | - Hiroshi Shiraishi
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan
| | - Toshikatsu Hanada
- Department of Cell Biology, Oita University, Faculty of Medicine, Oita, Japan.
| | - Kenji Ihara
- Department of Pediatrics, Oita University, Faculty of Medicine, Oita, Japan.
| |
Collapse
|
5
|
Ribeiro J, Araújo-Silva H, Fernandes M, da Silva JA, Pinto FDCL, Pessoa ODL, Santos HS, de Menezes JESA, Gomes AC. Petrosamine isolated from marine sponge Petrosia sp. demonstrates protection against neurotoxicity in vitro and in vivo. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:16. [PMID: 38383833 PMCID: PMC10881933 DOI: 10.1007/s13659-024-00439-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024]
Abstract
According to The World Alzheimer Report 2023 by Alzheimer's Disease International (ADI) estimates that 33 to 38.5 million people worldwide suffer from Alzheimer's Disease (AD). A crucial hallmark associated with this disease is associated with the deficiency of the brain neurotransmitter acetylcholine, due to an affected acetylcholinesterase (AChE) activity. Marine organisms synthesize several classes of compounds, some of which exhibit significant AChE inhibition, such as petrosamine, a coloured pyridoacridine alkaloid. The aim of this work was to characterize the activity of petrosamine isolated for the first time from a Brazilian marine sponge, using two neurotoxicity models with aluminium chloride, as exposure to aluminium is associated with the development of neurodegenerative diseases. The in vitro model was based in a neuroblastoma cell line and the in vivo model exploited the potential of zebrafish (Danio rerio) embryos in mimicking hallmarks of AD. To our knowledge, this is the first report on petrosamine's activity over these parameters, either in vitro or in vivo, in order to characterize its full potential for tackling neurotoxicity.
Collapse
Affiliation(s)
- Joana Ribeiro
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Henrique Araújo-Silva
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Mário Fernandes
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
| | - Joilna Alves da Silva
- Program in Natural Sciences, Natural Products Chemistry Laboratory, State University of Ceará, Fortaleza, Ceará, Brazil
| | - Francisco das Chagas L Pinto
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Otília Deusdenia L Pessoa
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Hélcio Silva Santos
- Program in Natural Sciences, Natural Products Chemistry Laboratory, State University of Ceará, Fortaleza, Ceará, Brazil
- Department of Organic and Inorganic Chemistry, Science Center, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Andreia C Gomes
- CBMA (Centre of Molecular and Environmental Biology) / Aquatic Research Network (ARNET) Associate Laboratory, Department of Biology, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
6
|
Fan YL, Hsu FR, Wang Y, Liao LD. Unlocking the Potential of Zebrafish Research with Artificial Intelligence: Advancements in Tracking, Processing, and Visualization. Med Biol Eng Comput 2023; 61:2797-2814. [PMID: 37558927 DOI: 10.1007/s11517-023-02903-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Zebrafish have become a widely accepted model organism for biomedical research due to their strong cortisol stress response, behavioral strain differences, and sensitivity to both drug treatments and predators. However, experimental zebrafish studies generate substantial data that must be analyzed through objective, accurate, and repeatable analysis methods. Recently, advancements in artificial intelligence (AI) have enabled automated tracking, image recognition, and data analysis, leading to more efficient and insightful investigations. In this review, we examine key AI applications in zebrafish research, including behavior analysis, genomics, and neuroscience. With the development of deep learning technology, AI algorithms have been used to precisely analyze and identify images of zebrafish, enabling automated testing and analysis. By applying AI algorithms in genomics research, researchers have elucidated the relationship between genes and biology, providing a better basis for the development of disease treatments and gene therapies. Additionally, the development of more effective neuroscience tools could help researchers better understand the complex neural networks in the zebrafish brain. In the future, further advancements in AI technology are expected to enable more extensive and in-depth medical research applications in zebrafish, improving our understanding of this important animal model. This review highlights the potential of AI technology in achieving the full potential of zebrafish research by enabling researchers to efficiently track, process, and visualize the outcomes of their experiments.
Collapse
Affiliation(s)
- Yi-Ling Fan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Fang-Rong Hsu
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Yuhling Wang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Electrical Engineering, National United University, 2, Lien-Da, Nan-Shih Li, Miaoli, 360302, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan.
| |
Collapse
|
7
|
Saputra F, Lai YH, Roldan MJM, Alos HC, Aventurado CA, Vasquez RD, Hsiao CD. The Effect of the Pyrethroid Pesticide Fenpropathrin on the Cardiac Performance of Zebrafish and the Potential Mechanism of Toxicity. BIOLOGY 2023; 12:1214. [PMID: 37759613 PMCID: PMC10525504 DOI: 10.3390/biology12091214] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/04/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
Fenpropathrin, a pyrethroid insecticide, has been widely used for many years in agricultural fields. It works by disturbing the voltage-gated sodium channel, leading to paralysis and the death of the target animal. While past studies have focused on neurodegeneration following fenpropathrin poisoning in humans, relatively few pieces of research have examined its effect on other peripheral organs. This study successfully investigated the potential toxicity of fenpropathrin on the cardiovascular system using zebrafish as an animal model. Zebrafish larvae exposed to varying doses of fenpropathrin underwent an evaluation of cardiac physiology by measuring the heart rate, stroke volume, cardiac output, and shortening fraction. The blood flow velocity and the dorsal aorta diameter were also measured to assess the impact of fenpropathrin exposure on the vascular system. Furthermore, molecular docking was performed to evaluate the pesticide binding affinity to various proteins associated with the cardiovascular system, revealing the potential mechanism of the fenpropathrin cardiotoxic effect. The findings demonstrated a significant dose-dependent increase in the heart rate stroke volume, cardiac output, shortening fraction, and ejection fraction of zebrafish larvae after 24 h of acute treatment with fenpropathrin. Additionally, zebrafish treated at a concentration of 1 ppm exhibited significantly larger blood vessels in diameter and an increased blood flow velocity compared to the control group. According to molecular docking, fenpropathrin showed a high affinity for various voltage-gated sodium channels like scn1lab, cacna1sb, and clcn3. Finally, from the results, we found that fenpropathrin caused cardiomegaly, which may have been induced by the voltage-gated sodium channel disruption. This study highlights the significant disruption of fenpropathrin in the cardiovascular system and emphasizes the need for further research on the health implications of this pesticide.
Collapse
Affiliation(s)
- Ferry Saputra
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan;
| | - Marri Jmelou M. Roldan
- The Graduate School, University of Santo Tomas, Manila 1008, Philippines; (M.J.M.R.); (H.C.A.); (C.A.A.)
| | - Honeymae C. Alos
- The Graduate School, University of Santo Tomas, Manila 1008, Philippines; (M.J.M.R.); (H.C.A.); (C.A.A.)
| | - Charlaine A. Aventurado
- The Graduate School, University of Santo Tomas, Manila 1008, Philippines; (M.J.M.R.); (H.C.A.); (C.A.A.)
| | - Ross D. Vasquez
- The Graduate School, University of Santo Tomas, Manila 1008, Philippines; (M.J.M.R.); (H.C.A.); (C.A.A.)
- Department of Pharmacy, Faculty of Pharmacy, University of Santo Tomas, Manila 1008, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila 1008, Philippines
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan;
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| |
Collapse
|
8
|
Fujita H, Kaneshiro J, Takeda M, Sasaki K, Yamamoto R, Umetsu D, Kuranaga E, Higo S, Kondo T, Asano Y, Sakata Y, Miyagawa S, Watanabe TM. Estimation of crossbridge-state during cardiomyocyte beating using second harmonic generation. Life Sci Alliance 2023; 6:e202302070. [PMID: 37236659 PMCID: PMC10215972 DOI: 10.26508/lsa.202302070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Estimation of dynamic change of crossbridge formation in living cardiomyocytes is expected to provide crucial information for elucidating cardiomyopathy mechanisms, efficacy of an intervention, and others. Here, we established an assay system to dynamically measure second harmonic generation (SHG) anisotropy derived from myosin filaments depended on their crossbridge status in pulsating cardiomyocytes. Experiments utilizing an inheritable mutation that induces excessive myosin-actin interactions revealed that the correlation between sarcomere length and SHG anisotropy represents crossbridge formation ratio during pulsation. Furthermore, the present method found that ultraviolet irradiation induced an increased population of attached crossbridges that lost the force-generating ability upon myocardial differentiation. Taking an advantage of infrared two-photon excitation in SHG microscopy, myocardial dysfunction could be intravitally evaluated in a Drosophila disease model. Thus, we successfully demonstrated the applicability and effectiveness of the present method to evaluate the actomyosin activity of a drug or genetic defect on cardiomyocytes. Because genomic inspection alone may not catch the risk of cardiomyopathy in some cases, our study demonstrated herein would be of help in the risk assessment of future heart failure.
Collapse
Affiliation(s)
- Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Junichi Kaneshiro
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Maki Takeda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kensuke Sasaki
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Rikako Yamamoto
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Shuichiro Higo
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takumi Kondo
- Department of Medical Therapeutics for Heart Failure, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomonobu M Watanabe
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
- Laboratory for Comprehensive Bioimaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
9
|
Chang Y, Tsai JF, Chen PJ, Huang YT, Liu BH. Thallium exposure interfered with heart development in embryonic zebrafish (Danio rerio): From phenotype to genotype. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 878:162901. [PMID: 36948317 DOI: 10.1016/j.scitotenv.2023.162901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/11/2023] [Accepted: 03/12/2023] [Indexed: 05/13/2023]
Abstract
Thallium (Tl) is a rare trace metal element but increasingly detected in wastewater produced by coal-burning, smelting, and more recently, high-tech manufacturing industries. However, the adverse effects of Tl, especially cardiotoxicity, on aquatic biota remain unclear. In this study, zebrafish model was used to elucidate the effects and mechanisms of Tl(I) cardiotoxicity in developing embryos. Exposure of embryonic zebrafish to low-dose Tl(I) (25-100 μg/L) decreased heart rate and blood flow activity, and subsequently impaired swim bladder inflation and locomotive behavior of larvae. Following high-level Tl(I) administration (200-800 μg/L), embryonic zebrafish exhibited pericardial edema, incorrect heart looping, and thinner myocardial layer. Based on RNA-sequencing, Tl(I) altered pathways responsible for protein folding and transmembrane transport, as well as negative regulation of heart rate and cardiac jelly development. The gene expression of nppa, nppb, ucp1, and ucp3, biomarkers of cardiac damage, were significantly upregulated by Tl(I). Our findings demonstrate that Tl(I) at environmentally relevant concentrations interfered with cardiac development with respect to anatomy, function, and transcriptomic alterations. The cardiotoxic mechanisms of Tl(I) provide valuable information in the assessment of Tl-related ecological risk in freshwater environment.
Collapse
Affiliation(s)
- Yung Chang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jui-Feng Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Jen Chen
- Department of Agricultural Chemistry, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan
| | - Ying-Tzu Huang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Biing-Hui Liu
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
10
|
Kämmer N, Reimann T, Ovcharova V, Braunbeck T. A novel automated method for the simultaneous detection of breathing frequency and amplitude in zebrafish (Danio rerio) embryos and larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 258:106493. [PMID: 36963131 DOI: 10.1016/j.aquatox.2023.106493] [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: 01/21/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 06/18/2023]
Abstract
Stress responses of fish to disruption of oxygen homeostasis include adjusted oxygen consumption rate (MO2) as well as the hyperventilation consisting of changes in breathing frequency (fv) and amplitude (fampl). However, studying the HVR in very small organisms such as zebrafish (Danio rerio) embryos and larvae is challenging, and breathing movements (i.e., fv) are usually manually counted, which is time- and human resource-intense, error-prone and does not provide information on the amplitude of breathing movements of the response, the breathing amplitude (fampl). Hence, in the present study, a new automated method was developed to simultaneously measure fv and fampl in small zebrafish embryos and larvae with the computer software DanioScope™. To compare HVR strategies at different life-stages of zebrafish and the physiologically linked MO2, hatched 4 d old embryos and early gill-breathing 12 d old larvae were treated with the HVR-inducing neurotoxic compound lindane (γ-hexachlorocyclohexane; γ-HCH) as a model substance. Comparison of manually counted fv with fv data measured by DanioScope™ at both life-stages showed high to moderate agreement between the two methods with respect to fv in control fish and in fish treated with lower lindane concentrations (3 - 18% deviation at 25 µg/L γ-HCH). With increasing lindane concentrations (100 and 400 µg/L γ-HCH), however, manual counts showed an average underestimation of fv by up to 30%, mainly due to very fast, rapidly successive, and indistinct movements of the fish, which cannot be properly detected by manual counts. Automated measurement thus proved significantly more sensitive, although several pre- and post-processing steps are needed. The improved automated detection of fv and the first reliable estimation of fampl in small fish embryos and larvae, as well as the inclusion of MO2, may provide new insights into different respiratory strategies and may, thus, represent a tool to lower the detection limit for reactions of different life-stages of fish to environmental stressors. In the present study, this became evident, as early gill-breathing 12 d old zebrafish larvae showed symptoms of respiratory failure (i.e., increase in fv, fampl and MO2, followed by subsequent lethargy) after exposure to lindane, whereas skin-breathing in 4 d old embryos proved mainly insensitive to the paralytic effects of lindane.
Collapse
Affiliation(s)
- Nadine Kämmer
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg D-69210, Germany.
| | - Tanja Reimann
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg D-69210, Germany
| | - Viktoriia Ovcharova
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg D-69210, Germany
| | - Thomas Braunbeck
- Aquatic Ecology and Toxicology Section, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, Heidelberg D-69210, Germany.
| |
Collapse
|
11
|
de Araujo-Neto JH, Guedes APM, Leite CM, Moraes CAF, Santos AL, Brito RDS, Rocha TL, Mello-Andrade F, Ellena J, Batista AA. "Half-Sandwich" Ruthenium Complexes with Alizarin as Anticancer Agents: In Vitro and In Vivo Studies. Inorg Chem 2023; 62:6955-6969. [PMID: 37099760 DOI: 10.1021/acs.inorgchem.3c00183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Upon exploration of the chemistry of the combination of ruthenium/arene with anthraquinone alizarin (L), three new complexes with the general formulas [Ru(L)Cl(η6-p-cymene)] (C1), [Ru(L)(η6-p-cymene)(PPh3)]PF6 (C2), and [Ru(L)(η6-p-cymene)(PEt3)]PF6 (C3) were synthesized and characterized using spectroscopic techniques (mass, IR, and 1D and 2D NMR), molar conductivity, elemental analysis, and X-ray diffraction. Complex C1 exhibited fluorescence, such as free alizarin, while in C2 and C3, the emission was probably quenched by monophosphines and the crystallographic data showed that hydrophobic interactions are predominant in intermolecular contacts. The cytotoxicity of the complexes was evaluated in the MDA-MB-231 (triple-negative breast cancer), MCF-7 (breast cancer), and A549 (lung) tumor cell lines and MCF-10A (breast) and MRC-5 (lung) nontumor cell lines. Complexes C1 and C2 were more selective to the breast tumor cell lines, and C2 was the most cytotoxic (IC50 = 6.5 μM for MDA-MB-231). In addition, compound C1 performs a covalent interaction with DNA, while C2 and C3 present only weak interactions; however, internalization studies by flow cytometry and confocal microscopy showed that complex C1 does not accumulate in viable MDA-MB-231 cells and is detected in the cytoplasm only after cell permeabilization. Investigations of the mechanism of action of the complexes indicate that C2 promotes cell cycle arrest in the Sub-G1 phase in MDA-MB-231, inhibits its colony formation, and has a possible antimetastatic action, impeding cell migration in the wound-healing experiment (13% of wound healing in 24 h). The in vivo toxicological experiments with zebrafish indicate that C1 and C3 exhibit the most zebrafish embryo developmental toxicity (inhibition of spontaneous movements and heartbeats), while C2, the most promising anticancer drug in the in vitro preclinical tests, revealed the lowest toxicity in in vivo preclinical screening.
Collapse
Affiliation(s)
- João Honorato de Araujo-Neto
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Adriana P M Guedes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Celisnolia M Leite
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Carlos André F Moraes
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| | - Andressa L Santos
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Rafaella da S Brito
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Thiago L Rocha
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
| | - Francyelli Mello-Andrade
- Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Goiás 74605-050, Brazil
- Instituto Federal de Educação Ciência e Tecnologia (IFG), Goiânia, Goiás 74055-110, Brazil
| | - Javier Ellena
- Instituto de Física de São Carlos, Universidade de São Paulo (USP), São Carlos, São Paulo 13566-590, Brazil
| | - Alzir A Batista
- Departamento de Química, Universidade Federal de São Carlos, São Carlos, São Paulo 13565-905, Brazil
| |
Collapse
|
12
|
Lin HC, Saputra F, Audira G, Lai YH, Roldan MJM, Alos HC, Aventurado CA, Vasquez RD, Tsai GJ, Lim KH, Hsiao CD. Investigating Potential Cardiovascular Toxicity of Two Anti-Leukemia Drugs of Asciminib and Ponatinib in Zebrafish Embryos. Int J Mol Sci 2022; 23:ijms231911711. [PMID: 36233014 PMCID: PMC9570146 DOI: 10.3390/ijms231911711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
BCR-ABL, a fusion protein kinase, is a druggable target exclusively expressed in patients with chronic myeloid leukemia (CML). Several anti-leukemia medicines targeting this protein have been developed in recent years. However, therapeutic options are limited for CML patients bearing multiple BCR-ABL1 mutations. Ponatinib (PON), a potent tyrosinase inhibitor, was one of the approved drugs for managing BCR-ABL1 T315I mutant disease. However, treatment of patients with PON reported severe side effects related to cardiovascular events. Asciminib (ASC) was the first allosteric inhibitor approved to target the myristoyl pocket of BCR-ABL protein to inhibit protein activity. The different mechanism of inhibition opens the possibility of co-exposure with both medicines. Reports on cardiovascular side effects due to the combination use of PON + ASC in pre-clinical and clinical studies are minimal. Thus, this study aimed to observe the potential cardiovascular-related side effect after co-exposure to ASC and PON using zebrafish as an animal model. In this study, zebrafish were acutely exposed to both compounds. The cardiovascular physiology parameters and gene expression related to cardiovascular development were evaluated. We demonstrate that combining ASC with PON at no observed effect concentration (NOEC) did not cause any significant change in the cardiac performance parameter in zebrafish. However, a significant increase in nkx2.5 expression level and a substantial decrease in blood flow velocity were recorded, suggesting that combining these compounds at NOEC can cause mild cardiovascular-related side effects.
Collapse
Affiliation(s)
- Huan-Chau Lin
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei 10449, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, Mackay Memorial Hospital, No. 45, Minsheng Road, Tamsui District, New Taipei City 25160, Taiwan
| | - Ferry Saputra
- Department of Chemistry, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| | - Gilbert Audira
- Department of Chemistry, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan
| | - Marri Jmelou M. Roldan
- The Graduate School, Faculty of Pharmacy, University of Santo Tomas, Manila 1008, Philippines
| | - Honeymae C. Alos
- The Graduate School, University of Santo Tomas, Manila 1008, Philippines
| | | | - Ross D. Vasquez
- Department of Pharmacy, Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila 1008, Philippines
| | - Guan-Jhe Tsai
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei 10449, Taiwan
| | - Ken-Hong Lim
- Division of Hematology and Oncology, Department of Internal Medicine, Mackay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Taipei 10449, Taiwan
- Laboratory of Good Clinical Research Center, Department of Medical Research, Mackay Memorial Hospital, No. 45, Minsheng Road, Tamsui District, New Taipei City 25160, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- Correspondence: (K.-H.L.); (C.-D.H.)
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Center of Nanotechnology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Chung-Li, Taoyuan City 320314, Taiwan
- Correspondence: (K.-H.L.); (C.-D.H.)
| |
Collapse
|
13
|
Ibbini Z, Spicer JI, Truebano M, Bishop J, Tills O. HeartCV: a tool for transferrable, automated measurement of heart rate and heart rate variability in transparent animals. J Exp Biol 2022; 225:276574. [PMID: 36073614 PMCID: PMC9659326 DOI: 10.1242/jeb.244729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022]
Abstract
Heart function is a key component of whole-organismal physiology. Bioimaging is commonly, but not exclusively, used for quantifying heart function in transparent individuals, including early developmental stages of aquatic animals, many of which are transparent. However, a central limitation of many imaging-related methods is the lack of transferability between species, life-history stages and experimental approaches. Furthermore, locating the heart in mobile individuals remains challenging. Here, we present HeartCV: an open-source Python package for automated measurement of heart rate and heart rate variability that integrates automated localization and is transferrable across a wide range of species. We demonstrate the efficacy of HeartCV by comparing its outputs with measurements made manually for a number of very different species with contrasting heart morphologies. Lastly, we demonstrate the applicability of the software to different experimental approaches and to different dataset types, such as those corresponding to longitudinal studies.
Collapse
Affiliation(s)
- Ziad Ibbini
- Marine Biology and Ecology Research Centre, Plymouth University, Plymouth PL4 8AA, UK
- Author for correspondence ()
| | - John I. Spicer
- Marine Biology and Ecology Research Centre, Plymouth University, Plymouth PL4 8AA, UK
| | - Manuela Truebano
- Marine Biology and Ecology Research Centre, Plymouth University, Plymouth PL4 8AA, UK
| | - John Bishop
- Marine Biological Association of the UK, Citadel Hill Laboratory, Plymouth PL1 2PB, UK
| | - Oliver Tills
- Marine Biology and Ecology Research Centre, Plymouth University, Plymouth PL4 8AA, UK
| |
Collapse
|
14
|
Suryanto ME, Saputra F, Kurnia KA, Vasquez RD, Roldan MJM, Chen KHC, Huang JC, Hsiao CD. Using DeepLabCut as a Real-Time and Markerless Tool for Cardiac Physiology Assessment in Zebrafish. BIOLOGY 2022; 11:1243. [PMID: 36009871 PMCID: PMC9405297 DOI: 10.3390/biology11081243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/13/2022] [Accepted: 08/19/2022] [Indexed: 11/21/2022]
Abstract
DeepLabCut (DLC) is a deep learning-based tool initially invented for markerless pose estimation in mammals. In this study, we explored the possibility of adopting this tool for conducting markerless cardiac physiology assessment in an important aquatic toxicology model of zebrafish (Danio rerio). Initially, high-definition videography was applied to capture heartbeat information at a frame rate of 30 frames per second (fps). Next, 20 videos from different individuals were used to perform convolutional neural network training by labeling the heart chamber (ventricle) with eight landmarks. Using Residual Network (ResNet) 152, a neural network with 152 convolutional neural network layers with 500,000 iterations, we successfully obtained a trained model that can track the heart chamber in a real-time manner. Later, we validated DLC performance with the previously published ImageJ Time Series Analysis (TSA) and Kymograph (KYM) methods. We also evaluated DLC performance by challenging experimental animals with ethanol and ponatinib to induce cardiac abnormality and heartbeat irregularity. The results showed that DLC is more accurate than the TSA method in several parameters tested. The DLC-trained model also detected the ventricle of zebrafish embryos even in the occurrence of heart abnormalities, such as pericardial edema. We believe that this tool is beneficial for research studies, especially for cardiac physiology assessment in zebrafish embryos.
Collapse
Affiliation(s)
- Michael Edbert Suryanto
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Ferry Saputra
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Kevin Adi Kurnia
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| | - Ross D. Vasquez
- Department of Pharmacy, Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila 1008, Philippines
| | - Marri Jmelou M. Roldan
- Faculty of Pharmacy, The Graduate School, University of Santo Tomas, Manila 1008, Philippines
| | - Kelvin H.-C. Chen
- Department of Applied Chemistry, National Pingtung University, Pingtung 90003, Taiwan
| | - Jong-Chin Huang
- Department of Applied Chemistry, National Pingtung University, Pingtung 90003, Taiwan
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Center for Nanotechnology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
- Research Center for Aquatic Toxicology and Pharmacology, Chung Yuan Christian University, Taoyuan 320314, Taiwan
| |
Collapse
|
15
|
Magalhães ACM, Correia PMM, Oliveira RG, Encarnação PMCC, Domingues I, Veloso JFCA, Silva ALM. New Enclosure for in vivo Medical Imaging of Zebrafish With Vital Signs Monitoring. Front Physiol 2022; 13:906110. [PMID: 35846002 PMCID: PMC9278279 DOI: 10.3389/fphys.2022.906110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Lately, the use of zebrafish has gained increased interest in the scientific community as an animal model in preclinical research. However, there is a lack of in vivo imaging tools that ensure animal welfare during acquisition procedures. The use of functional imaging techniques, like Positron Emission Tomography (PET), in zebrafish is limited since it requires the animal to be alive, representing a higher instrumentation complexity when compared to morphological imaging systems. In the present work, a new zebrafish enclosure was developed to acquire in vivo images while monitoring the animal’s welfare through its heartbeat. The temperature, dissolved oxygen, and pH range in a closed aquatic environment were tested to ensure that the conditions stay suitable for animal welfare during image acquisitions. The developed system, based on an enclosure with a bed and heartbeat sensors, was tested under controlled conditions in anesthetized fishes. Since the anesthetized zebrafish do not affect the water quality over time, there is no need to incorporate water circulation for the expected time of PET exams (about 30 min). The range of values obtained for the zebrafish heart rate was 88–127 bpm. The developed system has shown promising results regarding the zebrafish’s heart rate while keeping the fish still during the long imaging exams. The zebrafish enclosure ensures the animal’s well-being during the acquisition of in vivo images in different modalities (PET, Computer Tomography, Magnetic Resonance Imaging), contributing substantially to the preclinical research.
Collapse
Affiliation(s)
| | - P. M. M. Correia
- Department of Physics, I3N, University of Aveiro, Aveiro, Portugal
| | - R. G. Oliveira
- Department of Physics, I3N, University of Aveiro, Aveiro, Portugal
| | | | - I. Domingues
- CESAM, Department of Biology, University of Aveiro, Aveiro, Portugal
| | | | - A. L. M. Silva
- Department of Physics, I3N, University of Aveiro, Aveiro, Portugal
- *Correspondence: A. L. M. Silva,
| |
Collapse
|
16
|
Ling D, Chen H, Chan G, Lee SMY. Quantitative measurements of zebrafish heartrate and heart rate variability: A survey between 1990-2020. Comput Biol Med 2021; 142:105045. [PMID: 34995954 DOI: 10.1016/j.compbiomed.2021.105045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/14/2021] [Accepted: 11/14/2021] [Indexed: 12/19/2022]
Abstract
Zebrafish is an essential model organism for studying cardiovascular diseases, given its advantages of fast proliferation and high gene homology with humans. Zebrafish embryos/larvae are valuable experimental models used in toxicology studies to analyze drug toxicity, including hepatoxicity, nephrotoxicity and cardiotoxicity, as well as for drug discovery and drug safety screening in the preclinical stage. Heart rate (HR) serves as a functional endpoint in studies of cardiotoxicity, while heart rate variability (HRV) serves as an indicator of cardiac arrhythmia. Cardiotoxicity is a major cause of early and late termination of drug trials, so a more comprehensive understanding of zebrafish HR and HRV is important. This review summarized HR and HRV in a specific range of applications and fields, focusing on zebrafish heartbeat detection procedures, signal analysis technology and well-established commercial software, such as LabVIEW, Rvlpulse, and ZebraLab. We also compared HR detection algorithms and electrocardiography (ECG)-based methods of heart signal extraction. The relationship between HR and HRV was also systematically analyzed; HR was shown to have an inverse correlation with HRV. Applications to drug testing are also highlighted in this review. Furthermore, HR and HRV were shown to be regulated by the automatic nervous system; their connections with ECG measurements are also summarized herein.
Collapse
Affiliation(s)
- Dongmin Ling
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macao, China
| | - Huanxian Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macao, China
| | - Ging Chan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macao, China; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macao, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macao, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
| |
Collapse
|
17
|
Narumanchi S, Wang H, Perttunen S, Tikkanen I, Lakkisto P, Paavola J. Zebrafish Heart Failure Models. Front Cell Dev Biol 2021; 9:662583. [PMID: 34095129 PMCID: PMC8173159 DOI: 10.3389/fcell.2021.662583] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023] Open
Abstract
Heart failure causes significant morbidity and mortality worldwide. The understanding of heart failure pathomechanisms and options for treatment remain incomplete. Zebrafish has proven useful for modeling human heart diseases due to similarity of zebrafish and mammalian hearts, fast easily tractable development, and readily available genetic methods. Embryonic cardiac development is rapid and cardiac function is easy to observe and quantify. Reverse genetics, by using morpholinos and CRISPR-Cas9 to modulate gene function, make zebrafish a primary animal model for in vivo studies of candidate genes. Zebrafish are able to effectively regenerate their hearts following injury. However, less attention has been given to using zebrafish models to increase understanding of heart failure and cardiac remodeling, including cardiac hypertrophy and hyperplasia. Here we discuss using zebrafish to study heart failure and cardiac remodeling, and review zebrafish genetic, drug-induced and other heart failure models, discussing the advantages and weaknesses of using zebrafish to model human heart disease. Using zebrafish models will lead to insights on the pathomechanisms of heart failure, with the aim to ultimately provide novel therapies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Suneeta Narumanchi
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Hong Wang
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Sanni Perttunen
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Ilkka Tikkanen
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center Nephrology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland.,Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Jere Paavola
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| |
Collapse
|
18
|
Marrs JA, Sarmah S. The Genius of the Zebrafish Model: Insights on Development and Disease. Biomedicines 2021; 9:biomedicines9050577. [PMID: 34065228 PMCID: PMC8160874 DOI: 10.3390/biomedicines9050577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/06/2021] [Indexed: 01/20/2023] Open
|
19
|
Abstract
Inherited cardiac arrhythmias contribute substantially to sudden cardiac death in the young. The underlying pathophysiology remains incompletely understood because of the lack of representative study models and the labour-intensive nature of electrophysiological patch clamp experiments. Whereas patch clamp is still considered the gold standard for investigating electrical properties in a cell, optical mapping of voltage and calcium transients has paved the way for high-throughput studies. Moreover, the development of human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CMs) has enabled the study of patient specific cell lines capturing the full genomic background. Nevertheless, hiPSC-CMs do not fully address the complex interactions between various cell types in the heart. Studies using in vivo models, are therefore necessary. Given the analogies between the human and zebrafish cardiovascular system, zebrafish has emerged as a cost-efficient model for arrhythmogenic diseases. In this review, we describe how hiPSC-CM and zebrafish are employed as models to study primary electrical disorders. We provide an overview of the contemporary electrophysiological phenotyping tools and discuss in more depth the different strategies available for optical mapping. We consider the current advantages and disadvantages of both hiPSC-CM and zebrafish as a model and optical mapping as phenotyping tool and propose strategies for further improvement. Overall, the combination of experimental readouts at cellular (hiPSC-CM) and whole organ (zebrafish) level can raise our understanding of the complexity of inherited cardiac arrhythmia disorders to the next level.
Collapse
|